101
|
LUO XN, YAO HL, SONG J, SONG QQ, SHI BT, XIA D, HAN J. Coxsackievirus B3 Infection Triggers Autophagy through 3 Pathways of Endoplasmic Reticulum Stress. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2018; 31:867-875. [PMID: 30636656 PMCID: PMC7126911 DOI: 10.3967/bes2018.115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 11/22/2018] [Indexed: 06/09/2023]
Abstract
OBJECTIVE Autophagy is a highly conserved intracellular degradation pathway. Many picornaviruses induce autophagy to benefit viral replication, but an understanding of how autophagy occurs remains incomplete. In this study, we explored whether coxsackievirus B3 (CVB3) infection induced autophagy through endoplasmic reticulum (ER) stress. METHODS In CVB3-infected HeLa cells, the specific molecules of ER stress and autophagy were detected using Western blotting, reverse transcription polymerase chain reaction (RT-PCR), and confocal microscopy. Then PKR-like ER protein kinase (PERK) inhibitor, inositol-requiring protein-1 (IRE1) inhibitor, or activating transcription factor-6 (ATF6) inhibitor worked on CVB3-infected cells, their effect on autophagy was assessed by Western blotting for detecting microtubule-associated protein light chain 3 (LC3). RESULTS CVB3 infection induced ER stress, and ER stress sensors PERK/eIF2α, IRE1/XBP1, and ATF6 were activated. CVB3 infection increased the accumulation of green fluorescent protein (GFP)-LC3 punctuation and induced the conversion from LC3-I to phosphatidylethanolamine-conjugated LC3-1 (LC3-II). CVB3 infection still decreased the expression of mammalian target of rapamycin (mTOR) and p-mTOR. Inhibition of PERK, IRE1, or ATF6 significantly decreased the ratio of LC3-II to LC3-I in CVB3-infected HeLa cells. CONCLUSION CVB3 infection induced autophagy through ER stress in HeLa cells, and PERK, IRE1, and ATF6a pathways participated in the regulation of autophagy. Our data suggested that ER stress may inhibit mTOR signaling pathway to induce autophagy during CVB3 infection.
Collapse
Affiliation(s)
- Xiao Nuan LUO
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hai Lan YAO
- Molecular Immunology Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Juan SONG
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qin Qin SONG
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Bing Tian SHI
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Dong XIA
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jun HAN
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
102
|
Tan Z, Zhang W, Sun J, Fu Z, Ke X, Zheng C, Zhang Y, Li P, Liu Y, Hu Q, Wang H, Zheng Z. ZIKV infection activates the IRE1-XBP1 and ATF6 pathways of unfolded protein response in neural cells. J Neuroinflammation 2018; 15:275. [PMID: 30241539 PMCID: PMC6151056 DOI: 10.1186/s12974-018-1311-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Many viruses depend on the extensive membranous network of the endoplasmic reticulum (ER) for their translation, replication, and packaging. Certain membrane modifications of the ER can be a trigger for ER stress, as well as the accumulation of viral protein in the ER by viral infection. Then, unfolded protein response (UPR) is activated to alleviate the stress. Zika virus (ZIKV) is a mosquito-borne flavivirus and its infection causes microcephaly in newborns and serious neurological complications in adults. Here, we investigated ER stress and the regulating model of UPR in ZIKV-infected neural cells in vitro and in vivo. METHODS Mice deficient in type I and II IFN receptors were infected with ZIKV via intraperitoneal injection and the nervous tissues of the mice were assayed at 5 days post-infection. The expression of phospho-IRE1, XBP1, and ATF6 which were the key markers of ER stress were analyzed by immunohistochemistry assay in vivo. Additionally, the nuclear localization of XBP1s and ATF6n were analyzed by immunohistofluorescence. Furthermore, two representative neural cells, neuroblastoma cell line (SK-N-SH) and astrocytoma cell line (CCF-STTG1), were selected to verify the ER stress in vitro. The expression of BIP, phospho-elF2α, phospho-IRE1, and ATF6 were analyzed through western blot and the nuclear localization of XBP1s was performed by confocal immunofluorescence microscopy. RT-qPCR was also used to quantify the mRNA level of the UPR downstream genes in vitro and in vivo. RESULTS ZIKV infection significantly upregulated the expression of ER stress markers in vitro and in vivo. Phospho-IRE1 and XBP1 expression significantly increased in the cerebellum and mesocephalon, while ATF6 expression significantly increased in the mesocephalon. ATF6n and XBP1s were translocated into the cell nucleus. The levels of BIP, ATF6, phospho-elf2α, and spliced xbp1 also significantly increased in vitro. Furthermore, the downstream genes of UPR were detected to investigate the regulating model of the UPR during ZIKV infection in vitro and in vivo. The transcriptional levels of atf4, gadd34, chop, and edem-1 in vivo and that of gadd34 and chop in vitro significantly increased. CONCLUSION Findings in this study demonstrated that ZIKV infection activates ER stress in neural cells. The results offer clues to further study the mechanism of neuropathogenesis caused by ZIKV infection.
Collapse
Affiliation(s)
- Zhongyuan Tan
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Wanpo Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 China
| | - Jianhong Sun
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Zuquan Fu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070 China
| | - Xianliang Ke
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Caishang Zheng
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Yuan Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Penghui Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yan Liu
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Hanzhong Wang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| | - Zhenhua Zheng
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
| |
Collapse
|
103
|
Tan B, Jia R, Wang G, Yang J. Astragaloside attenuates the progression of prostate cancer cells through endoplasmic reticulum stress pathways. Oncol Lett 2018; 16:3901-3906. [PMID: 30128005 DOI: 10.3892/ol.2018.9071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/09/2018] [Indexed: 11/06/2022] Open
Abstract
Astragaloside (As) has been demonstrated extensively to serve roles in a variety of tumor types, including glioma, lung cancer, colorectal cancer, breast cancer and cervical cancer, and has therefore been widely used in Traditional Chinese Medicine. To the best of our knowledge, the present study was the first to investigate the efficacy of the Traditional Chinese Medicine astragaloside on tumor growth and the apoptosis of prostate cancer cells. In addition, further investigation into the underlying molecular mechanisms via the endoplasmic reticulum (ER) stress pathway was also performed. In the present study, the human prostate cancer DU-145 cell line was employed as an experimental model in vitro and cells were divided into five treatment groups: Dimethyl sulfoxide (DMSO) group (control), low-dose astragaloside group (L-As; 20 nmol/l), moderate-dose astragaloside group (M-As; 50 nmol/l), high-dose astragaloside group (H-As, 100 nmol/l) and ER stress suppressor group (tauroursodeoxycholic acid; TUDCA). The proliferative ability and apoptosis rate of the DU-145 cells were detected via Cell Counting kit-8 methods and flow cytometry, respectively. Furthermore, the ER stress factors [binding immunoglobulin protein (BiP), C/EBP homologous protein (CHOP) and caspase-12] were assessed through reverse transcription polymerase chain reaction. Additionally, the protein expression levels of inositol-requiring enzyme 1 (IRE1), phosphorylated protein kinase R-like ER kinase (p-PERK), iron-regulated transcriptional activator Aft (AFT)4 and AFT6 were measured detected by western blot analysis. Administration of As significantly reduced the cell viability and promoted apoptosis (P<0.05) in a dose-dependent manner. Expression of ER-stressed genes BiP, CHOP and caspase-12 mRNA was increased by As administration, while TUDCA treatment led to a lower mRNA expression of these genes, compared with the control group. Results of western blot analysis indicated that the protein expression of IRE1, AFT4 and AFT6 was upregulated in the H-As group, and that the ratio of p-PERK/PERK was also higher than in the other groups. The administration of As demonstrated significant therapeutic effects on the proliferation of prostate cancer cells, as well as the expression of related proteins and genes. The results of the present study suggested future clinical potential of As for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Bo Tan
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Renfeng Jia
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Gang Wang
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Jinhui Yang
- Department of Urology, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| |
Collapse
|
104
|
Yang M, Zhang Y, Xie X, Yue N, Li J, Wang XB, Han C, Yu J, Liu Y, Li D. Barley stripe mosaic virus γb Protein Subverts Autophagy to Promote Viral Infection by Disrupting the ATG7-ATG8 Interaction. THE PLANT CELL 2018; 30:1582-1595. [PMID: 29848767 PMCID: PMC6096602 DOI: 10.1105/tpc.18.00122] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/18/2018] [Accepted: 05/22/2018] [Indexed: 05/04/2023]
Abstract
Autophagy is a conserved defense strategy against viral infection. However, little is known about the counterdefense strategies of plant viruses involving interference with autophagy. Here, we show that γb protein from Barley stripe mosaic virus (BSMV), a positive single-stranded RNA virus, directly interacts with AUTOPHAGY PROTEIN7 (ATG7). BSMV infection suppresses autophagy, and overexpression of γb protein is sufficient to inhibit autophagy. Furthermore, silencing of autophagy-related gene ATG5 and ATG7 in Nicotiana benthamiana plants enhanced BSMV accumulation and viral symptoms, indicating that autophagy plays an antiviral role in BSMV infection. Molecular analyses indicated that γb interferes with the interaction of ATG7 with ATG8 in a competitive manner, whereas a single point mutation in γb, Tyr29Ala (Y29A), made this protein deficient in the interaction with ATG7, which was correlated with the abolishment of autophagy inhibition. Consistently, the mutant BSMVY29A virus showed reduced symptom severity and viral accumulation. Taken together, our findings reveal that BSMV γb protein subverts autophagy-mediated antiviral defense by disrupting the ATG7-ATG8 interaction to promote plant RNA virus infection, and they provide evidence that ATG7 is a target of pathogen effectors that functions in the ongoing arms race of plant defense and viral counterdefense.
Collapse
Affiliation(s)
- Meng Yang
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Yongliang Zhang
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Xialin Xie
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Ning Yue
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Jinlin Li
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Xian-Bing Wang
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Chenggui Han
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Jialin Yu
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| | - Yule Liu
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Dawei Li
- State Key Laboratory of Agro-Biotechnology and Ministry of Agriculture Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, P.R. China
| |
Collapse
|
105
|
Wang Q, Yuan X, Chen Y, Zheng Q, Xu L, Wu Y. Endoplasmic Reticulum Stress Mediated MDRV p10.8 Protein-Induced Cell Cycle Arrest and Apoptosis Through the PERK/eIF2α Pathway. Front Microbiol 2018; 9:1327. [PMID: 29977231 PMCID: PMC6021497 DOI: 10.3389/fmicb.2018.01327] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
In this study, the mechanism of Muscovy duck reovirus (MDRV) p10.8 protein-induced pathogenesis was investigated, with a focus on endoplasmic reticulum (ER) stress. In chicken embryo fibroblasts cell lines (DF1), pCI-neo-flg-p10.8 protein transfection increased the phosphorylation (p-) levels of PERK and eIF2α as shown by Western blotting analysis and led to the dissociation of BiP from PERK as shown by co-immunoprecipitation (Co-IP) analysis. Results of treatment with both ER stress activator and inhibitor further confirmed that p10.8 protein induced ER stress. Subsequently, using flow cytometry analysis, it was also found that p10.8 protein induced cell cycle arrest during the G0/G1 phase. Furthermore, p10.8 transfection increased the phosphorylation levels of PERK and eIF2α, and reduced the expression levels of CDK2, CDK4, and Cyclin E according to Western blotting analysis. Treatment with ER stress activator and ER stress inhibitor after p10.8 protein transfection in DF1 cells further indicated that p10.8 protein induced ER stress, which resulted in cell cycle arrest. The results of knockdown of either PERK or eIF2α genes further confirmed that p10.8 protein-induced ER stress led to cell cycle arrest through the PERK/eIF2α pathway. Further results showed that p10.8 protein induced ER stress and apoptosis in DF1 cells. The expression levels of p-PERK, p-eIF2α, CHOP, cleaved-Caspase12, and cleaved-Caspase3 were increased by p10.8 protein. Test results of treatment with each of Tunicamycin, TUDCA and knockdown of PERK, and eIF2α, confirmed that p10.8 protein induced ER stress involving apoptosis via the PERK/eIF2α pathway. In conclusion, MDRV p10.8 protein induced ER stress that caused cell cycle arrest and apoptosis through the PERK/eIF2α pathway.
Collapse
Affiliation(s)
- Quanxi Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China.,Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaoqin Yuan
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China.,Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yuan Chen
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China.,Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qingli Zheng
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China.,Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lihui Xu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yijian Wu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
106
|
Rodrigues LOCP, Graça RSF, Carneiro LAM. Integrated Stress Responses to Bacterial Pathogenesis Patterns. Front Immunol 2018; 9:1306. [PMID: 29930559 PMCID: PMC5999787 DOI: 10.3389/fimmu.2018.01306] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/25/2018] [Indexed: 12/25/2022] Open
Abstract
Activation of an appropriate innate immune response to bacterial infection is critical to limit microbial spread and generate cytokines and chemokines to instruct appropriate adaptive immune responses. Recognition of bacteria or bacterial products by pattern recognition molecules is crucial to initiate this response. However, it is increasingly clear that the context in which this recognition occurs can dictate the quality of the response and determine the outcome of an infection. The cross talk established between host and pathogen results in profound alterations on cellular homeostasis triggering specific cellular stress responses. In particular, the highly conserved integrated stress response (ISR) has been shown to shape the host response to bacterial pathogens by sensing cellular insults resulting from infection and modulating transcription of key genes, translation of new proteins and cell autonomous antimicrobial mechanisms such as autophagy. Here, we review the growing body of evidence demonstrating a role for the ISR as an integral part of the innate immune response to bacterial pathogens.
Collapse
Affiliation(s)
- Larissa O C P Rodrigues
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo S F Graça
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia A M Carneiro
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
107
|
Chen D, Feng C, Tian X, Zheng N, Wu Z. Promyelocytic Leukemia Restricts Enterovirus 71 Replication by Inhibiting Autophagy. Front Immunol 2018; 9:1268. [PMID: 29922292 PMCID: PMC5996053 DOI: 10.3389/fimmu.2018.01268] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022] Open
Abstract
The promyelocytic leukemia (PML) protein, also known as TRIM19, functions as a major organizer of PML nuclear bodies (NBs) in most mammalian cells and plays important roles in antiviral activities against both DNA and RNA viruses. In this study, we found that the downregulation of PML rendered HeLa cells more susceptible to infection by enterovirus 71 (EV71), and the overexpression of the PMLIII or PMLIV isoforms inhibited viral protein expression and resulted in viral titers that were 2–3 log units lower than those in the control. Using short interfering RNAs, the downregulation of either the PMLIII or PMLIV isoform increased both viral protein VP1 expression and viral production. The PML repression of EV71 replication was partially mediated by the inhibition of autophagy, and PML deficiency triggered autophagy. Furthermore, the EV71 infection resulted in a reduction in PML independent of the proteasome pathway. Instead, PML degradation was mediated by virus protease 3Cpro. In conclusion, PML contributes to a cellular antiviral effect by inhibiting autophagy, which is countered by a disruption of promyelocytic leukemia protein-nuclear bodies mediated by viral protease 3Cpro.
Collapse
Affiliation(s)
- Deyan Chen
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Chunhong Feng
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Nan Zheng
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
108
|
Wu S, Yang Y, Li F, Huang L, Han Z, Wang G, Yu H, Li H. Chelerythrine induced cell death through ROS-dependent ER stress in human prostate cancer cells. Onco Targets Ther 2018; 11:2593-2601. [PMID: 29780252 PMCID: PMC5951218 DOI: 10.2147/ott.s157707] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Prostate cancer is the most common noncutaneous cancer and the second leading cause of cancer-related mortality worldwide and the third in USA in 2017. Chelerythrine (CHE), a naturalbenzo[c]phenanthridine alkaloid, formerly identified as a protein kinase C inhibitor, has also shown anticancer effect through a number of mechanisms. Herein, effect and mechanism of the CHE-induced apoptosis via reactive oxygen species (ROS)-mediated endoplasmic reticulum (ER) stress in prostate cancer cells were studied for the first time. METHODS In our present study, we investigated whether CHE induced cell viability decrease, colony formation inhibition, and apoptosis in a dose-dependent manner in PC-3 cells. In addition, we showed that CHE increases intracellular ROS and leads to ROS-dependent ER stress and cell apoptosis. RESULTS Pre-treatment with N-acetyl cysteine, an ROS scavenger, totally reversed the CHE-induced cancer cell apoptosis as well as ER stress activation, suggesting that the ROS generation was responsible for the anticancer effects of CHE. CONCLUSION Taken together, our findings support one of the anticancer mechanisms by which CHE increased ROS accumulation in prostate cancer cells, thereby leading to ER stress and caused intrinsic apoptotic signaling. The study reveals that CHE could be a potential candidate for application in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Songjiang Wu
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Yanying Yang
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Feiping Li
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Lifu Huang
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Zihua Han
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Guanfu Wang
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Hongyuan Yu
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| | - Haiping Li
- Department of Urology, Enze Hospital of Taizhou Enze Medical Center (Group), Taizhou, China
| |
Collapse
|
109
|
Celeste DB, Miller MS. Reviewing the evidence for viruses as environmental risk factors for ALS: A new perspective. Cytokine 2018; 108:173-178. [PMID: 29684753 DOI: 10.1016/j.cyto.2018.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/05/2018] [Accepted: 04/07/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease whose etiology remains poorly understood. Since the genetic basis of disease is known in only a small subset of cases, there has been substantial interest in determining whether environmental factors act as triggers of ALS. Viruses have received longstanding attention as potential ALS triggers. Yet, existing studies have not provided a compelling case for causation. This review summarizes the evidence supporting a link between viral infection and motor neuron disease, with a focus on ALS. Limitations of prior studies are discussed and contextualized, and recent work that has provided stronger mechanistic evidence for viruses in disease pathogenesis is highlighted. Finally, we offer a new perspective on the association of viruses with ALS, and underscore the need for multidisciplinary approaches bridging neurology and infectious diseases research to move the field forward in the future.
Collapse
Affiliation(s)
- Daniel B Celeste
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Matthew S Miller
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
110
|
Li YC, Zhang MQ, Zhang JP. Opposite Effects of Two Human ATG10 Isoforms on Replication of a HCV Sub-genomic Replicon Are Mediated via Regulating Autophagy Flux in Zebrafish. Front Cell Infect Microbiol 2018; 8:109. [PMID: 29670865 PMCID: PMC5893791 DOI: 10.3389/fcimb.2018.00109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a host mechanism for cellular homeostatic control. Intracellular stresses are symptoms of, and responses to, dysregulation of the physiological environment of the cell. Alternative gene transcription splicing is a mechanism potentially used by a host to respond to physiological or pathological challenges. Here, we aimed to confirm opposite effects of two isoforms of the human autophagy-related protein ATG10 on an HCV subgenomic replicon in zebrafish. A liver-specific HCV subreplicon model was established and exhibited several changes in gene expression typically induced by HCV infection, including overexpression of several HCV-dependent genes (argsyn, leugpcr, rasgbd, and scaf-2), as well as overexpression of several ER stress related genes (atf4, chop, atf6, and bip). Autophagy flux was blocked in the HCV model. Our results indicated that the replication of the HCV subreplicon was suppressed via a decrease in autophagosome formation caused by the autophagy inhibitor 3MA, but enhanced via dysfunction in the lysosomal degradation caused by another autophagy inhibitor CQ. Human ATG10, a canonical isoform in autophagy, facilitated the amplification of the HCV-subgenomic replicon via promoting autophagosome formation. ATG10S, a non-canonical short isoform of the ATG10 protein, promoted autophagy flux, leading to lysosomal degradation of the HCV-subgenomic replicon. Human ATG10S may therefore inhibit HCV replication, and may be an appropriate target for future antiviral drug screening.
Collapse
Affiliation(s)
- Yu-Chen Li
- Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao-Qing Zhang
- Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Pu Zhang
- Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
111
|
Wang R, Moniruzzaman M, Shuffle E, Lourie R, Hasnain SZ. Immune regulation of the unfolded protein response at the mucosal barrier in viral infection. Clin Transl Immunology 2018; 7:e1014. [PMID: 29632667 PMCID: PMC5881172 DOI: 10.1002/cti2.1014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 01/12/2023] Open
Abstract
Protein folding in the endoplasmic reticulum (ER) is subject to stringent quality control. When protein secretion demand exceeds the protein folding capacity of the ER, the unfolded protein response (UPR) is triggered as a consequence of ER stress. Due to the secretory function of epithelial cells, UPR plays an important role in maintaining epithelial barrier function at mucosal sites. ER stress and activation of the UPR are natural mechanisms by which mucosal epithelial cells combat viral infections. In this review, we discuss the important role of UPR in regulating mucosal epithelium homeostasis. In addition, we review current insights into how the UPR is involved in viral infection at mucosal barriers and potential therapeutic strategies that restore epithelial cell integrity following acute viral infections via cytokine and cellular stress manipulation.
Collapse
Affiliation(s)
- Ran Wang
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Md Moniruzzaman
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Eric Shuffle
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia
| | - Rohan Lourie
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Translational Research Institute Inflammatory Bowel Disease Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia
| | - Sumaira Z Hasnain
- Translational Research Institute Immunopathology Group at Mater Research Institute - The University of Queensland Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| |
Collapse
|
112
|
Deficiency of the IRE1α-Autophagy Axis Enhances the Antitumor Effects of the Oncolytic Virus M1. J Virol 2018; 92:JVI.01331-17. [PMID: 29263275 DOI: 10.1128/jvi.01331-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022] Open
Abstract
Oncolytic virotherapy is an emerging treatment modality that uses replication-competent viruses to destroy cancer cells. M1 is a naturally occurring alphavirus (Togaviridae) which shows potent oncolytic activities against many cancers. Accumulation of unfolded proteins during virus replication leads to a transcriptional/translational response known as the unfolded protein response (UPR), which might counteract the antitumor effect of the oncolytic virus. In this report, we show that either pharmacological or biological inhibition of IRE1α or PERK, but not ATF6, substantially increases the oncolytic effects of the M1 virus. Moreover, inhibition of IRE1α blocks M1 virus-induced autophagy, which restricts the antitumor effects of the M1 virus through degradation of viral protein, in glioma cells. In addition, IRE1α suppression significantly increases the oncolytic effect of M1 virus in an orthotopic glioma model. From a molecular pathology study, we found that IRE1α is expressed at lower levels in higher-grade gliomas, suggesting greater antitumor efficacy of the oncolytic virus M1. Taken together, these findings illustrate a defensive mechanism of glioma cells against the oncolytic virus M1 and identify possible approaches to enhance the oncolytic viral protein accumulation and the subsequent lysis of tumor cells.IMPORTANCE Although oncolytic virotherapy is showing great promise in clinical applications, not all patients are benefiting. Identifying inhibitory signals in refractory cancer cells for each oncolytic virus would provide a good chance to increase the therapeutic effect. Here we describe that infection with the oncolytic virus M1 triggers the unfolded protein response (UPR) and subsequent autophagy, while blocking the UPR-autophagy axis significantly potentiates the antitumor efficacy of M1 in vitro and in vivo A survey of cancer tissue banks revealed that IRE1α, a key element in the UPR pathway, is commonly downregulated in higher-grade human gliomas, suggesting favorable prospects for the application of M1. Our work provides a potential predictor and target for enhancement of the therapeutic effectiveness of the M1 virus. We predict that the mechanism-based combination therapy will promote cancer virotherapy in the future.
Collapse
|
113
|
Reverendo M, Mendes A, Argüello RJ, Gatti E, Pierre P. At the crossway of ER-stress and proinflammatory responses. FEBS J 2018; 286:297-310. [PMID: 29360216 DOI: 10.1111/febs.14391] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/12/2018] [Accepted: 01/18/2018] [Indexed: 12/13/2022]
Abstract
Immune cells detect specific microbes or damage to tissue integrity in order to initiate efficient immune responses. Abnormal accumulation of proteins in the endoplasmic reticulum (ER) can be seen as a sign of cellular malfunction and stress that triggers a collection of conserved emergency rescue programs. These different signaling cascades, which favor ER proteostasis and promote cell survival, are collectively known as the unfolded protein response (UPR). In recent years, a synergy between the UPR and inflammatory cytokine production has been unraveled, with different branches of the UPR entering in a cross-talk with specialized microbe sensing pathways, which turns on or amplify inflammatory cytokines production. Complementary to this synergetic activity, UPR induction alone, can itself be seen as a danger signal, and triggers directly or indirectly inflammation in different cellular and pathological models, this independently of the presence of pathogens. Here, we discuss recent advances on the nature of these cross-talks and how innate immunity, metabolism dysregulation, and ER-signaling pathways intersect in specialized immune cells, such as dendritic cells (DCs), and contribute to the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Marisa Reverendo
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille cedex 9, France.,International Associated Laboratory (LIA) CNRS 'Mistra', Marseille cedex 9, France
| | - Andreia Mendes
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille cedex 9, France.,International Associated Laboratory (LIA) CNRS 'Mistra', Marseille cedex 9, France
| | - Rafael J Argüello
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille cedex 9, France
| | - Evelina Gatti
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille cedex 9, France.,International Associated Laboratory (LIA) CNRS 'Mistra', Marseille cedex 9, France.,Institute for Research in Biomedicine (iBiMED) and Ilidio Pinho Foundation, Department of Medical Sciences, University of Aveiro, Portugal
| | - Philippe Pierre
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille cedex 9, France.,International Associated Laboratory (LIA) CNRS 'Mistra', Marseille cedex 9, France.,Institute for Research in Biomedicine (iBiMED) and Ilidio Pinho Foundation, Department of Medical Sciences, University of Aveiro, Portugal
| |
Collapse
|
114
|
Jheng JR, Lau KS, Lan YW, Horng JT. A novel role of ER stress signal transducer ATF6 in regulating enterovirus A71 viral protein stability. J Biomed Sci 2018; 25:9. [PMID: 29386036 PMCID: PMC5793394 DOI: 10.1186/s12929-018-0412-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background Due to limited coding capacity of viral genome, enterovirus A71 (EV-A71) co-opts host nuclear proteins for its replication. Upon ER stress, the ER-localized 90 kDa activating transcription factor 6 (p90ATF6) is proteolytically cleaved to produce the transcriptionally active amino-terminal 50 kDa (p50ATF6) product where it enters the nucleus to activate a subset of unfolded protein response and ER-associated degradation (also known as ERAD) genes. During EV-A71 infection, however, this p50ATF6 product was not detected in the nucleus, and its downstream target genes were not activated. Methods We examined the role of ATF6 during EV-A71 infection, including its cleavage process and its role in viral life cycle by silencing or overexpressing ATF6. Results We showed that a potential cleavage in the middle of p90ATF6 produced an amino-terminal ~ 45 kDa fragment in a viral protease-independent but EV-A71-dependent manner. The disappearance of ATF6 was not restricted to a specific strain of EV-A71 or cell type, and was not simply caused by picornavirus-mediated global translational shutoff. This cleavage of ATF6, which was most likely mediated by the host response, was nevertheless independent of both cellular caspases and XBP1-associated proteasomes. The silencing of ATF6 expression by small interfering RNA suppressed viral titers due to reduced viral protein stability. This effect was markedly restored by the ectopic expression of p90ATF6. Conclusion Our findings indicate that ATF6 plays a distinct role in viral protein stability and that the host uses different cleavage strategies, rather than conventional cleavage by generating p50ATF6, to combat viral infection. Electronic supplementary material The online version of this article (10.1186/s12929-018-0412-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jia-Rong Jheng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, 259 Wen-Hwa First Road, Kweishan, 333, Taoyuan, Taiwan
| | - Kean-Seng Lau
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, 259 Wen-Hwa First Road, Kweishan, 333, Taoyuan, Taiwan
| | - Yueh-Wen Lan
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, 259 Wen-Hwa First Road, Kweishan, 333, Taoyuan, Taiwan
| | - Jim-Tong Horng
- Department of Biochemistry and Molecular Biology, College of Medicine, Chang Gung University, 259 Wen-Hwa First Road, Kweishan, 333, Taoyuan, Taiwan. .,Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kweishan, 333, Taoyuan, Taiwan. .,Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, 333, Taoyuan, Taiwan. .,Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, 333, Taoyuan, Taiwan.
| |
Collapse
|
115
|
Genotype 2 Strains of Porcine Reproductive and Respiratory Syndrome Virus Dysregulate Alveolar Macrophage Cytokine Production via the Unfolded Protein Response. J Virol 2018; 92:JVI.01251-17. [PMID: 29070690 DOI: 10.1128/jvi.01251-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/17/2017] [Indexed: 11/20/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) infects alveolar macrophages (AMϕ), causing dysregulated alpha interferon (IFN-α) and tumor necrosis factor alpha (TNF-α) production through a mechanism(s) yet to be resolved. Here, we show that AMϕ infected with PRRSV secreted a reduced quantity of IFN-α following exposure of the cell to synthetic double-stranded RNA (dsRNA). This reduction did not correlate with reduced IFNA1 gene transcription. Rather, it coincided with two events that occurred late during infection and that were indicative of translational attenuation, specifically, the activation of eukaryotic translation initiation factor 2α (eIF2α) and the appearance of stress granules. Notably, the typical rapid production of TNF-α by AMϕ exposed to lipopolysaccharide (LPS) was suppressed or enhanced by PRRSV, depending on when the LPS exposure occurred after virus infection. If exposure was delayed until 6 h postinfection (hpi) so that the development of the cytokine response coincided with the time in which phosphorylation of eIF2α by the stress sensor PERK (protein kinase RNA [PKR]-like ER kinase) occurred, inhibition of TNF-α production was observed. However, if LPS exposure occurred at 2 hpi, prior to a detectable onset of eIF2α phosphorylation, a synergistic response was observed due to the earlier NF-κB activation via the stress sensor IRE1α (inositol-requiring kinase 1α). These results suggest that the asynchronous actions of two branches of the unfolded protein response (UPR), namely, IRE1α, and PERK, activated by ER stress resulting from the virus infection, are associated with enhancement or suppression of TNF-α production, respectively.IMPORTANCE The activation of AMϕ is controlled by the microenvironment to deter excessive proinflammatory cytokine responses to microbes that could impair lung function. However, viral pneumonias frequently become complicated by secondary bacterial infections, triggering severe inflammation, lung dysfunction, and death. Although dysregulated cytokine production is considered an integral component of the exacerbated inflammatory response in viral-bacterial coinfections, the mechanism responsible for this event is unknown. Here, we show that PRRSV replication in porcine AMϕ triggers activation of the IRE1α branch of the UPR, which causes a synergistic TNF-α response to LPS exposure. Thus, the severe pneumonias typically observed in pigs afflicted with PRRSV-bacterial coinfections could result from dysregulated, overly robust TNF-α production in response to opportunistic pathogens that is not commensurate with the typical restrained reaction by uninfected AMϕ. This notion could help in the design of therapies to mitigate the severity of viral and bacterial coinfections.
Collapse
|
116
|
Emerging Role for the PERK/eIF2α/ATF4 in Human Cutaneous Leishmaniasis. Sci Rep 2017; 7:17074. [PMID: 29213084 PMCID: PMC5719050 DOI: 10.1038/s41598-017-17252-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 11/23/2017] [Indexed: 02/02/2023] Open
Abstract
Leishmania parasites utilize adaptive evasion mechanisms in infected macrophages to overcome host defenses and proliferate. We report here that the PERK/eIF2α/ATF4 signaling branch of the integrated endoplasmic reticulum stress response (IERSR) is activated by Leishmania and this pathway is important for Leishmania amazonensis infection. Knocking down PERK or ATF4 expression or inhibiting PERK kinase activity diminished L. amazonensis infection. Knocking down ATF4 decreased NRF2 expression and its nuclear translocation, reduced HO-1 expression and increased nitric oxide production. Meanwhile, the increased expression of ATF4 and HO-1 mRNAs were observed in lesions derived from patients infected with the prevalent related species L.(V.) braziliensis. Our data demonstrates that Leishmania parasites activate the PERK/eIF2α/ATF-4 pathway in cultured macrophages and infected human tissue and that this pathway is important for parasite survival and progression of the infection.
Collapse
|
117
|
He W, Xu H, Gou H, Yuan J, Liao J, Chen Y, Fan S, Xie B, Deng S, Zhang Y, Chen J, Zhao M. CSFV Infection Up-Regulates the Unfolded Protein Response to Promote Its Replication. Front Microbiol 2017; 8:2129. [PMID: 29163417 PMCID: PMC5673830 DOI: 10.3389/fmicb.2017.02129] [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: 08/04/2017] [Accepted: 10/18/2017] [Indexed: 01/07/2023] Open
Abstract
Classical swine fever (CSF) is an OIE-listed, highly contagious animal disease caused by classical swine fever virus (CSFV). The endoplasmic reticulum (ER) is an organelle in which the replication of many RNA viruses takes place. During viral infection, a series of events elicited in cells can destroy the ER homeostasis that cause ER stress and induce an unfolded protein response (UPR). In this study, we demonstrate that ER stress was induced during CSFV infection as several UPR-responsive elements such as XBP1(s), GRP78 and CHOP were up-regulated. Specifically, CSFV transiently activated IRE1 pathway at the initial stage of infection but rapidly switched off, likely due to the reduction in cytoplasm Ca2+ after viral incubation. Additionally, our data show that the ER stress induced by CSFV can promote CSFV production, which the IRE1 pathway play an important role in it. Evidence of ER stress in vivo was also confirmed by the marked elevation of GRP78 in CSFV-infected pig PBMC and tissues. Collectively, these data indicate that the ER stress was induced upon CSFV infection and that the activation of the IRE1 pathway benefits CSFV replication.
Collapse
Affiliation(s)
- Wencheng He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hailuan Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongchao Gou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jin Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiedan Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Baoming Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shaofeng Deng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yangyi Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
118
|
Hepatitis C Virus-Induced Autophagy and Host Innate Immune Response. Viruses 2017; 9:v9080224. [PMID: 28805674 PMCID: PMC5580481 DOI: 10.3390/v9080224] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/04/2017] [Accepted: 08/11/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a catabolic process that is important for maintaining cellular homeostasis. This pathway in hepatocytes is stimulated and controlled by the hepatitis C virus (HCV)—upon infection—to promote its own replication. HCV induces autophagy indirectly and directly through different mechanisms and temporally controls the autophagic flux. This enables the virus to maximize its replication and attenuate the innate immune responses that it activates. In this review, we discuss the relationship between HCV and autophagy, and the crosstalk between HCV-induced autophagy and host innate immune responses.
Collapse
|
119
|
Monel B, Compton AA, Bruel T, Amraoui S, Burlaud-Gaillard J, Roy N, Guivel-Benhassine F, Porrot F, Génin P, Meertens L, Sinigaglia L, Jouvenet N, Weil R, Casartelli N, Demangel C, Simon-Lorière E, Moris A, Roingeard P, Amara A, Schwartz O. Zika virus induces massive cytoplasmic vacuolization and paraptosis-like death in infected cells. EMBO J 2017; 36:1653-1668. [PMID: 28473450 PMCID: PMC5470047 DOI: 10.15252/embj.201695597] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 03/23/2017] [Accepted: 03/28/2017] [Indexed: 12/15/2022] Open
Abstract
The cytopathic effects of Zika virus (ZIKV) are poorly characterized. Innate immunity controls ZIKV infection and disease in most infected patients through mechanisms that remain to be understood. Here, we studied the morphological cellular changes induced by ZIKV and addressed the role of interferon-induced transmembrane proteins (IFITM), a family of broad-spectrum antiviral factors, during viral replication. We report that ZIKV induces massive vacuolization followed by "implosive" cell death in human epithelial cells, primary skin fibroblasts and astrocytes, a phenomenon which is exacerbated when IFITM3 levels are low. It is reminiscent of paraptosis, a caspase-independent, non-apoptotic form of cell death associated with the formation of large cytoplasmic vacuoles. We further show that ZIKV-induced vacuoles are derived from the endoplasmic reticulum (ER) and dependent on the PI3K/Akt signaling axis. Inhibiting the Sec61 ER translocon in ZIKV-infected cells blocked vacuole formation and viral production. Our results provide mechanistic insight behind the ZIKV-induced cytopathic effect and indicate that IFITM3, by acting as a gatekeeper for incoming virus, restricts virus takeover of the ER and subsequent cell death.
Collapse
Affiliation(s)
| | | | | | - Sonia Amraoui
- Virus & Immunity Unit, Institut Pasteur, Paris, France
| | - Julien Burlaud-Gaillard
- INSERM U966 & Platefome IBiSA de Microscopie Electronique, Université François Rabelais and CHRU de Tours, Paris, France
| | - Nicolas Roy
- Virus & Immunity Unit, Institut Pasteur, Paris, France
| | | | | | - Pierre Génin
- Signaling and Pathogenesis Laboratory and CNRS UMR3691, Institut Pasteur, Paris, France
| | - Laurent Meertens
- INSERM U944, CNRS 7212 Laboratoire de Pathologie et Virologie Moléculaire, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Laura Sinigaglia
- Viral Genomics and Vaccination Unit, Institut Pasteur, Paris, France
- UMR CNRS 3569, Paris, France
| | - Nolwenn Jouvenet
- Viral Genomics and Vaccination Unit, Institut Pasteur, Paris, France
- UMR CNRS 3569, Paris, France
| | - Robert Weil
- Signaling and Pathogenesis Laboratory and CNRS UMR3691, Institut Pasteur, Paris, France
| | | | | | - Etienne Simon-Lorière
- Institut Pasteur, Unité de Génétique Fonctionnelle des Maladies Infectieuses, Paris, France
- CNRS URA 3012, Paris, France
| | - Arnaud Moris
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1135, CNRS ERL 8255, Center for Immunology and Microbial Infections - CIMI-Paris, Paris, France
| | - Philippe Roingeard
- INSERM U966 & Platefome IBiSA de Microscopie Electronique, Université François Rabelais and CHRU de Tours, Paris, France
| | - Ali Amara
- INSERM U944, CNRS 7212 Laboratoire de Pathologie et Virologie Moléculaire, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - Olivier Schwartz
- Virus & Immunity Unit, Institut Pasteur, Paris, France
- UMR CNRS 3569, Paris, France
| |
Collapse
|
120
|
Wang X, Xu X, Wang W, Yu Z, Wen L, He K, Fan H. MicroRNA-30a-5p promotes replication of porcine circovirus type 2 through enhancing autophagy by targeting 14-3-3. Arch Virol 2017; 162:2643-2654. [PMID: 28530014 DOI: 10.1007/s00705-017-3400-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/31/2017] [Indexed: 12/11/2022]
Abstract
Accumulating evidence demonstrates that autophagy and microRNAs (miRNAs) play key roles in regulating virus-host interactions and can restrict or facilitate viral replication. In the present study we examined whether a functional relationship exists between autophagy, miRNA and porcine circovirus type 2 (PCV2) infection, using several approaches. We demonstrated that there was a positive correlation between PCV2 infection and autophagy in 3D4/21 cells and autophagy induced by PCV2 infection triggered PCV2 replication. Four miRNA were selected by real-time PCR and further studied, but only miR-30a-5p mimic had a significant effect on PCV2 replication. Overexpression of miR-30a-5p significantly enhanced PCV2 infection and autophagy in a dose-dependent manner. Blockage of miR-30a-5p significantly decreased PCV2 replication. We provided further evidence that miR-30a-5p regulate the link between PCV2 infection and host immune system. Furthermore, miR-30a-5p targeted and regulated 14-3-3 gene, which is a regulator of autophagy. Flow cytometry data demonstrated that miR-30a-5p promotes cell cycle arrest at the G2 phase to regulate PCV2 replication and autophagy by interacting directly with 14-3-3, but not with the PCV2 genome. These data not only provide new insights into virus-host interactions during PCV2 infection but also suggest a potential new antiviral therapeutic strategy against PCV2 infection.
Collapse
Affiliation(s)
- Xiaomin Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China
| | - Xianglan Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.,Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China
| | - Wei Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China
| | - Zhengyu Yu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China
| | - Libin Wen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China
| | - Kongwang He
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, National Center for Engineering Research of Veterinary Bio-products, Nanjing, 210014, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| | - Hongjie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
121
|
Feng C, Fu Y, Chen D, Wang H, Su A, Zhang L, Chang L, Zheng N, Wu Z. miR-127-5p negatively regulates enterovirus 71 replication by directly targeting SCARB2. FEBS Open Bio 2017; 7:747-758. [PMID: 28593131 PMCID: PMC5458453 DOI: 10.1002/2211-5463.12197] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/27/2016] [Accepted: 01/16/2017] [Indexed: 12/27/2022] Open
Abstract
Enterovirus 71 (EV71) is the major causative agent of hand‐foot‐and‐mouth disease in young children and can cause severe cerebral and pulmonary complications and even fatality. This study aimed at elucidating whether and how EV71 infection is regulated by a cellular microRNA, miR‐127‐5p. We found that miR‐127‐5p can downregulate the expression of SCARB2, a main receptor of EV71, by targeting two potential sites in its 3′ UTR region and inhibit EV71 infection. Meanwhile, miR‐127‐5p expression was upregulated during EV71 infection. Notably, transfecting cells with miR‐127‐5p mimics led to a significant decrease in viral replication, while inhibition of endogenous miR‐127‐5p facilitated viral replication. Furthermore, our evidence showed that miR‐127‐5p did not affect postentry viral replication. Taken together, these results indicated that miR‐127‐5p inhibited EV71 replication by targeting the SCARB2 mRNA.
Collapse
Affiliation(s)
- Chunhong Feng
- Center for Public Health Research Medical School Nanjing University China.,School of life sciences Nanjing University China
| | - Yuxuan Fu
- Center for Public Health Research Medical School Nanjing University China
| | - Deyan Chen
- Center for Public Health Research Medical School Nanjing University China
| | - Huanru Wang
- Center for Public Health Research Medical School Nanjing University China
| | - Airong Su
- Center for Public Health Research Medical School Nanjing University China
| | - Li Zhang
- Center for Public Health Research Medical School Nanjing University China
| | - Liang Chang
- Center for Public Health Research Medical School Nanjing University China
| | - Nan Zheng
- Center for Public Health Research Medical School Nanjing University China.,State Key Lab of Analytical Chemistry for Life Science Nanjing University China.,Medical School and Jiangsu Key Laboratory of Molecular Medicine Nanjing University China
| | - Zhiwei Wu
- Center for Public Health Research Medical School Nanjing University China.,State Key Lab of Analytical Chemistry for Life Science Nanjing University China.,Medical School and Jiangsu Key Laboratory of Molecular Medicine Nanjing University China
| |
Collapse
|
122
|
Benzyl isothiocyanate induces protective autophagy in human lung cancer cells through an endoplasmic reticulum stress-mediated mechanism. Acta Pharmacol Sin 2017; 38:539-550. [PMID: 28112178 DOI: 10.1038/aps.2016.146] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/15/2016] [Indexed: 02/08/2023]
Abstract
Isothiocyanates, such as allyl isothiocya¬nate (AITC), benzyl isothiocyanate (BITC), phenethyl isothio¬cyanate (PEITC) and sulforaphane (SFN), are natural compounds abundant in cruciferous vegetables, which have substantial chemopreventive activities against various human malignancies. However, the mechanisms underlying the inhibition of tumor cell growth by isothiocyanates are not fully understood. Since autophagy has dual functions in cancer, in the present study we investigated the effects of BITC on autophagy induction in human lung cancer cells in vitro and in vivo. BITC (1-100 μmol/L) dose-dependently inhibited the growth of 3 different human lung cancer cell lines A549 (adenocarcinoma), H661 (large cell carcinoma) and SK-MES-1 (squamous cell carcinoma) with IC50 values of 30.7±0.14, 15.9±0.22 and 23.4±0.11 μmol/L, respectively. BITC (10-40 μmol/L) induced autophagy in the lung cancer cells, evidenced by the formation of acidic vesicular organelles (AVOs), the accumulation of LC3-II, the punctate pattern of LC3, and the expression of Atg5. Pretreatment with the autophagy inhibitor 3-MA (5 mmol/L) significantly enhanced the BITC-caused growth inhibition in the lung cancer cells. Furthermore, BITC (20-40 μmol/L) activated ER stress, as shown by the increased cytosolic Ca2+ level and the phosphorylation of the ER stress marker proteins PERK and eIF2α in the lung cancer cells. Pretreatment with the ER stress inhibitor 4-PBA (5 mmol/L) attenuated the autophagy induction and potentiated the BITC-induced cell growth inhibition. In nude mice bearing A549 xenografts, administration of BITC (100 mg·kg-1·d-1, ip) for 8 weeks markedly suppressed the lung tumor growth, and significantly enhanced both autophagy and ER stress in the tumor tissues. Our results demonstrate that BITC inhibits human lung cancer cell growth in vitro and in vivo. In addition, BITC induces autophagy in the lung cancer cells, which protects the cancer cells against the inhibitory action of BITC; the autophagy induction is mediated by the ER stress response.
Collapse
|
123
|
Chinchwadkar S, Padmanabhan S, Mishra P, Singh S, Suresh SN, Vats S, Barve G, Ammanathan V, Manjithaya R. Multifaceted Housekeeping Functions of Autophagy. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0015-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
124
|
Evaluation of Possible Consequences of Zika Virus Infection in the Developing Nervous System. Mol Neurobiol 2017; 55:1620-1629. [DOI: 10.1007/s12035-017-0442-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/03/2017] [Indexed: 01/05/2023]
|
125
|
Zhang J, Feng Z, Wang C, Zhou H, Liu W, Kanchana K, Dai X, Zou P, Gu J, Cai L, Liang G. Curcumin derivative WZ35 efficiently suppresses colon cancer progression through inducing ROS production and ER stress-dependent apoptosis. Am J Cancer Res 2017; 7:275-288. [PMID: 28337376 PMCID: PMC5336501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/04/2016] [Indexed: 06/06/2023] Open
Abstract
Colon cancer is characterized by its fast progression and poor prognosis, and novel agents of treating colon cancer are urgently needed. WZ35, a synthetic curcumin derivative, has been reported to exhibit promising antitumor activity. Here, we investigated the in vitro and in vivo activities of WZ35 and explored the underlying mechanisms in colon cancer cell lines. WZ35 treatment significantly decreased the cell viability associated with G2/M cell cycle arrest and apoptosis induction in colon cancer cell lines. We also show that WZ35 is highly effective in inhibiting tumor growth in a CT26 xenograft mouse model. Mechanistically, WZ35 treatment significantly induced reactive oxygen species (ROS) generation and endoplasmic reticulum (ER) stress in CT26 cells. Abrogation of ROS production by N-acetylcysteine (NAC) co-treatment almost totally reversed the WZ35-induced cell apoptosis and ER stress activation. Inhibition of p-PERK by GSK2606414 can significantly reverse WZ35-induced cell apoptosis in CT26 cells. Taken together, the curcumin derivative WZ35 exhibited anti-tumor effects in colon cancer cells both in vitro and in vivo, via a ROS-ER stress-mediated mechanism. These findings indicate that activating ROS generation could be an important strategy for the treatment of colon cancers.
Collapse
Affiliation(s)
- Junru Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
- School of Pharmaceutical Science, Binzhou Medical UniversityYantai, Shandong 264003, China
| | - Zhiguo Feng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| | - Chunhua Wang
- School of Pharmaceutical Science, Binzhou Medical UniversityYantai, Shandong 264003, China
| | - Huiping Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| | - Weidong Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| | - Karvannan Kanchana
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| | - Xuanxuan Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| | - Peng Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| | - Junlian Gu
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
- Kosair Children’s Hospital Research Institute, Department of Pediatrics of The University of LouisvilleLouisville, KY 40202, USA
| | - Lu Cai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
- Kosair Children’s Hospital Research Institute, Department of Pediatrics of The University of LouisvilleLouisville, KY 40202, USA
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical UniversityWenzhou, Zhejiang 325035, China
| |
Collapse
|
126
|
Fan X, Han S, Yan D, Gao Y, Wei Y, Liu X, Liao Y, Guo H, Sun S. Foot-and-mouth disease virus infection suppresses autophagy and NF-кB antiviral responses via degradation of ATG5-ATG12 by 3C pro. Cell Death Dis 2017; 8:e2561. [PMID: 28102839 PMCID: PMC5386389 DOI: 10.1038/cddis.2016.489] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/30/2016] [Accepted: 12/19/2016] [Indexed: 12/22/2022]
Abstract
Autophagy-related protein ATG5-ATG12 is an essential complex for the autophagophore elongation in autophagy, which has been reported to be involved in foot-and-mouth disease virus (FMDV) replication. Previous reports show that ATG5-ATG12 positively or negatively regulates type I interferon (IFN-α/β) pathway during virus infection. In this study, we found that FMDV infection rapidly induced LC3 lipidation and GFP-LC3 subcellular redistribution at the early infection stage in PK-15 cells. Along with infection time course to 2-5 h.p.i., the levels of LC3II and ATG5-ATG12 were gradually reduced. Further study showed that ATG5-ATG12 was degraded by viral protein 3Cpro, demonstrating that FMDV suppresses autophagy along with viral protein production. Depletion of ATG5-ATG12 by siRNA knock down significantly increased the FMDV yields, whereas overexpression of ATG5-ATG12 had the opposite effects, suggesting that degradation of ATG5-ATG12 benefits virus growth. Further experiment showed that overexpression of ATG5-ATG12 positively regulated NF-кB pathway during FMDV infection, marked with promotion of IKKα/β phosphorylation and IκBα degradation, inhibition of p65 degradation, and facilitation of p65 nuclear translocation. Meanwhile, ATG5-ATG12 also promoted the phosphorylation of TBK1 and activation of IRF3 via preventing TRAF3 degradation. The positive regulation of NF-кB and IRF3 pathway by ATG5-ATG12 resulted in enhanced expression of IFN-β, chemokines/cytokines, and IFN stimulated genes, including anti-viral protein PKR. Altogether, above findings suggest that ATG5-ATG12 positively regulate anti-viral NF-κB and IRF3 signaling during FMDV infection, thereby limiting FMDV proliferation. FMDV has evolved mechanisms to counteract the antiviral function of ATG5-ATG12, via degradation of them by viral protein 3Cpro.
Collapse
Affiliation(s)
- Xuxu Fan
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Shichong Han
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing, P. R. China
| | - Dan Yan
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Yuan Gao
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Yanquan Wei
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Xiangtao Liu
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Ying Liao
- Department of Avian Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, P. R. China
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Shiqi Sun
- State Key Laboratory of Veterinary Etiological Biology and National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| |
Collapse
|
127
|
Takahashi S, Sato N, Kikuchi J, Kakinuma H, Okawa J, Masuyama Y, Iwasa S, Irokawa H, Hwang GW, Naganuma A, Kohara M, Kuge S. Immature Core protein of hepatitis C virus induces an unfolded protein response through inhibition of ERAD-L in a yeast model system. Genes Cells 2017; 22:160-173. [PMID: 28097745 DOI: 10.1111/gtc.12464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/29/2016] [Indexed: 01/18/2023]
Abstract
The structural protein Core of hepatitis C virus (HCV), a cytosolic protein, induces endoplasmic reticulum (ER) stress and unfolded protein response (UPR) in hepatocytes, and is responsible for the pathogenesis of persistent HCV infection. Using yeast as a model system, we evaluated mechanisms underlying Core-induced interference of ER homeostasis and UPR, and found that UPR is induced by the immature Core (aa 1-191, Core191) but not by the mature Core (aa 1-177, Core177). Interestingly, Core191 inhibits both ERAD-L, a degradation system responsible for misfolded/unfolded proteins in the ER lumen, and ERAD-M, a degradation system responsible for proteins carrying a misfolded/unfolded region in the ER membrane. In contrast, Core177 inhibits ERAD-M but not ERAD-L. In addition, requirement of an unfolded protein sensor in the ER lumen suggested that inhibition of ERAD-L is probably responsible for Core191-dependent UPR activation. These results implicate inadequate maturation of Core as a trigger for induction of ER stress and UPR.
Collapse
Affiliation(s)
- Shota Takahashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Naoko Sato
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan.,Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Junichi Kikuchi
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Hideaki Kakinuma
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Jun Okawa
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Yukiko Masuyama
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Singo Iwasa
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Hayato Irokawa
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Gi-Wook Hwang
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Akira Naganuma
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Shusuke Kuge
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan.,Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| |
Collapse
|
128
|
Moon JY, Park JM. Cross-Talk in Viral Defense Signaling in Plants. Front Microbiol 2016; 7:2068. [PMID: 28066385 PMCID: PMC5174109 DOI: 10.3389/fmicb.2016.02068] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/07/2016] [Indexed: 01/19/2023] Open
Abstract
Viruses are obligate intracellular parasites that have small genomes with limited coding capacity; therefore, they extensively use host intracellular machinery for their replication and infection in host cells. In recent years, it was elucidated that plants have evolved intricate defense mechanisms to prevent or limit damage from such pathogens. Plants employ two major strategies to counteract virus infections: resistance (R) gene-mediated and RNA silencing-based defenses. In this review, plant defenses and viral counter defenses are described, as are recent studies examining the cross-talk between different plant defense mechanisms.
Collapse
Affiliation(s)
- Ju Y. Moon
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and BiotechnologyDaejeon, South Korea
- Department of Biosystems and Bioengineering, University of Science and TechnologyDaejeon, South Korea
| | - Jeong M. Park
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and BiotechnologyDaejeon, South Korea
- Department of Biosystems and Bioengineering, University of Science and TechnologyDaejeon, South Korea
| |
Collapse
|
129
|
Hu DD, Mai JN, He LY, Li PQ, Chen WX, Yan JJ, Zhu WD, Deng L, Wei D, Liu DH, Yang SD, Yao ZB. Glucocorticoids Prevent Enterovirus 71 Capsid Protein VP1 Induced Calreticulin Surface Exposure by Alleviating Neuronal ER Stress. Neurotox Res 2016; 31:204-217. [PMID: 27848175 DOI: 10.1007/s12640-016-9670-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/26/2016] [Accepted: 09/15/2016] [Indexed: 12/11/2022]
Abstract
Severe hand-foot-and-mouth disease (HFMD) caused by Enterovirus 71 (EV71) always accompanies with inflammation and neuronal damage in the central nervous system (CNS). During neuronal injuries, cell surface-exposed calreticulin (Ecto-CRT) is an important mediator for primary phagocytosis of viable neurons by microglia. Our data confirmed that brainstem neurons underwent neuronophagia by glia in EV71-induced death cases of HFMD. EV71 capsid proteins VP1, VP2, VP3, or VP4 did not induce apoptosis of brainstem neurons. Interestingly, we found VP1-activated endoplasmic reticulum (ER) stress and autophagy could promote Ecto-CRT upregulation, but ER stress or autophagy alone was not sufficient to induce CRT exposure. Furthermore, we demonstrated that VP1-induced autophagy activation was mediated by ER stress. Meaningfully, we found dexamethasone treatment could attenuate Ecto-CRT upregulation by alleviating VP1-induced ER stress. Altogether, these findings identify VP1-promoted Ecto-CRT upregulation as a novel mechanism of EV71-induced neuronal cell damage and highlight the potential of the use of glucocorticoids to treat severe HFMD patients with CNS complications.
Collapse
Affiliation(s)
- Dan-Dan Hu
- Department of Anatomy Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.,Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Jian-Ning Mai
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Li-Ya He
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Pei-Qing Li
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Wen-Xiong Chen
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Jian-Jiang Yan
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Wei-Dong Zhu
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Li Deng
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China
| | - Dan Wei
- First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Di-Hui Liu
- United Family Healthcare Guangzhou Clinic, Guangzhou, 510600, China
| | - Si-Da Yang
- Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong, China.
| | - Zhi-Bin Yao
- Department of Anatomy Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
130
|
Khongwichit S, Wikan N, Abere B, Thepparit C, Kuadkitkan A, Ubol S, Smith DR. Cell-type specific variation in the induction of ER stress and downstream events in chikungunya virus infection. Microb Pathog 2016; 101:104-118. [PMID: 27863885 DOI: 10.1016/j.micpath.2016.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 09/27/2016] [Accepted: 11/14/2016] [Indexed: 12/25/2022]
Abstract
Over the last decade infections with the mosquito transmitted chikungunya virus (CHIKV) have become a major worldwide concern, and considerable efforts have been made in understanding the interaction of this virus with the host cell machinery. Studies have documented the induction of the unfolded protein response (UPR), as well as the induction of apoptosis and autophagy in response to CHIKV infection. This study comparatively analysed these three processes in two cell lines, Hela and HepG2. Infection of Hela cells was characterized by activation of the PERK/eIF2α branch of the UPR, the induction of autophagy and early apoptosis, while infection of HepG2 cells was characterized by activation of the IRE/XBP1 branch of the UPR, limited or no activation of autophagy and comparatively later apoptosis. These results show that the specific cell context is an important mediator of the host cell response to CHIKV infection.
Collapse
Affiliation(s)
- Sarawut Khongwichit
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Nakhon Pathom, Thailand
| | - Nitwara Wikan
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Nakhon Pathom, Thailand
| | - Bizunesh Abere
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Nakhon Pathom, Thailand
| | - Chutima Thepparit
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Nakhon Pathom, Thailand
| | - Atichat Kuadkitkan
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Nakhon Pathom, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Nakhon Pathom, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Nakhon Pathom, Thailand; Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Nakhon Pathom, Thailand.
| |
Collapse
|
131
|
Inhibition of autophagy enhances heat-induced apoptosis in human non-small cell lung cancer cells through ER stress pathways. Arch Biochem Biophys 2016; 607:55-66. [DOI: 10.1016/j.abb.2016.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 08/17/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022]
|
132
|
The innate immune response to RSV: Advances in our understanding of critical viral and host factors. Vaccine 2016; 35:481-488. [PMID: 27686836 DOI: 10.1016/j.vaccine.2016.09.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/30/2016] [Accepted: 09/15/2016] [Indexed: 12/14/2022]
Abstract
Respiratory syncytial virus (RSV) causes mild to severe respiratory illness in humans and is a major cause of hospitalizations of infants and the elderly. Both the innate and the adaptive immune responses contribute to the control of RSV infection, but despite successful viral clearance, protective immunity against RSV re-infection is usually suboptimal and infections recur. Poor understanding of the mechanisms limiting the induction of long-lasting immunity has delayed the development of an effective vaccine. The innate immune response plays a critical role in driving the development of adaptive immunity and is thus a crucial determinant of the infection outcome. Advances in recent years have improved our understanding of cellular and viral factors that influence the onset and quality of the innate immune response to RSV. These advances include the identification of a complex system of cellular sensors that mediate RSV detection and stimulate transcriptome changes that lead to virus control and the discovery that cell stress and apoptosis participate in the control of RSV infection. In addition, it was recently demonstrated that defective viral genomes (DVGs) generated during RSV replication are the primary inducers of the innate immune response. Newly discovered host pathways involved in the innate response to RSV, together with the potential generation of DVG-derived oligonucleotides, present various novel opportunities for the design of vaccine adjuvants able to induce a protective response against RSV and similar viruses.
Collapse
|
133
|
Frabutt DA, Zheng YH. Arms Race between Enveloped Viruses and the Host ERAD Machinery. Viruses 2016; 8:v8090255. [PMID: 27657106 PMCID: PMC5035969 DOI: 10.3390/v8090255] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/12/2022] Open
Abstract
Enveloped viruses represent a significant category of pathogens that cause serious diseases in animals. These viruses express envelope glycoproteins that are singularly important during the infection of host cells by mediating fusion between the viral envelope and host cell membranes. Despite low homology at protein levels, three classes of viral fusion proteins have, as of yet, been identified based on structural similarities. Their incorporation into viral particles is dependent upon their proper sub-cellular localization after being expressed and folded properly in the endoplasmic reticulum (ER). However, viral protein expression can cause stress in the ER, and host cells respond to alleviate the ER stress in the form of the unfolded protein response (UPR); the effects of which have been observed to potentiate or inhibit viral infection. One important arm of UPR is to elevate the capacity of the ER-associated protein degradation (ERAD) pathway, which is comprised of host quality control machinery that ensures proper protein folding. In this review, we provide relevant details regarding viral envelope glycoproteins, UPR, ERAD, and their interactions in host cells.
Collapse
Affiliation(s)
- Dylan A Frabutt
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.
| | - Yong-Hui Zheng
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
134
|
Acosta EG, Bartenschlager R. The quest for host targets to combat dengue virus infections. Curr Opin Virol 2016; 20:47-54. [PMID: 27648486 DOI: 10.1016/j.coviro.2016.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/15/2022]
Abstract
Dengue virus (DENV) is a human threat of increasing importance. Although a tetravalent vaccine has been recently approved, owing to limited efficacy there is still an urgent need for antiviral drugs to prevent or treat DENV infections. Traditionally, antiviral drug discovery has focused on molecules targeting viral factors. However, thus far the identification of direct-acting antiviral drugs with potent DENV pan-serotypic activity has been problematic. An alternative are host-targeting antiviral drugs that hold great promise for broad-spectrum activity. In this review, we summarize cellular factors and pathways required by DENV for efficient replication and in principle suitable for antiviral therapy, including host-directed inhibitors that have even been advanced into clinical trials.
Collapse
Affiliation(s)
- Eliana G Acosta
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany.
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany; German Center for Infection Research (DZIF), Partner-site Heidelberg, Germany.
| |
Collapse
|
135
|
Affiliation(s)
- Yin Mo
- Ministry of Health Holdings, University Medicine Cluster, National University Health System, 1 Maritime Square, Singapore 099253, Singapore Division of Infectious Diseases, University Medicine Cluster, National University Health System, 1E Kent Ridge Road, Level 10, 119228, Singapore
| | - Brenda Mae Alferez Salada
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, 1E Kent Ridge Road, Level 10, 119228, Singapore
| | - Paul Anantharajah Tambyah
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, 1E Kent Ridge Road, Level 10, 119228, Singapore Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Rd 119228, NUHS Tower Block, Level 11, Singapore 117597, Singapore
| |
Collapse
|
136
|
Abstract
Understanding the molecular mechanisms behind the capacity of cancer cells to adapt to the tumor microenvironment and to anticancer therapies is a major challenge. In this context, cancer is believed to be an evolutionary process where random mutations and the selection process shape the mutational pattern and phenotype of cancer cells. This article challenges the notion of randomness of some cancer-associated mutations by describing molecular mechanisms involving stress-mediated biogenesis of mRNA-derived small RNAs able to target and increase the local mutation rate of the genomic loci they originate from. It is proposed that the probability of some mutations at specific loci could be increased in a stress-specific and RNA-depending manner. This would increase the probability of generating mutations that could alleviate stress situations, such as those triggered by anticancer drugs. Such a mechanism is made possible because tumor- and anticancer drug-associated stress situations trigger both cellular reprogramming and inflammation, which leads cancer cells to express molecular tools allowing them to “attack” and mutate their own genome in an RNA-directed manner.
Collapse
Affiliation(s)
- Didier Auboeuf
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, Lyon, France
| |
Collapse
|
137
|
Proteomic analysis of the secretome of HepG2 cells indicates differential proteolytic processing after infection with dengue virus. J Proteomics 2016; 151:106-113. [PMID: 27427332 DOI: 10.1016/j.jprot.2016.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 06/27/2016] [Accepted: 07/13/2016] [Indexed: 01/04/2023]
Abstract
Secretome analysis can be described as a subset of proteomics studies consisting in the analysis of the molecules secreted by cells or tissues. Dengue virus (DENV) infection can lead to a broad spectrum of clinical manifestations, with the severe forms of the disease characterized by hemostasis abnormalities and liver injury. The hepatocytes are a relevant site of viral replication and a major source of plasma proteins. Until now, we had limited information on the small molecules secreted by hepatic cells after infection by DENV. In the present study, we analysed a fraction of the secretome of mock- and DENV-infected hepatic cells (HepG2 cells) containing molecules with <10kDa, using different proteomic approaches. We identified 175 proteins, with 57 detected only in the samples from mock-infected cells, 59 only in samples from DENV-infected cells, and 59 in both conditions. Most of the peptides identified were derived from proteins larger than 10kDa, suggesting a proteolytic processing of the secreted molecules. Using in silico analysis, we predicted consistent differences between the proteolytic processing occurring in mock and DENV-infected samples, raising, for the first time, the hypothesis that differential proteolysis of secreted molecules would be involved in the pathogenesis of dengue. BIOLOGICAL SIGNIFICANCE Since the liver, one of the targets of DENV infection, is responsible for producing molecules involved in distinct biological processes, the identification of proteins and peptides secreted by hepatocytes after infection would help to a better understanding of the physiopathology of dengue. Proteomic analyses of molecules with <10kDa secreted by HepG2 cells after infection with DENV revealed differential proteolytic processing as an effect of DENV infection.
Collapse
|
138
|
Wang A, Zhou X. ER Stress, UPR and Virus Infections in Plants. CURRENT RESEARCH TOPICS IN PLANT VIROLOGY 2016. [PMCID: PMC7123154 DOI: 10.1007/978-3-319-32919-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
The endoplasmic reticulum (ER) endomembrane is a central site for protein synthesis. Perturbation of ER homeostasis can result in an accumulation of unfolded proteins within the ER lumen, causing ER stress and the unfolded protein response (UPR). In humans, ER stress and UPR are closely associated with a vast number of diseases, including viral diseases. In plants, two arms that govern the UPR signaling network have been described: one that contains two ER membrane–associated transcription factors (bZIP17 and bZIP28) and the other that encompasses a dual protein kinase (RNA-splicing factor IRE1) and its target RNA (bZIP60). Although early studies mainly focus on the essential roles of the UPR in abiotic stresses, the significance of UPR in plant diseases caused by virus infections has recently drawn much attention. This chapter summarizes the latest scenario of ER stress and UPR in virus-infected plant cells, highlights the emerging roles of the IRE1 pathway in virus infections, and outlines exciting future directions to spark more research interest in the UPR field in plants.
Collapse
Affiliation(s)
- Aiming Wang
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, Ontario Canada
| | - Xueping Zhou
- State Key Laboratory for Biology of Plan, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
139
|
Prieto-Domínguez N, Ordóñez R, Fernández A, García-Palomo A, Muntané J, González-Gallego J, Mauriz JL. Modulation of Autophagy by Sorafenib: Effects on Treatment Response. Front Pharmacol 2016; 7:151. [PMID: 27375485 PMCID: PMC4896953 DOI: 10.3389/fphar.2016.00151] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
The multikinase inhibitor sorafenib is, at present, the only drug approved for the treatment of hepatocellular carcinoma (HCC), one of the most lethal types of cancer worldwide. However, the increase in the number of sorafenib tumor resistant cells reduces efficiency. A better knowledge of the intracellular mechanism of the drug leading to reduced cell survival could help to improve the benefits of sorafenib therapy. Autophagy is a bulk cellular degradation process activated in a broad range of stress situations, which allows cells to degrade misfolded proteins or dysfunctional organelles. This cellular route can induce survival or death, depending on cell status and media signals. Sorafenib, alone or in combination with other drugs is able to induce autophagy, but cell response to the drug depends on the complex integrative crosstalk of different intracellular signals. In cancerous cells, autophagy can be regulated by different cellular pathways (Akt-related mammalian target of rapamycin (mTOR) inhibition, 5′ AMP-activated protein kinase (AMPK) induction, dissociation of B-cell lymphoma 2 (Bcl-2) family proteins from Beclin-1), or effects of some miRNAs. Inhibition of mTOR signaling by sorafenib and diminished interaction between Beclin-1 and myeloid cell leukemia 1 (Mcl-1) have been related to induction of autophagy in HCC. Furthermore, changes in some miRNAs, such as miR-30α, are able to modulate autophagy and modify sensitivity in sorafenib-resistant cells. However, although AMPK phosphorylation by sorafenib seems to play a role in the antiproliferative action of the drug, it does not relate with modulation of autophagy. In this review, we present an updated overview of the effects of sorafenib on autophagy and its related activation pathways, analyzing in detail the involvement of autophagy on sorafenib sensitivity and resistance.
Collapse
Affiliation(s)
- Nestor Prieto-Domínguez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - Raquel Ordóñez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - Anna Fernández
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - Andres García-Palomo
- Service of Clinical Oncology, Complejo Asistencial Universitario de León (Hospital of León) León, Spain
| | - Jordi Muntané
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Department of General Surgery"Virgen del Rocío"-"Virgen Macarena" University Hospital/IBiS/CSIC/Universidad de Sevilla, Spain
| | - Javier González-Gallego
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| | - José L Mauriz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)León, Spain; Institute of Biomedicine (IBIOMED), University of LeónLeón, Spain
| |
Collapse
|
140
|
Ravindran MS, Bagchi P, Cunningham CN, Tsai B. Opportunistic intruders: how viruses orchestrate ER functions to infect cells. Nat Rev Microbiol 2016; 14:407-420. [PMID: 27265768 PMCID: PMC5272919 DOI: 10.1038/nrmicro.2016.60] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Viruses exploit the functions of the endoplasmic reticulum (ER) to promote both early and later stages of their life cycle, including entry, translation, replication, assembly, morphogenesis and egress. This observation reveals a shared principle that underlies virus–host cell relationships. Viral entry often requires disassembly of the incoming virus particle. This is best exemplified in the case of polyomavirus entry, in which ER-associated machineries are hijacked to disassemble the virus and promote entry to the cytosol en route to the nucleus. Many enveloped viruses, such as HIV and influenza virus, co-opt the ER-associated protein biosynthetic machinery to translate their genome and produce structural proteins that are necessary for the formation of virus particles and non-structural proteins that are essential during genome replication. Replication of the viral genome, particularly for positive-sense RNA ((+)RNA) viruses including hepatitis C virus (HCV), dengue virus (DENV) and West Nile virus (WNV), occurs in virus-induced membranous structures that are most often derived from the ER. The formation of these structures requires morphological changes to the ER membrane, involving membrane rearrangements that are induced by viral non-structural proteins that are targeted to the ER. As virus assembly is often coupled to genome replication, the assembly process frequently relies on the ER membrane. This strategy is seen for both RNA and DNA viruses. Morphogenesis of assembled virus particles can also take advantage of the ER. This is best observed in the non-enveloped rotavirus, for which a transient enveloped intermediate is converted to the mature and infectious particle in the lumen of the ER. After maturation in the ER, progeny virus particles egress the host through the ER-dependent secretory pathway, which provides a physical conduit to the extracellular environment. The overall observations that the ER actively promotes all steps of viral infection have therapeutic implications. The development of chemical inhibitors of selective ER-associated components is emerging as a potential avenue of antiviral therapy, provided that these inhibitors have minimal toxicity to the host cell.
Many host structures are vital for viral infection and the endoplasmic reticulum (ER), in particular, is essential. In this Review, Tsai and colleagues highlight examples of subversion of the ER by diverse viruses to promote all stages of their life cycle, from entry to egress. Viruses subvert the functions of their host cells to replicate and form new viral progeny. The endoplasmic reticulum (ER) has been identified as a central organelle that governs the intracellular interplay between viruses and hosts. In this Review, we analyse how viruses from vastly different families converge on this unique intracellular organelle during infection, co-opting some of the endogenous functions of the ER to promote distinct steps of the viral life cycle from entry and replication to assembly and egress. The ER can act as the common denominator during infection for diverse virus families, thereby providing a shared principle that underlies the apparent complexity of relationships between viruses and host cells. As a plethora of information illuminating the molecular and cellular basis of virus–ER interactions has become available, these insights may lead to the development of crucial therapeutic agents.
Collapse
Affiliation(s)
- Madhu Sudhan Ravindran
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Room 3043, Ann Arbor, Michigan 48109, USA
| | - Parikshit Bagchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Room 3043, Ann Arbor, Michigan 48109, USA
| | - Corey Nathaniel Cunningham
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Room 3043, Ann Arbor, Michigan 48109, USA
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Room 3043, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
141
|
Barel M, Harduin-Lepers A, Portier L, Slomianny MC, Charbit A. Host glycosylation pathways and the unfolded protein response contribute to the infection by Francisella. Cell Microbiol 2016; 18:1763-1781. [PMID: 27185209 DOI: 10.1111/cmi.12614] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/09/2016] [Accepted: 05/09/2016] [Indexed: 12/14/2022]
Abstract
Protein glycosylation processes play a crucial role in most physiological functions, including cell signalling, cellular differentiation and adhesion. We previously demonstrated that rapid deglycosylation of membrane proteins was specifically triggered after infection of human macrophages by the bacterial pathogen Francisella tularensis. Using a glycan processing gene microarray, we found here that Francisella infection modulated expression of numerous glycosidase and glycosyltransferase genes. Furthermore, analysis of cell extracts from infected macrophages by Lectin and Western blotting revealed an important increase of N- and O-protein glycosylation. We chose to focus in the present work on one of the O-glycosylated proteins identified by mass spectrometry, the multifunctional endoplasmic reticulum chaperone BiP (HSPA5/GRP78). We demonstrate that BiP expression is modulated upon Francisella infection and is required to support its intracellular multiplication. Moreover, we show that Francisella differentially modulates the BiP-dependent activation of three key proteins of the unfolded protein response (UPR), IRE1, PERK and ATF6. The effects exerted on human cells by Francisella may thus constitute a novel excample of UPR manipulation contributing to intracellular bacterial adaptation.
Collapse
Affiliation(s)
- Monique Barel
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Institut Necker Enfants-Malades INSERM, U1151, Team 11, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FR 59000, Lille, France.,UGSF, Bat. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Lucie Portier
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FR 59000, Lille, France.,UGSF, Bat. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Marie-Christine Slomianny
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FR 59000, Lille, France.,UGSF, Bat. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Alain Charbit
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Institut Necker Enfants-Malades INSERM, U1151, Team 11, Unité de Pathogénie des Infections Systémiques, Paris, France
| |
Collapse
|
142
|
Hepatitis C Virus Infection Induces Autophagy as a Prosurvival Mechanism to Alleviate Hepatic ER-Stress Response. Viruses 2016; 8:v8050150. [PMID: 27223299 PMCID: PMC4885105 DOI: 10.3390/v8050150] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/04/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022] Open
Abstract
Hepatitis C virus (HCV) infection frequently leads to chronic liver disease, liver cirrhosis and hepatocellular carcinoma (HCC). The molecular mechanisms by which HCV infection leads to chronic liver disease and HCC are not well understood. The infection cycle of HCV is initiated by the attachment and entry of virus particles into a hepatocyte. Replication of the HCV genome inside hepatocytes leads to accumulation of large amounts of viral proteins and RNA replication intermediates in the endoplasmic reticulum (ER), resulting in production of thousands of new virus particles. HCV-infected hepatocytes mount a substantial stress response. How the infected hepatocyte integrates the viral-induced stress response with chronic infection is unknown. The unfolded protein response (UPR), an ER-associated cellular transcriptional response, is activated in HCV infected hepatocytes. Over the past several years, research performed by a number of laboratories, including ours, has shown that HCV induced UPR robustly activates autophagy to sustain viral replication in the infected hepatocyte. Induction of the cellular autophagy response is required to improve survival of infected cells by inhibition of cellular apoptosis. The autophagy response also inhibits the cellular innate antiviral program that usually inhibits HCV replication. In this review, we discuss the physiological implications of the HCV-induced chronic ER-stress response in the liver disease progression.
Collapse
|
143
|
Culjat M, Darling SE, Nerurkar VR, Ching N, Kumar M, Min SK, Wong R, Grant L, Melish ME. Clinical and Imaging Findings in an Infant With Zika Embryopathy. Clin Infect Dis 2016; 63:805-11. [PMID: 27193747 DOI: 10.1093/cid/ciw324] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/11/2016] [Indexed: 01/02/2023] Open
Abstract
Recent Zika virus (ZIKV) outbreaks have been associated with an increased incidence of neonatal microcephaly. Subsequently, tropism for the brain was established in human fetal brain tissue. We present the first congenital ZIKV infection in the United States, confirmed by high ZIKV immunoglobulin M antibody titers in serum and cerebrospinal fluid. The phenotypic characteristics of the patient fall within fetal brain disruption sequence, suggesting impaired brain development in the second half of gestation. Brain imaging revealed an almost agyric brain with diffuse parenchymal calcifications, hydrocephalus ex vacuo, and cerebellar hypoplasia. Ophthalmologic examination revealed macular pigment stippling and optic nerve atrophy. Liver, lungs, heart, and bone marrow were not affected. The patient had progressive neurologic deterioration in the first month of life. The discovery of ZIKV infection in human fetal brain tissue along with serologic confirmation proves the vertical transmission of ZIKV. Therefore, ZIKV has joined the group of congenital infections.
Collapse
Affiliation(s)
| | - Stephen E Darling
- Department of Radiology, Kapi'olani Medical Center for Women and Children
| | - Vivek R Nerurkar
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, University of Hawai'i at Manoa
| | | | - Mukesh Kumar
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, University of Hawai'i at Manoa
| | | | - Rupa Wong
- Kapi'olani Medical Center for Women and Children
| | | | - Marian E Melish
- Kapi'olani Medical Center for Women and Children Department of Pediatrics, Department of Tropical Medicine, Medical Microbiology and Pharmacology, Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, Honolulu, Hawaii
| |
Collapse
|
144
|
Cervantes-Ortiz SL, Zamorano Cuervo N, Grandvaux N. Respiratory Syncytial Virus and Cellular Stress Responses: Impact on Replication and Physiopathology. Viruses 2016; 8:v8050124. [PMID: 27187445 PMCID: PMC4885079 DOI: 10.3390/v8050124] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/14/2016] [Accepted: 04/21/2016] [Indexed: 02/08/2023] Open
Abstract
Human respiratory syncytial virus (RSV), a member of the Paramyxoviridae family, is a major cause of severe acute lower respiratory tract infection in infants, elderly and immunocompromised adults. Despite decades of research, a complete integrated picture of RSV-host interaction is still missing. Several cellular responses to stress are involved in the host-response to many virus infections. The endoplasmic reticulum stress induced by altered endoplasmic reticulum (ER) function leads to activation of the unfolded-protein response (UPR) to restore homeostasis. Formation of cytoplasmic stress granules containing translationally stalled mRNAs is a means to control protein translation. Production of reactive oxygen species is balanced by an antioxidant response to prevent oxidative stress and the resulting damages. In recent years, ongoing research has started to unveil specific regulatory interactions of RSV with these host cellular stress responses. Here, we discuss the latest findings regarding the mechanisms evolved by RSV to induce, subvert or manipulate the ER stress, the stress granule and oxidative stress responses. We summarize the evidence linking these stress responses with the regulation of RSV replication and the associated pathogenesis.
Collapse
Affiliation(s)
- Sandra L Cervantes-Ortiz
- CRCHUM-Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X 0A9, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| | - Natalia Zamorano Cuervo
- CRCHUM-Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X 0A9, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| | - Nathalie Grandvaux
- CRCHUM-Centre Hospitalier de l'Université de Montréal, Montréal, QC H2X 0A9, Canada.
- Faculty of Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
145
|
Guerrero CA, Acosta O. Inflammatory and oxidative stress in rotavirus infection. World J Virol 2016; 5:38-62. [PMID: 27175349 PMCID: PMC4861870 DOI: 10.5501/wjv.v5.i2.38] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/23/2015] [Accepted: 01/29/2016] [Indexed: 02/05/2023] Open
Abstract
Rotaviruses are the single leading cause of life-threatening diarrhea affecting children under 5 years of age. Rotavirus entry into the host cell seems to occur by sequential interactions between virion proteins and various cell surface molecules. The entry mechanisms seem to involve the contribution of cellular molecules having binding, chaperoning and oxido-reducing activities. It appears to be that the receptor usage and tropism of rotaviruses is determined by the species, cell line and rotavirus strain. Rotaviruses have evolved functions which can antagonize the host innate immune response, whereas are able to induce endoplasmic reticulum (ER) stress, oxidative stress and inflammatory signaling. A networking between ER stress, inflammation and oxidative stress is suggested, in which release of calcium from the ER increases the generation of mitochondrial reactive oxygen species (ROS) leading to toxic accumulation of ROS within ER and mitochondria. Sustained ER stress potentially stimulates inflammatory response through unfolded protein response pathways. However, the detailed characterization of the molecular mechanisms underpinning these rotavirus-induced stressful conditions is still lacking. The signaling events triggered by host recognition of virus-associated molecular patterns offers an opportunity for the development of novel therapeutic strategies aimed at interfering with rotavirus infection. The use of N-acetylcysteine, non-steroidal anti-inflammatory drugs and PPARγ agonists to inhibit rotavirus infection opens a new way for treating the rotavirus-induced diarrhea and complementing vaccines.
Collapse
|
146
|
Bu X, Zhao Y, Zhang Z, Wang M, Li M, Yan Y. Recombinant Newcastle disease virus (rL-RVG) triggers autophagy and apoptosis in gastric carcinoma cells by inducing ER stress. Am J Cancer Res 2016; 6:924-936. [PMID: 27293989 PMCID: PMC4889710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/10/2016] [Indexed: 06/06/2023] Open
Abstract
We have reported that the recombinant avirulent Newcastle disease virus (NDV) LaSota strain expressing the rabies virus glycoprotein (rL-RVG) could induce autophagy and apoptosis in gastric carcinoma cells. In the present study, we explored the upstream regulators, endoplasmic reticulum (ER) stress that induce autophagy and apoptosis and the relationships among them. For this purpose, SGC-7901 and HGC cells were infected with rL-RVG. NDV LaSota strain and phosphate-buffered saline (PBS) were treated as the control groups. Western blotting and immunofluorescence microscopy were used to detect the expression of the ER stress-related proteins glucose-regulated protein 78 (GRP78) and the transcription factor GADD153 (CHOP), among others. The expression of beclin-1 and the conversion of light chain (LC) 3-I were used to determine the occurrence of autophagy, and flow cytometry (FCM) and western blotting were used to examine apoptosis-related protein expression. Transmission electron microscopy was also performed to monitor the ultrastructure of the cells. Moreover, small interfering (si) RNA was used to knock down CHOP expression. rL-RVG treatment increased the expression of ER stress-related proteins, such as GRP78, CHOP, activating transcriptional factor 6 (ATF6), X-box-binding protein 1 (XBP-1), and phosphorylated eukaryotic initiation factor 2 (p-eIF2α), in a time- and concentration-dependent manner, and knockdown of CHOP reduced LC3-II conversion and beclin-1 expression. When ER stress was inhibited with 4-PBA, the expression of both autophagy-related proteins and apoptosis-related proteins markedly decreased. Interestingly, inhibition of autophagy with 3-methyladenine (3MA) decreased not only apoptosis-related protein expression but also ER stress-related protein expression. Moreover, we found that downregulation of the c-Jun N-terminal kinase (JNK) pathway by SP600125 reduced LC3-II conversion, beclin-1 expression and caspase-3 activation. Collectively, the results suggest that rL-RVG increased ER stress in three branch pathways (ATF6, inositol-requiring enzyme 1 (IRE1), and PKR-like ER protein kinase (PERK)) that are upstream regulators of autophagy and apoptosis. Moreover, the IRE1-JNK pathway played an important role in switching ER stress to autophagy. These findings will provide molecular bases for developing rL-RVG into a drug candidate for the treatment of gastric carcinoma.
Collapse
Affiliation(s)
- Xuefeng Bu
- Department of General Surgery, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Yinghai Zhao
- Department of General Surgery, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang, Jiangsu, China
- Clinical Medicine College of Jiangsu UniversityZhenjiang, China
| | - Zhijian Zhang
- Clinical Medicine College of Jiangsu UniversityZhenjiang, China
| | - Mubin Wang
- Clinical Medicine College of Jiangsu UniversityZhenjiang, China
| | - Mi Li
- Clinical Medicine College of Jiangsu UniversityZhenjiang, China
| | - Yulan Yan
- Department of Internal Medicine, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang, China
| |
Collapse
|
147
|
Olagnier D, Amatore D, Castiello L, Ferrari M, Palermo E, Diamond MS, Palamara AT, Hiscott J. Dengue Virus Immunopathogenesis: Lessons Applicable to the Emergence of Zika Virus. J Mol Biol 2016; 428:3429-48. [PMID: 27130436 DOI: 10.1016/j.jmb.2016.04.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/14/2016] [Accepted: 04/16/2016] [Indexed: 01/07/2023]
Abstract
Dengue is the leading mosquito-transmitted viral infection in the world. There are more than 390 million new infections annually; while the majority of infected individuals are asymptomatic or develop a self-limited dengue fever, up to 1 million clinical cases develop severe manifestations, including dengue hemorrhagic fever and shock syndrome, resulting in ~25,000 deaths annually, mainly in children. Gaps in our understanding of the mechanisms that contribute to dengue infection and immunopathogenesis have hampered the development of vaccines and antiviral agents. Some of these limitations are highlighted by the explosive re-emergence of another arthropod-borne flavivirus-Zika virus-spread by the same vector, the Aedes aegypti mosquito, that also carries dengue, yellow fever and chikungunya viruses. This review will discuss the early virus-host interactions in dengue infection, with emphasis on the interrelationship between oxidative stress and innate immune pathways, and will provide insight as to how lessons learned from dengue research may expedite therapeutic strategies for Zika virus.
Collapse
Affiliation(s)
- David Olagnier
- Lady Davis Institute, Jewish General Hospital, McGill University Montreal, Canada
| | - Donatella Amatore
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Matteo Ferrari
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Enrico Palermo
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University at St. Louis, St. Louis, MO, USA
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy; Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy
| | - John Hiscott
- Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
148
|
Enterovirus 71 induces dsRNA/PKR-dependent cytoplasmic redistribution of GRP78/BiP to promote viral replication. Emerg Microbes Infect 2016; 5:e23. [PMID: 27004760 PMCID: PMC4820672 DOI: 10.1038/emi.2016.20] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 12/16/2022]
Abstract
GRP78/BiP is an endoplasmic reticulum (ER) chaperone protein with the important function of maintaining ER homeostasis, and the overexpression of GRP78/BiP alleviates ER stress. Our previous studies showed that infection with enterovirus 71 (EV71), a (+)RNA picornavirus, induced GRP78/BiP upregulation; however, ectopic GRP78/BiP overexpression in ER downregulates virus replication and viral particle formation. The fact that a virus infection increases GRP78/BiP expression, which is unfavorable for virus replication, is counterintuitive. In this study, we found that the GRP78/BiP protein level was elevated in the cytoplasm instead of in the ER in EV71-infected cells. Cells transfected with polyinosinic-polycytidylic acid, a synthetic analog of replicative double-stranded RNA (dsRNA), but not with viral proteins, also exhibited upregulation and elevation of GRP78/BiP in the cytosol. Our results further demonstrate that EV71 infections induce the dsRNA/protein kinase R-dependent cytosolic accumulation of GRP78/BiP. The overexpression of a GRP78/BiP mutant lacking a KDEL retention signal failed to inhibit both dithiothreitol-induced eIF2α phosphorylation and viral replication in the context of viral protein synthesis and viral titers. These data revealed that EV71 infection might cause upregulation and aberrant redistribution of GRP78/BiP to the cytosol, thereby facilitating virus replication.
Collapse
|
149
|
LIU KS, PENG ZH, CHENG WJ, DAI CF, TONG H. Endoplasmic reticulum stress-induced apoptosis in the development of reproduction. REPRODUCTION AND CONTRACEPTION 2016; 27:51-59. [PMID: 32288402 PMCID: PMC7129369 DOI: 10.7669/j.issn.1001-7844.2016.01.0051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Indexed: 12/21/2022]
Abstract
Proteins synthesized in the endoplasmic reticulum (ER) are properly folded with the assistance of ER chaperones. Accumulation of misfolded protein in the ER triggers an adaptive ER stress (ERS) response termed the unfolded protein response. Recent interest has focused on the possibility that the accumulation of misfolded proteins can also contribute to reproductive response, including preimplantation embryos, testicular germ cell, placenta, and unexplained intrauterine growth restriction (IUGR). The major ERS pathway constituents are present at all stages of preimplantation development and that the activation of ERS pathways can be induced at the 8-cell, morula and blastocyst stage. This review mainly introduced the research progress of ERS induced apoptosis of reproductive cells, providing a new direction for the research of reproductive disease therapy.
Collapse
Affiliation(s)
- Kang-sheng LIU
- Department of Clinical Laboratory, State Key Laboratory of Reproductive Medicine Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Zheng-hang PENG
- School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Weng-jun CHENG
- Department of Maternity and Child Health Care, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Chun-fan DAI
- Department of Obstetrical, Nanjing Maternity and Child Health Care Hospital, Nanjing 210009, China
| | - Hua TONG
- Department of Scientific Research and Education, Nanjing Maternity and Child Health Care Hospital, Nanjing 210009, China
| |
Collapse
|
150
|
Porcine Circovirus 2 Deploys PERK Pathway and GRP78 for Its Enhanced Replication in PK-15 Cells. Viruses 2016; 8:v8020056. [PMID: 26907328 PMCID: PMC4776210 DOI: 10.3390/v8020056] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/06/2016] [Accepted: 02/17/2016] [Indexed: 12/15/2022] Open
Abstract
Porcine circovirus type 2 (PCV2) infection induces autophagy and apoptosis. These cellular responses could be connected with endoplasmic reticulum (ER) stress. It remains unknown if PCV2 induces ER stress and if autophagy or apoptosis is primary to PCV2 infection or secondary responses following ER stress. Here, we demonstrate that PCV2 triggered unfolded protein response (UPR) in PK-15 cells by activating the PERK/eIF2α pathway without concomitant activation of IRE1 or ATF6. Since ATF4 and CHOP were induced later than PERK/eIF2α, it is clear that persistent PCV2 infection could lead to selective activation of PERK via the PERK-eIF2α-ATF4-CHOP axis. Therefore, PERK activation could be part of the pro-apoptotic signaling via induced expression of CHOP by PCV2. Since PERK inhibition by GSK2606414 or RNA silencing or suppression of eIF2α dephosphorylation by salubrinal limited viral replication, we suppose that PCV2 deploys UPR to enhance its replication. Over-expression of GRP78 or treatment with tauroursodeoxycholic acid could enhance viral capsid expression and/or viral titers, indicating that these chaperones, endogenous or exogenous, could help correct folding of viral proteins. Our findings provide the first evidence that ER stress plays a role in the pathogenesis of PCV2 infection probably as part of autophagic and apoptotic responses.
Collapse
|