201
|
Li Y, Huang J, Pang S, Wang H, Zhang A, Hawley RG, Yan B. Novel and functional ATG12 gene variants in sporadic Parkinson's disease. Neurosci Lett 2017; 643:22-26. [PMID: 28229934 DOI: 10.1016/j.neulet.2017.02.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/05/2023]
Abstract
Parkinson's disease (PD) is a common and progressive neurodegenerative disease, including familial and sporadic cases. To date, genetic causes for sporadic PD, majority of PD cases, remain largely unknown. Accumulating evidence indicates that dysfunctional autophagy, a highly conserved cellular process, is involved in the PD pathogenesis. We speculated that changed expression levels of autophagy-related genes (ATG) may contribute to PD development. Previously, we have genetically analyzed ATG5 and ATG7 genes in sporadic PD patients and identified several functional DNA sequence variants (DSVs). In groups of sporadic PD patients and ethic-matched healthy controls in this study, we further genetically and functionally analyzed the promoter of ATG12, a critical gene for autophagososme formation. The results showed that three DNA sequence variants (DSVs), g.115842507G>T,g.115842394C>T and g.115841817_18del, were identified three PD patients, which significantly altered transcriptional activity of ATG12 gene promoter, probably due to abolishing or creating binding sites for transcription factors. The transcriptional activity of ATG12 gene promoter was not significantly affected by other two DSVs identified in PD patients, g.115842640A>C and g.115842242G>C, which may not alter binding sites for transcription factors. Therefore, these three functional DSVs identified in PD patient may change ATG12 protein levels, contributing to PD development as a risk factor by interfering with autophagy as well as non-autophagy functions.
Collapse
Affiliation(s)
- Yuequn Li
- Division of Transcranial Doppler Ultrasound, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Jian Huang
- Shandong Provincial Sino-US Cooperation Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
| | - Shuchao Pang
- Shandong Provincial Sino-US Cooperation Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
| | - Haihua Wang
- Shandong Provincial Sino-US Cooperation Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China
| | - Aimei Zhang
- Division of Neurology, Affiliated Hospital of Jining Medical University, Jining Medical, University, Jining, Shandong 272029, China
| | - Robert G Hawley
- Shandong Provincial Sino-US Cooperation Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China; Department of Anatomy and Regenerative Biology, The George Washington University, 2300, Eye Street, NW, Washington, DC 20037, USA.
| | - Bo Yan
- Shandong Provincial Sino-US Cooperation Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272029, China; Department of Anatomy and Regenerative Biology, The George Washington University, 2300, Eye Street, NW, Washington, DC 20037, USA.
| |
Collapse
|
202
|
Liu J, Wang H, Gu J, Deng T, Yuan Z, Hu B, Xu Y, Yan Y, Zan J, Liao M, DiCaprio E, Li J, Su S, Zhou J. BECN1-dependent CASP2 incomplete autophagy induction by binding to rabies virus phosphoprotein. Autophagy 2017; 13:739-753. [PMID: 28129024 PMCID: PMC5388250 DOI: 10.1080/15548627.2017.1280220] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autophagy is an essential component of host immunity and used by viruses for survival. However, the autophagy signaling pathways involved in virus replication are poorly documented. Here, we observed that rabies virus (RABV) infection triggered intracellular autophagosome accumulation and results in incomplete autophagy by inhibiting autophagy flux. Subsequently, we found that RABV infection induced the reduction of CASP2/caspase 2 and the activation of AMP-activated protein kinase (AMPK)-AKT-MTOR (mechanistic target of rapamycin) and AMPK-MAPK (mitogen-activated protein kinase) pathways. Further investigation revealed that BECN1/Beclin 1 binding to viral phosphoprotein (P) induced an incomplete autophagy via activating the pathways CASP2-AMPK-AKT-MTOR and CASP2-AMPK-MAPK by decreasing CASP2. Taken together, our data first reveals a crosstalk of BECN1 and CASP2-dependent autophagy pathways by RABV infection.
Collapse
Affiliation(s)
- Juan Liu
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China
| | - Hailong Wang
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China
| | - Jinyan Gu
- b Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University , Nanjing , China
| | - Tingjuan Deng
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China
| | - Zhuangchuan Yuan
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China
| | - Boli Hu
- b Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University , Nanjing , China
| | - Yunbin Xu
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China
| | - Yan Yan
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China.,c Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University , Hangzhou , China
| | - Jie Zan
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China
| | - Min Liao
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China.,c Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University , Hangzhou , China
| | - Erin DiCaprio
- d Department of Veterinary Biosciences , College of Veterinary Medicine, Ohio State University , Columbus , OH , USA
| | - Jianrong Li
- d Department of Veterinary Biosciences , College of Veterinary Medicine, Ohio State University , Columbus , OH , USA
| | - Shuo Su
- b Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University , Nanjing , China
| | - Jiyong Zhou
- a Key Laboratory of Animal Virology of Ministry of Agriculture , Zhejiang University , Hangzhou , China.,c Collaborative Innovation Center and State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University , Hangzhou , China
| |
Collapse
|
203
|
Li X, Li Y, Fang S, Su J, Jiang J, Liang B, Huang J, Zhou B, Zang N, Ho W, Li J, Li Y, Chen H, Ye L, Liang H. Downregulation of autophagy-related gene ATG5 and GABARAP expression by IFN-λ1 contributes to its anti-HCV activity in human hepatoma cells. Antiviral Res 2017; 140:83-94. [PMID: 28131804 DOI: 10.1016/j.antiviral.2017.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Type-III interferon (IFN-λ), the most recently discovered family of IFNs, shares common features with type I IFNs, but also has many distinctive activities. It is not clear that whether IFN-λ has additional antiviral mechanisms. In this study, we investigated the effects of IFN-λ on autophagy, a cellular process closely related to hepatitis C virus (HCV) infection in human hepatoma Huh7 cells. Our results showed that IFN-λ1 treatment inhibit autophagic activity in Huh7 cells, as evidenced by the decreased expression of microtubule-associated protein 1 light chain 3B (LC3B)-II and conversion of LC3B-I to LC3B-II, decreased formation of GFP-LC3 puncta and accumulation of autophagosomes. IFN-λ1 could also inhibit HCV-induced or tunicamycin (a known inducer of autophagy with similar mechanism to HCV infection) -induced LC3B-II expression and autophagosome formation. Through PCR array, real time RT PCR, and western blot, two autophagy-related genes, ATG5 and GABARAP, were identified and verified to be down-regulated by IFN-λ1 treatment, either in HCV-uninfected Huh7 cells or in HCV JFH-1-infected cells. Overexpression of ATG5 and/or GABARAP could partly recover the IFN-λ1-inhibited HCV replication. Mechanism research demonstrated that IFN-λ1 could induce the expression of miR-181a and miR-214 (targeting ATG5 and GABARAP respectively), by which down-regulates ATG5 and GABARAP expression. Taken together, our results indicate that suppression of the autophagy response by IFN-λ1 contributes to IFN-λ1 anti-HCV activity. The results also provide a theoretical basis for improving the effectiveness of IFN treatment of HCV infection through inhibition of the HCV-induced autophagy response.
Collapse
Affiliation(s)
- Xu Li
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yu Li
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Medical Insurance Department, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi, China
| | - Shoucai Fang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jinming Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Division of HIV/AIDS Control and Prevention, Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning, 530021, Guangxi, China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Bingyu Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Bo Zhou
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ning Zang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenzhe Ho
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Jieliang Li
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, 19140, USA
| | - Yiping Li
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Geriatrics Digestion Department of Internal Medicine, The First Affiliated Hospital of GuangXi Medical University, Nanning, 530021, Guangxi, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
204
|
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: 8.0] [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
|
205
|
Liu Y, Olagnier D, Lin R. Host and Viral Modulation of RIG-I-Mediated Antiviral Immunity. Front Immunol 2017; 7:662. [PMID: 28096803 PMCID: PMC5206486 DOI: 10.3389/fimmu.2016.00662] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Innate immunity is the first line of defense against invading pathogens. Rapid and efficient detection of pathogen-associated molecular patterns via pattern-recognition receptors is essential for the host to mount defensive and protective responses. Retinoic acid-inducible gene-I (RIG-I) is critical in triggering antiviral and inflammatory responses for the control of viral replication in response to cytoplasmic virus-specific RNA structures. Upon viral RNA recognition, RIG-I recruits the mitochondrial adaptor protein mitochondrial antiviral signaling protein, which leads to a signaling cascade that coordinates the induction of type I interferons (IFNs), as well as a large variety of antiviral interferon-stimulated genes. The RIG-I activation is tightly regulated via various posttranslational modifications for the prevention of aberrant innate immune signaling. By contrast, viruses have evolved mechanisms of evasion, such as sequestrating viral structures from RIG-I detections and targeting receptor or signaling molecules for degradation. These virus–host interactions have broadened our understanding of viral pathogenesis and provided insights into the function of the RIG-I pathway. In this review, we summarize the recent advances regarding RIG-I pathogen recognition and signaling transduction, cell-intrinsic control of RIG-I activation, and the viral antagonism of RIG-I signaling.
Collapse
Affiliation(s)
- Yiliu Liu
- Jewish General Hospital, Lady Davis Institute, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - David Olagnier
- Jewish General Hospital, Lady Davis Institute, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Rongtuan Lin
- Jewish General Hospital, Lady Davis Institute, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| |
Collapse
|
206
|
Schott K, Riess M, König R. Role of Innate Genes in HIV Replication. Curr Top Microbiol Immunol 2017; 419:69-111. [PMID: 28685292 DOI: 10.1007/82_2017_29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cells use an elaborate innate immune surveillance and defense system against virus infections. Here, we discuss recent studies that reveal how HIV-1 is sensed by the innate immune system. Furthermore, we present mechanisms on the counteraction of HIV-1. We will provide an overview how HIV-1 actively utilizes host cellular factors to avoid sensing. Additionally, we will summarize effectors of the innate response that provide an antiviral cellular state. HIV-1 has evolved passive mechanism to avoid restriction and to regulate the innate response. We review in detail two prominent examples of these cellular factors: (i) NLRX1, a negative regulator of the innate response that HIV-1 actively usurps to block cytosolic innate sensing; (ii) SAMHD1, a restriction factor blocking the virus at the reverse transcription step that HIV-1 passively avoids to escape sensing.
Collapse
Affiliation(s)
- Kerstin Schott
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225, Langen, Germany
| | - Maximilian Riess
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225, Langen, Germany
| | - Renate König
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225, Langen, Germany. .,Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA. .,German Center for Infection Research (DZIF), 63225, Langen, Germany.
| |
Collapse
|
207
|
Weindel CG, Richey LJ, Mehta AJ, Shah M, Huber BT. Autophagy in Dendritic Cells and B Cells Is Critical for the Inflammatory State of TLR7-Mediated Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2016; 198:1081-1092. [PMID: 28031336 DOI: 10.4049/jimmunol.1601307] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022]
Abstract
Individuals suffering from autoimmune disorders possess a hyperactive cellular phenotype where tolerance to self-antigens is lost. Autophagy has been implicated in both the induction and prevention of autoimmunity, and modulators of this cellular recycling process hold high potential for the treatment of autoimmune diseases. In this study, we determine the effects of a loss of autophagy in dendritic cells (DCs), as well as both B cells and DCs, in a TLR7-mediated model of autoimmunity, similar to systemic lupus erythematosus, where both cell types are critical for disease. Although a loss of DC autophagy slowed disease, the combined loss of autophagy in both cell types resulted in a lethal sepsis-like environment, which included tissue inflammation and hyperproduction of inflammasome-associated cytokines. Ablation of B cell signaling reversed this phenotype, indicating that activation of these cells is an essential step in disease induction. Thus, autophagy plays a dichotomous role in this model of disease.
Collapse
Affiliation(s)
- Chi G Weindel
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Lauren J Richey
- Division of Laboratory Animal Medicine, Tufts University, Boston, MA 02111; and
| | - Abhiruchi J Mehta
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Mansi Shah
- Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Brigitte T Huber
- Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111; .,Department of Integrative Physiology and Pathobiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
208
|
Balasubramanian A, Teramoto T, Kulkarni AA, Bhattacharjee AK, Padmanabhan R. Antiviral activities of selected antimalarials against dengue virus type 2 and Zika virus. Antiviral Res 2016; 137:141-150. [PMID: 27889529 DOI: 10.1016/j.antiviral.2016.11.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/23/2022]
Abstract
In a previous study, twelve antimalarial compounds, amodiaquine (AQ) and derivatives, were shown to have potent anti-dengue viral (DENV) activity by using the stable DENV2 Renilla luciferase reporter replicon expressing BHK-21 cells, infectivity (plaque), and the qRT-PCR assays. In this study, we performed molecular modeling on these compounds to determine their stereo-electronic properties required for optimal antiviral activity. Based on the similarity of calculated stereo-electronic profiles, specifically the electrostatic potential profiles of the compounds, and in silico screening of related compounds from literature, we identified three additional compounds, Quinacrine (QC), Mefloquine (MQ), and GSK369796. Analysis of their antiviral activities indicated that all three compounds have high anti-DENV activity in the DENV2 replicon expressing cells with EC50 values of 5.30 ± 1.31 μM (QC), 3.22 ± 0.37 μM (MQ), and 5.06 ± 0.86 μM (GSK369796). The infectivity assays revealed the EC50 values of 7.09 ± 1.67 μM (QC), 4.36 ± 0.31 μM (MQ) and 3.03 ± 0.35 μM (GSK369796). The mode of action of these compounds is through inhibition of autophagy, thereby affecting DENV2 replication. Moreover, these compounds also showed antiviral activity against the rapidly emerging Zika virus (ZIKV) with EC50 values of 2.27 ± 0.14 μM (QC), 3.95 ± 0.21 μM (MQ), and 2.57 ± 0.09 μM (GSK369796).
Collapse
Affiliation(s)
- Anuradha Balasubramanian
- Department of Microbiology & Immunology, Georgetown University School of Medicine, Washington DC, USA
| | - Tadahisa Teramoto
- Department of Microbiology & Immunology, Georgetown University School of Medicine, Washington DC, USA
| | - Amol A Kulkarni
- Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington DC, USA
| | - Apurba K Bhattacharjee
- Department of Microbiology & Immunology, Georgetown University School of Medicine, Washington DC, USA.
| | - Radhakrishnan Padmanabhan
- Department of Microbiology & Immunology, Georgetown University School of Medicine, Washington DC, USA.
| |
Collapse
|
209
|
Autophagy regulates MAVS signaling activation in a phosphorylation-dependent manner in microglia. Cell Death Differ 2016; 24:276-287. [PMID: 28141795 DOI: 10.1038/cdd.2016.121] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/26/2016] [Accepted: 09/19/2016] [Indexed: 11/08/2022] Open
Abstract
Mitochondrial antiviral signaling (MAVS) protein has an important role in antiviral immunity and autoimmunity. However, the pathophysiological role of this signaling pathway, especially in the brain, remains elusive. Here we demonstrated that MAVS signaling existed and mediated poly(I:C)-induced inflammation in the brain. Along with the MAVS signaling activation, there was an induction of autophagic activation. Autophagy negatively regulated the activity of MAVS through direct binding of LC3 to the LIR motif Y(9)xxI(12) of MAVS. We also found that c-Abl kinase phosphorylated MAVS and regulated its interaction with LC3. Interestingly, tyrosine phosphorylation of MAVS was required for downstream signaling activation. Importantly, in vivo data showed that the deficiency of MAVS or c-Abl prevented MPTP-induced microglial activation and dopaminergic neuron loss. Together, our findings reveal the molecular mechanisms underlying the regulation of MAVS-dependent microglial activation in the nervous system, thus providing a potential target for the treatment of microglia-driven inflammatory brain diseases.
Collapse
|
210
|
Liu X, Qin H, Xu J. The role of autophagy in the pathogenesis of systemic lupus erythematosus. Int Immunopharmacol 2016; 40:351-361. [DOI: 10.1016/j.intimp.2016.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 08/15/2016] [Accepted: 09/19/2016] [Indexed: 02/07/2023]
|
211
|
Abstract
Autophagy has broad functions in immunity, ranging from cell-autonomous defence to coordination of complex multicellular immune responses. The successful resolution of infection and avoidance of autoimmunity necessitates efficient and timely communication between autophagy and pathways that sense the immune environment. The recent literature indicates that a variety of immune mediators induce or repress autophagy. It is also becoming increasingly clear that immune signalling cascades are subject to regulation by autophagy, and that a return to homeostasis following a robust immune response is critically dependent on this pathway. Importantly, examples of non-canonical forms of autophagy in mediating immunity are pervasive. In this article, the progress in elucidating mechanisms of crosstalk between autophagy and inflammatory signalling cascades is reviewed. Improved mechanistic understanding of the autophagy machinery offers hope for treating infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Ken Cadwell
- grid.137628.90000 0004 1936 8753and the Department of Microbiology, Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, 10016 New York USA
| |
Collapse
|
212
|
Moretti J, Blander JM. Cell-autonomous stress responses in innate immunity. J Leukoc Biol 2016; 101:77-86. [PMID: 27733577 DOI: 10.1189/jlb.2mr0416-201r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/10/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
The innate immune response of phagocytes to microbes has long been known to depend on the core signaling cascades downstream of pattern recognition receptors (PRRs), which lead to expression and production of inflammatory cytokines that counteract infection and induce adaptive immunity. Cell-autonomous responses have recently emerged as important mechanisms of innate immunity. Either IFN-inducible or constitutive, these processes aim to guarantee cell homeostasis but have also been shown to modulate innate immune response to microbes and production of inflammatory cytokines. Among these constitutive cell-autonomous responses, autophagy is prominent and its role in innate immunity has been well characterized. Other stress responses, such as metabolic stress, the ER stress/unfolded protein response, mitochondrial stress, or the DNA damage response, seem to also be involved in innate immunity, although the precise mechanisms by which they regulate the innate immune response are not yet defined. Of importance, these distinct constitutive cell-autonomous responses appear to be interconnected and can also be modulated by microbes and PRRs, which add further complexity to the interplay between innate immune signaling and cell-autonomous responses in the mediation of an efficient innate immune response.
Collapse
Affiliation(s)
- Julien Moretti
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - J Magarian Blander
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; .,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
213
|
Abstract
Autophagy is a catabolic process by which cells remove protein aggregates and damaged organelles for recycling. It can also be used by cells to remove intracellular microbial pathogens, including viruses, in a process known as xenophagy. However, many viruses have developed mechanisms to subvert this intracellular antiviral response and even use this pathway to support their own replications. Hepatitis C virus (HCV) is one such virus and is an important human pathogen that can cause severe liver diseases. Recent studies indicated that HCV could activate the autophagic pathway to support its replication. This review summarizes the current knowledge on the interplay between HCV and autophagy and how this interplay affects HCV replication and host innate immune responses.
Collapse
|
214
|
Abstract
Mitochondrial antiviral signalling protein (MAVS) acts as a critical adaptor protein to transduce antiviral signalling by physically interacting with activated RIG-I and MDA5 receptors. MAVS executes its functions at the outer membrane of mitochondria to regulate downstream antiviral signalling, indicating that the mitochondria provides a functional platform for innate antiviral signalling transduction. However, little is known about whether and how MAVS-mediated antiviral signalling contributes to mitochondrial homeostasis. Here we show that the activation of MAVS is sufficient to induce autophagic signalling, which may mediate the turnover of the damaged mitochondria. Importantly, we find MAVS directly interacts with LC3 through its LC3-binding motif ‘YxxI’, suggesting that MAVS might act as an autophagy receptor to mediate mitochondrial turnover upon excessive activation of RLR signalling. Furthermore, we provide evidence that both MAVS self-aggregation and its interaction with TRAF2/6 proteins are important for MAVS-mediated mitochondrial turnover. Collectively, our findings suggest that MAVS acts as a potential receptor for mitochondria-associated autophagic signalling to maintain mitochondrial homeostasis.
Collapse
|
215
|
Wang S, Yang YK, Chen T, Zhang H, Yang WW, Song SS, Zhai ZH, Chen DY. RNF123 has an E3 ligase-independent function in RIG-I-like receptor-mediated antiviral signaling. EMBO Rep 2016; 17:1155-68. [PMID: 27312109 PMCID: PMC4967948 DOI: 10.15252/embr.201541703] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/09/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are cytoplasmic sensors crucial for recognizing different species of viral RNAs, which triggers the production of type I interferons (IFNs) and inflammatory cytokines. Here, we identify RING finger protein 123 (RNF123) as a negative regulator of RIG-I and MDA5. Overexpression of RNF123 inhibits IFN-β production triggered by Sendai virus (SeV) and encephalomyocarditis picornavirus (EMCV). Knockdown or knockout of endogenous RNF123 potentiates IFN-β production triggered by SeV and EMCV, but not by the sensor of DNA viruses cGAS RNF123 associates with RIG-I and MDA5 in both endogenous and exogenous cases in a viral infection-inducible manner. The SPRY and coiled-coil, but not the RING, domains of RNF123 are required for the inhibitory function. RNF123 interacts with the N-terminal CARD domains of RIG-I/MDA5 and competes with the downstream adaptor VISA/MAVS/IPS-1/Cardif for RIG-I/MDA5 CARD binding. These findings suggest that RNF123 functions as a novel inhibitor of innate antiviral signaling mediated by RIG-I and MDA5, a function that does not depend on its E3 ligase activity.
Collapse
Affiliation(s)
- Shuai Wang
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Yong-Kang Yang
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Tao Chen
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Heng Zhang
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Wei-Wei Yang
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Sheng-Sheng Song
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Zhong-He Zhai
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| | - Dan-Ying Chen
- Key Laboratory of Cell Proliferation and Differentiation of The Ministry of Education, School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
216
|
Liu KY, Xia YQ, Zhou J, Chen ZW, Lu D, Zhang NZ, Liu XS, Ai H, Zhou LL. MOLECULAR CHARACTERIZATION OF AUTOPHAGY-RELATED GENE 5 FROM Spodoptera exigua AND EXPRESSION ANALYSIS UNDER VARIOUS STRESS CONDITIONS. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2016; 92:225-241. [PMID: 27226059 DOI: 10.1002/arch.21339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 04/11/2016] [Accepted: 04/28/2016] [Indexed: 06/05/2023]
Abstract
Autophagy is not only involved in development, but also has been proved to attend immune response against invading pathogens. Autophagy protein 5 (ATG5) is an important autophagic protein, which plays a crucial role in autophagosome elongation. Although ATG5 has been well studied in mammal, yeast, and Drosophila, little is known about ATG5 in lepidopteran insects. We cloned putative SeAtg5 gene from Spodoptera exigua larvae by the rapid amplification of cDNA ends method, and its characteristics and the influences of multiple exogenous factors on its expression levels were then investigated. The results showed that the putative S. exigua SeATG5 protein is highly homologous to other insect ATG5 proteins, which has a conserved Pfm domain and multiple phosphorylation sites. Next, fluorescence microscope observation showed that mCherry-SeATG5 was distributed in both nucleus and cytoplasm of Spodoptera litura Sl-HP cells and partially co-localized with BmATG6-GFP, but it almost has no significant co-localization with GFP-HaATG8. Then, the Western blot analysis demonstrated that GFP-SeATG5 conjugated with ATG12. Moreover, real-time PCR revealed that its expression levels significantly increased at the initiation of pupation and the stage of adult. In addition, the expression levels of SeAtg5 can be enhanced by the starvation, UV radiation, and infection of baculovirus and bacterium. However, the expression levels of SeAtg5 decreased at 24 h post treatments in all these treatments except in starvation. These results suggested that SeATG5 might be involved in response of S. exigua under various stress conditions.
Collapse
Affiliation(s)
- Kai-Yu Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Yu-Qian Xia
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Jing Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Zu-Wen Chen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Dandan Lu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Ning-Zhao Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Xu-Sheng Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Hui Ai
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, China
| | - Li-Lin Zhou
- Department of Plant Protection, Wuhan Vegetable Research Institute, Wuhan, China
| |
Collapse
|
217
|
Pei J, Deng J, Ye Z, Wang J, Gou H, Liu W, Zhao M, Liao M, Yi L, Chen J. Absence of autophagy promotes apoptosis by modulating the ROS-dependent RLR signaling pathway in classical swine fever virus-infected cells. Autophagy 2016; 12:1738-1758. [PMID: 27463126 DOI: 10.1080/15548627.2016.1196318] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A growing number of studies have demonstrated that both macroautophagy/autophagy and apoptosis are important inner mechanisms of cell to maintain homeostasis and participate in the host response to pathogens. We have previously reported that a functional autophagy pathway is trigged by infection of classical swine fever virus (CSFV) and is required for viral replication and release in host cells. However, the interplay of autophagy and apoptosis in CSFV-infected cells has not been clarified. In the present study, we demonstrated that autophagy induction with rapamycin facilitates cellular proliferation after CSFV infection, and that autophagy inhibition by knockdown of essential autophagic proteins BECN1/Beclin 1 or MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) promotes apoptosis via fully activating both intrinsic and extrinsic mechanisms in CSFV-infected cells. We also found that RIG-I-like receptor (RLR) signaling was amplified in autophagy-deficient cells during CSFV infection, which was closely linked to the activation of the intrinsic apoptosis pathway. Moreover, we discovered that virus infection of autophagy-impaired cells results in an increase in copy number of mitochondrial DNA and in the production of reactive oxygen species (ROS), which plays a significant role in enhanced RLR signaling and the activated extrinsic apoptosis pathway in cultured cells. Collectively, these data indicate that CSFV-induced autophagy delays apoptosis by downregulating ROS-dependent RLR signaling and thus contributes to virus persistent infection in host cells.
Collapse
Affiliation(s)
- Jingjing Pei
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Jieru Deng
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Zuodong Ye
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Jiaying Wang
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Hongchao Gou
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Wenjun Liu
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Mingqiu Zhao
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Ming Liao
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Lin Yi
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| | - Jinding Chen
- a College of Veterinary Medicine , South China Agricultural University , Guangzhou , China
| |
Collapse
|
218
|
Vij A, Randhawa R, Parkash J, Changotra H. Investigating regulatory signatures of human autophagy related gene 5 (ATG5) through functional in silico analysis. Meta Gene 2016; 9:237-48. [PMID: 27617225 PMCID: PMC5006144 DOI: 10.1016/j.mgene.2016.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an essential, homeostatic process which removes damaged cellular proteins and organelles for cellular renewal. ATG5, a part of E3 ubiquitin ligase-like complex (Atg12-Atg5/Atg16L1), is a key regulator involved in autophagosome formation - a crucial phase of autophagy. In this study, we used different in silico methods for comprehensive analysis of ATG5 to investigate its less explored regulatory activity. We have predicted various physico-chemical parameters and two possible transmembrane models that helped in exposing its functional regions. Twenty four PTM sites and 44 TFBS were identified which could be targeted to modulate the autophagy pathway. Furthermore, LD analysis identified 3 blocks of genotyped SNPs and 2 deleterious nsSNPs that may have damaging impact on protein function and thus could be employed for carrying genome-wide association studies. In conclusion, the information obtained in this study could be helpful for better understanding of regulatory roles of ATG5 and provides a base for its implication in population-based studies. ATG5 phylogenetic analysis shows its evolutionary relationship with other species. Two possible models for transmembrane regions detected in ATG5. 24 Post-translational modification sites were annotated over ATG5 domain structure. 44 Transcription factor binding sites were identified in ATG5. 2 nsSNPs were predicted to have damaging impact on ATG5 protein function.
Collapse
Affiliation(s)
- Avni Vij
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, 173 234, Himachal Pradesh, India
| | - Rohit Randhawa
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, 173 234, Himachal Pradesh, India
| | - Jyoti Parkash
- Centre for Animal Sciences, School of Basic and Applied Sciences, Central University Punjab, Mansa Road, Bathinda 151 001, India
| | - Harish Changotra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, 173 234, Himachal Pradesh, India
| |
Collapse
|
219
|
Han J, Xu G, Xu T. The miiuy croaker microRNA transcriptome and microRNA regulation of RIG-I like receptor signaling pathway after poly(I:C) stimulation. FISH & SHELLFISH IMMUNOLOGY 2016; 54:419-426. [PMID: 27131903 DOI: 10.1016/j.fsi.2016.04.126] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/18/2016] [Accepted: 04/26/2016] [Indexed: 06/05/2023]
Abstract
MicroRNAs (miRNAs) as endogenous small non-coding RNAs play key regulatory roles in diverse biological processes via degrading the target mRNAs or inhibiting protein translation. Previously many researchers have reported the identification, characteristic of miRNAs and the interaction with its target gene. But, the study on the regulation of miRNAs to biological processes via regulatory the key signaling pathway was still limited. In order to comprehend the regulatory mechanism of miRNAs, two small RNA libraries from the spleen of miiuy croaker individuals with or without poly(I:C) infection were constructed. The 197 conserved miRNAs and 75 novel miRNAs were identified, and 14 conserved and 8 novel miRNAs appeared significant variations. Those differently expressed miRNAs relate to immune regulation of miiuy croaker. Furthermore, expressions of four differently expressed miRNAs were validated by qRT-PCR, and the result was consistent with sequencing data. The target genes of the differently expressed miRNAs in the two libraries were predicted, and some candidate target genes were involved in the RIG-I-like receptor (RLR) signaling pathway. The negative regulation of miRNAs to target genes were confirmed by comparing the expression pattern of miRNAs and their target genes. The results of regulating target genes were that firstly directly or indirectly activating the downstream signaling cascades and subsequent inducting the type I interferon, inflammatory cytokines and apoptosis. These studies could help us to deeper understand the roles of miRNAs played in the fish immune system, and provide a new way to investigate the defense mechanism of fish.
Collapse
Affiliation(s)
- Jingjing Han
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Guoliang Xu
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Tianjun Xu
- Laboratory of Fish Biogenetics & Immune Evolution, College of Marine Science, Zhejiang Ocean University, Zhoushan, 316022, China.
| |
Collapse
|
220
|
Nucleic acid sensing and innate immunity: signaling pathways controlling viral pathogenesis and autoimmunity. CURRENT CLINICAL MICROBIOLOGY REPORTS 2016; 3:132-141. [PMID: 27857881 DOI: 10.1007/s40588-016-0043-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Innate immunity refers to the body's initial response to curb infection upon exposure to invading organisms. While the detection of pathogen-associated molecules is an ancient form of host defense, if dysfunctional, autoimmune disease may result. The innate immune response during pathogenic infection is initiated through the activation of receptors recognizing conserved molecular patterns, such as nucleic acids from a virus' genome or replicative cycle. Additionally, the host's own nucleic acids are capable of activating an immune response. Therefore, it follows that the nucleic acid-sensing pathways must be tightly controlled to avoid an autoimmune response from recognition of self, yet still be unimpeded to respond to viral infections. In this review, we will describe the nucleic acid sensing pathways and how they respond to virus infection. Moreover, we will discuss autoimmune diseases that develop when these pathways fail to signal properly and identify knowledge gaps that are prime for interrogation.
Collapse
|
221
|
Wu TT, Li WM, Yao YM. Interactions between Autophagy and Inhibitory Cytokines. Int J Biol Sci 2016; 12:884-97. [PMID: 27313501 PMCID: PMC4910606 DOI: 10.7150/ijbs.15194] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/12/2016] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a degradative pathway that plays an essential role in maintaining cellular homeostasis. Most early studies of autophagy focused on its involvement in age-associated degeneration and nutrient deprivation. However, the immunological functions of autophagy have become more widely studied in recent years. Autophagy has been shown to be an intrinsic cellular defense mechanism in the innate and adaptive immune responses. Cytokines belong to a broad and loose category of proteins and are crucial for innate and adaptive immunity. Inhibitory cytokines have evolved to permit tolerance to self while also contributing to the eradication of invading pathogens. Interactions between inhibitory cytokines and autophagy have recently been reported, revealing a novel mechanism by which autophagy controls the immune response. In this review, we discuss interactions between autophagy and the regulatory cytokines IL-10, transforming growth factor-β, and IL-27. We also mention possible interactions between two newly discovered cytokines, IL-35 and IL-37, and autophagy.
Collapse
Affiliation(s)
- Tian-Tian Wu
- 1. Department of Hepatobiliary Surgery, the 309th Hospital of Chinese PLA, Beijing 100091, People's Republic of China
| | - Wei-Min Li
- 1. Department of Hepatobiliary Surgery, the 309th Hospital of Chinese PLA, Beijing 100091, People's Republic of China
| | - Yong-Ming Yao
- 2. Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China; 3. State Key Laboratory of Kidney Disease, the Chinese PLA General Hospital, Beijing 100853, People's Republic of China
| |
Collapse
|
222
|
Tsuchiya Y, Jounai N, Takeshita F, Ishii KJ, Mizuguchi K. Ligand-induced Ordering of the C-terminal Tail Primes STING for Phosphorylation by TBK1. EBioMedicine 2016; 9:87-96. [PMID: 27333035 PMCID: PMC4972534 DOI: 10.1016/j.ebiom.2016.05.039] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/27/2016] [Accepted: 05/29/2016] [Indexed: 12/13/2022] Open
Abstract
The innate immune protein Stimulator of interferon genes (STING) promotes the induction of interferon beta (IFN-β) production via the phosphorylation of its C-terminal tail (CTT) by TANK-binding kinase 1 (TBK1). Potent ligands of STING are, therefore, promising candidates for novel anti-cancer drugs or vaccine adjuvants. However, the intrinsically flexible CTT poses serious problems in in silico drug discovery. Here, we performed molecular dynamics simulations of the STING fragment containing the CTT in ligand-bound and unbound forms and observed that the binding of a potent ligand cyclic GMP-AMP (cGAMP) induced a local structure in the CTT, reminiscent of the known structure of a TBK1 substrate. The subsequent molecular biological experiments confirmed the observed dynamics of the CTT and identified essential residues for the activation of the IFN-β promoter, leading us to propose a new mechanism of STING activation. The binding of the potent ligand cGAMP to STING induces local structural ordering in the flexible C-terminal tail of STING. Site-directed mutagenesis studies, designed based on this observation, elucidated residues essential for STING function. The local structure formation in the CTT was shown to be essential for the induction of IFN-β production.
Collapse
Affiliation(s)
- Yuko Tsuchiya
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan.
| | - Nao Jounai
- Laboratory of Aduvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Fumihiko Takeshita
- Laboratory of Aduvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Ken J Ishii
- Laboratory of Aduvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan.
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan.
| |
Collapse
|
223
|
Saitoh T, Akira S. Regulation of inflammasomes by autophagy. J Allergy Clin Immunol 2016; 138:28-36. [PMID: 27373323 DOI: 10.1016/j.jaci.2016.05.009] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022]
Abstract
Inflammasomes detect pathogen-associated molecular patterns to induce inflammatory innate immune responses and play a key role in host defense against infectious agents. However, inflammasomes are often wrongly activated by metabolites, amyloids, and environmental irritants. This induces massive inflammation, causing severe tissue damage, and results in the development of inflammatory diseases. Hence cellular machineries regulating both "activation" and "inactivation" of inflammasomes are definitely important. Recent studies have shown that autophagy, an intracellular degradation system associated with maintenance of cellular homeostasis, plays a key role in inflammasome inactivation. Notably, autophagy deficiency caused by gene mutation disrupts organelle elimination and thus induces aberrant activation of inflammasomes, leading to severe tissue damage. Here we review recent findings regarding the involvement of autophagy in the regulation of inflammasome activation and development of inflammatory disorders.
Collapse
Affiliation(s)
- Tatsuya Saitoh
- Department of Inflammation Biology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Research Center Immunology Frontier Research Center, Osaka, Japan; Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
224
|
Pabon MA, Ma KC, Choi AMK. Autophagy and Obesity-Related Lung Disease. Am J Respir Cell Mol Biol 2016; 54:636-46. [PMID: 26900794 PMCID: PMC5455357 DOI: 10.1165/rcmb.2016-0045ps] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/22/2016] [Indexed: 12/11/2022] Open
Abstract
Obesity-related disease is a significant source of premature death and economic burden globally. It is also a common comorbidity in patients suffering from lung disease, affecting both severity and treatment success. However, this complex association between obesity and the lung is poorly understood. Autophagy is a self-recycling homeostatic process that has been linked to beneficial or deleterious effects, depending on the specific lung disease. Obesity affects autophagy in a tissue-specific manner, activating autophagy in adipocytes and impairing autophagy in hepatocytes, immune cells, and pancreatic β-cells, among others. Obesity is also characterized by chronic low-grade inflammation that can be modulated by the pro- and antiinflammatory effects of the autophagic machinery. Scant evidence exists regarding the impact of autophagy in obesity-related lung diseases, but there are communal pathways that could be related to disease pathogenesis. Important signaling molecules in obesity, including IL-17, leptin, adiponectin, NLRP3 inflammasome, and TLR-4, have been implicated in the pathogenesis of lung disease. These mediators are known to be modulated by autophagy activity. In this perspective, we highlight the recent advances in the understanding of autophagy in obesity-related conditions, as well as the potential mechanisms that can link autophagy and obesity in the pathogenesis of lung disease.
Collapse
Affiliation(s)
- Maria A Pabon
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Kevin C Ma
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
225
|
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: 2.4] [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
|
226
|
Jin S, Tian S, Chen Y, Zhang C, Xie W, Xia X, Cui J, Wang RF. USP19 modulates autophagy and antiviral immune responses by deubiquitinating Beclin-1. EMBO J 2016; 35:866-80. [PMID: 26988033 PMCID: PMC4972138 DOI: 10.15252/embj.201593596] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
Autophagy, mediated by a number of autophagy-related (ATG) proteins, plays an important role in the bulk degradation of cellular constituents. Beclin-1 (also known as Atg6 in yeast) is a core protein essential for autophagic initiation and other biological processes. The activity of Beclin-1 is tightly regulated by multiple post-translational modifications, including ubiquitination, yet the molecular mechanism underpinning its reversible deubiquitination remains poorly defined. Here, we identified ubiquitin-specific protease 19 (USP19) as a positive regulator of autophagy, but a negative regulator of type I interferon (IFN) signaling.USP19 stabilizes Beclin-1 by removing the K11-linked ubiquitin chains of Beclin-1 at lysine 437. Moreover, we foundthat USP19 negatively regulates type IIFNsignaling pathway, by blockingRIG-I-MAVSinteraction in a Beclin-1-dependent manner. Depletion of eitherUSP19 or Beclin-1 inhibits autophagic flux and promotes type IIFNsignaling as well as cellular antiviral immunity. Our findings reveal novel dual functions of theUSP19-Beclin-1 axis by balancing autophagy and the production of type IIFNs.
Collapse
Affiliation(s)
- Shouheng Jin
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuo Tian
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yamei Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Chuanxia Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weihong Xie
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaojun Xia
- Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China Department of Experimental Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China
| | - Jun Cui
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Rong-Fu Wang
- Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
227
|
Shibasaki S, Karasaki M, Aburaya S, Morisaka H, Takeda Y, Aoki W, Kitano S, Kitano M, Ueda M, Sano H, Iwasaki T. A comparative proteomics study of a synovial cell line stimulated with TNF-α. FEBS Open Bio 2016; 6:418-24. [PMID: 27419047 PMCID: PMC4856420 DOI: 10.1002/2211-5463.12049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/18/2016] [Accepted: 02/29/2016] [Indexed: 12/29/2022] Open
Abstract
To elucidate the pathogenesis of rheumatoid arthritis (RA), we used proteomic analysis to determine the protein profile in a synovial cell line, MH7A, established from patients with RA. Proteins were extracted from MH7A cells that were or were not stimulated with tumor necrosis factor‐α (TNF‐α), and then analyzed on a liquid chromatography/mass spectrometry system equipped with a unique long monolithic silica capillary. On the basis of the results of this proteomic analysis, we identified 2650 proteins from untreated MH7A cells and 2688 proteins from MH7A cells stimulated with TNF‐α. Next, we selected 269 differentially produced proteins that were detected only under TNF‐α stimulation, and classified these proteins by performing gene ontology analysis by using DAVID as a functional annotation tool. In TNF‐α‐stimulated MH7A cells, we observed substantial production of plasminogen‐activator inhibitor 2 and apoptosis‐regulating proteins such as BH3‐interacting domain death agonist, autophagy protein 5, apolipoprotein E, and caspase‐3. These results indicate that the upregulation of plasminogen‐activator inhibitor 2 and apoptosis‐regulating proteins in synovial cells in response to TNF‐α stimulation might represent a predominant factor that contributes to the pathogenesis of RA.
Collapse
Affiliation(s)
- Seiji Shibasaki
- General Education Center Hyogo University of Health Sciences Kobe Japan
| | - Miki Karasaki
- General Education Center Hyogo University of Health Sciences Kobe Japan
| | - Shunsuke Aburaya
- Division of Applied Life Science Graduate School of Agriculture Kyoto University Japan
| | - Hironobu Morisaka
- Division of Applied Life Science Graduate School of Agriculture Kyoto University Japan
| | - Yumiko Takeda
- Division of Applied Life Science Graduate School of Agriculture Kyoto University Japan
| | - Wataru Aoki
- Division of Applied Life Science Graduate School of Agriculture Kyoto University Japan
| | - Sachie Kitano
- Division of Rheumatology Department of Internal Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Masayasu Kitano
- Division of Rheumatology Department of Internal Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Mitsuyoshi Ueda
- Division of Applied Life Science Graduate School of Agriculture Kyoto University Japan
| | - Hajime Sano
- Division of Rheumatology Department of Internal Medicine Hyogo College of Medicine Nishinomiya Japan
| | - Tsuyoshi Iwasaki
- Division of Pharmacotherapy Department of Pharmacy School of Pharmacy Hyogo University of Health Sciences Kobe Japan
| |
Collapse
|
228
|
Nguyen NT, Now H, Kim WJ, Kim N, Yoo JY. Ubiquitin-like modifier FAT10 attenuates RIG-I mediated antiviral signaling by segregating activated RIG-I from its signaling platform. Sci Rep 2016; 6:23377. [PMID: 26996158 PMCID: PMC4800306 DOI: 10.1038/srep23377] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/04/2016] [Indexed: 12/30/2022] Open
Abstract
RIG-I is a key cytosolic RNA sensor that mediates innate immune defense against RNA virus. Aberrant RIG-I activity leads to severe pathological states such as autosomal dominant multi-system disorder, inflammatory myophathies and dermatomyositis. Therefore, identification of regulators that ensure efficient defense without harmful immune-pathology is particularly critical to deal with RIG-I-associated diseases. Here, we presented the inflammatory inducible FAT10 as a novel negative regulator of RIG-I-mediated inflammatory response. In various cell lines, FAT10 protein is undetectable unless it is induced by pro-inflammatory cytokines. FAT10 non-covalently associated with the 2CARD domain of RIG-I, and inhibited viral RNA-induced IRF3 and NF-kB activation through modulating the RIG-I protein solubility. We further demonstrated that FAT10 was recruited to RIG-I-TRIM25 to form an inhibitory complex where FAT10 was stabilized by E3 ligase TRIM25. As the result, FAT10 inhibited the antiviral stress granules formation contains RIG-I and sequestered the active RIG-I away from the mitochondria. Our study presented a novel mechanism to dampen RIG-I activity. Highly accumulated FAT10 is observed in various cancers with pro-inflammatory environment, therefore, our finding which uncovered the suppressive effect of the accumulated FAT10 during virus-mediated inflammatory response may also provide molecular clue to understand the carcinogenesis related with infection and inflammation.
Collapse
Affiliation(s)
- Nhung T.H. Nguyen
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea Hyoja-dong 31, Pohang, 790-784, Republic of Korea
| | - Hesung Now
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea Hyoja-dong 31, Pohang, 790-784, Republic of Korea
| | - Woo-Jong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea Hyoja-dong 31, Pohang, 790-784, Republic of Korea
| | - Nari Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea Hyoja-dong 31, Pohang, 790-784, Republic of Korea
| | - Joo-Yeon Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea Hyoja-dong 31, Pohang, 790-784, Republic of Korea
| |
Collapse
|
229
|
Paul P, Münz C. Autophagy and Mammalian Viruses: Roles in Immune Response, Viral Replication, and Beyond. Adv Virus Res 2016; 95:149-95. [PMID: 27112282 DOI: 10.1016/bs.aivir.2016.02.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is an important cellular catabolic process conserved from yeast to man. Double-membrane vesicles deliver their cargo to the lysosome for degradation. Hence, autophagy is one of the key mechanisms mammalian cells deploy to rid themselves of intracellular pathogens including viruses. However, autophagy serves many more functions during viral infection. First, it regulates the immune response through selective degradation of immune components, thus preventing possibly harmful overactivation and inflammation. Additionally, it delivers virus-derived antigens to antigen-loading compartments for presentation to T lymphocytes. Second, it might take an active part in the viral life cycle by, eg, facilitating its release from cells. Lastly, in the constant arms race between host and virus, autophagy is often hijacked by viruses and manipulated to their own advantage. In this review, we will highlight key steps during viral infection in which autophagy plays a role. We have selected some exemplary viruses and will describe the molecular mechanisms behind their intricate relationship with the autophagic machinery, a result of host-pathogen coevolution.
Collapse
Affiliation(s)
- P Paul
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - C Münz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
230
|
Grimm WA, Messer JS, Murphy SF, Nero T, Lodolce JP, Weber CR, Logsdon MF, Bartulis S, Sylvester BE, Springer A, Dougherty U, Niewold TB, Kupfer SS, Ellis N, Huo D, Bissonnette M, Boone DL. The Thr300Ala variant in ATG16L1 is associated with improved survival in human colorectal cancer and enhanced production of type I interferon. Gut 2016; 65:456-64. [PMID: 25645662 PMCID: PMC4789828 DOI: 10.1136/gutjnl-2014-308735] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/11/2014] [Indexed: 12/15/2022]
Abstract
OBJECTIVE ATG16L1 is an autophagy gene known to control host immune responses to viruses and bacteria. Recently, a non-synonymous single-nucleotide polymorphism in ATG16L1 (Thr300Ala), previously identified as a risk factor in Crohn's disease (CD), was associated with more favourable clinical outcomes in thyroid cancer. Mechanisms underlying this observation have not been proposed, nor is it clear whether an association between Thr300Ala and clinical outcomes will be observed in other cancers. We hypothesised that Thr300Ala influences clinical outcome in human colorectal cancer (CRC) and controls innate antiviral pathways in colon cancer cells. DESIGN We genotyped 460 patients with CRC and assessed for an association between ATG16L1 Thr300Ala and overall survival and clinical stage. Human CRC cell lines were targeted by homologous recombination to examine the functional consequence of loss of ATG16L1, or introduction of the Thr300Ala variant. RESULTS We found an association between longer overall survival, reduced metastasis and the ATG16L1 Ala/Ala genotype. Tumour sections from ATG16L1 Ala/Ala patients expressed elevated type I interferons (IFN-I)-inducible, MxA, suggesting that differences in cytokine production may influence disease progression. When introduced into human CRC cells by homologous recombination, the Thr300Ala variant did not affect bulk autophagy, but increased basal production of type I IFN. Introduction of Thr300Ala resulted in increased sensitivity to the dsRNA mimic poly(I:C) through a mitochondrial antiviral signalling (MAVS)-dependent pathway. CONCLUSIONS The CD-risk allele, Thr300Ala, in ATG16L1 is associated with improved overall survival in human CRC, generating a rationale to genotype ATG16L1 Thr300Ala in patients with CRC. We found that Thr300A alters production of MAVS-dependent type I IFN in CRC cells, providing a mechanism that may influence clinical outcomes.
Collapse
Affiliation(s)
- Wesley A Grimm
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Jeannette S Messer
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Stephen F Murphy
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Thomas Nero
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - James P Lodolce
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | - Mark F Logsdon
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Sarah Bartulis
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Brooke E Sylvester
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Amanda Springer
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Urszula Dougherty
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Timothy B Niewold
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Sonia S Kupfer
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Nathan Ellis
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Dezheng Huo
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Marc Bissonnette
- Departments of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - David L Boone
- Departments of Microbiology and Immunology, Indiana University School of Medicine—South Bend, South Bend, Indiana, USA
| |
Collapse
|
231
|
Huang X, Yue Y, Li D, Zhao Y, Qiu L, Chen J, Pan Y, Xi J, Wang X, Sun Q, Li Q. Antibody-dependent enhancement of dengue virus infection inhibits RLR-mediated Type-I IFN-independent signalling through upregulation of cellular autophagy. Sci Rep 2016; 6:22303. [PMID: 26923481 PMCID: PMC4770412 DOI: 10.1038/srep22303] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/11/2016] [Indexed: 12/25/2022] Open
Abstract
Antibody dependent enhancement (ADE) of dengue virus (DENV) infection is identified as the main risk factor of severe Dengue diseases. Through opsonization by subneutralizing or non-neutralizing antibodies, DENV infection suppresses innate cell immunity to facilitate viral replication. However, it is largely unknown whether suppression of type-I IFN is necessary for a successful ADE infection. Here, we report that both DENV and DENV-ADE infection induce an early ISG (NOS2) expression through RLR-MAVS signalling axis independent of the IFNs signaling. Besides, DENV-ADE suppress this early antiviral response through increased autophagy formation rather than induction of IL-10 secretion. The early induced autophagic proteins ATG5-ATG12 participate in suppression of MAVS mediated ISGs induction. Our findings suggest a mechanism for DENV to evade the early antiviral response before IFN signalling activation. Altogether, these results add knowledge about the complexity of ADE infection and contribute further to research on therapeutic strategies.
Collapse
Affiliation(s)
- Xinwei Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
- Key Laboratory of The Second Affiliated Hospital of Kuming Medical College, Kunming 650101, PR China
| | - Yaofei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Duo Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Yujiao Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Lijuan Qiu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Junying Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Juemin Xi
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Xiaodan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| |
Collapse
|
232
|
Abstract
Autophagy is a lysosomal degradation process crucial for adaptation to stress and cellular homeostasis. In cancer, autophagy has been demonstrated to serve multifaceted roles in tumor initiation and progression. Although genetic evidence corroborates a role for autophagy as a tumor suppressor mechanism during tumor initiation, autophagy also sustains metabolic pathways in cancer cells and promotes survival within the harsh tumor microenvironment and in response to diverse anticancer therapies. Moreover, though traditionally viewed as an autodigestive process, more recent work demonstrates that autophagy also facilitates cellular secretion; the importance of these new functions of the autophagy pathway is being increasingly appreciated during cancer progression and treatment. In this review, we discuss how these evolving and diverse roles for autophagy both impede and promote tumorigenesis.
Collapse
Affiliation(s)
- J Liu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - J Debnath
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
233
|
Abstract
Salmonella enterica is an intracellular pathogen that causes diseases ranging from gastroenteritis to typhoid fever. Salmonella bacteria trigger an autophagic response in host cells upon infection but have evolved mechanisms for suppressing this response, thereby enhancing intracellular survival. We recently reported that S. enterica serovar Typhimurium actively recruits the host tyrosine kinase focal adhesion kinase (FAK) to the surface of the Salmonella-containing vacuole (SCV) (K. A. Owen et al., PLoS Pathog 10:e1004159, 2014). FAK then suppresses autophagy through activation of the Akt/mTORC1 signaling pathway. In FAK−/− macrophages, bacteria are captured in autophagosomes and intracellular survival is attenuated. Here we show that the cell-autonomous bacterial suppression of autophagy also suppresses the broader innate immune response by inhibiting production of beta interferon (IFN-β). Induction of bacterial autophagy (xenophagy), but not autophagy alone, triggers IFN-β production through a pathway involving the adapter TRIF and endosomal Toll-like receptor 3 (TLR3) and TLR4. Selective FAK knockout in macrophages resulted in rapid bacterial clearance from mucosal tissues after oral infection. Clearance correlated with increased IFN-β production by intestinal macrophages and with IFN-β-dependent induction of IFN-γ by intestinal NK cells. Blockade of either IFN-β or IFN-γ increased host susceptibility to infection, whereas experimental induction of IFN-β was protective. Thus, bacterial suppression of autophagy not only enhances cell-autonomous survival but also suppresses more-systemic innate immune responses by limiting type I and type II interferons. Salmonella enterica serovar Typhimurium represents one of the most commonly identified bacterial causes of foodborne illness worldwide. S. Typhimurium has developed numerous strategies to evade detection by the host immune system. Autophagy is a cellular process that involves the recognition and degradation of defective proteins and organelles. More recently, autophagy has been described as an important means by which host cells recognize and eliminate invading intracellular pathogens and plays a key role in the production of cytokines. Previously, we determined that Salmonella bacteria are able to suppress their own autophagic capture and elimination by macrophages. Building on that study, we show here that the inhibition of autophagy by Salmonella also prevents the induction of a protective cytokine response mediated by beta interferon (IFN-β) and IFN-γ. Together, these findings identify a novel virulence strategy whereby Salmonella bacteria prevent cell autonomous elimination via autophagy and suppress the activation of innate immune responses.
Collapse
|
234
|
Apoptosis, autophagy and unfolded protein response pathways in Arbovirus replication and pathogenesis. Expert Rev Mol Med 2016; 18:e1. [PMID: 26781343 PMCID: PMC4836210 DOI: 10.1017/erm.2015.19] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Arboviruses are pathogens that widely affect the health of people in different communities around the world. Recently, a few successful approaches toward production of effective vaccines against some of these pathogens have been developed, but treatment and prevention of the resulting diseases remain a major health and research concern. The arbovirus infection and replication processes are complex, and many factors are involved in their regulation. Apoptosis, autophagy and the unfolded protein response (UPR) are three mechanisms that are involved in pathogenesis of many viruses. In this review, we focus on the importance of these pathways in the arbovirus replication and infection processes. We provide a brief introduction on how apoptosis, autophagy and the UPR are initiated and regulated, and then discuss the involvement of these pathways in regulation of arbovirus pathogenesis.
Collapse
|
235
|
Maiuri MC, De Stefano D. Pathophysiologic Role of Autophagy in Human Airways. AUTOPHAGY NETWORKS IN INFLAMMATION 2016. [PMCID: PMC7123327 DOI: 10.1007/978-3-319-30079-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lung diseases are among the most common and widespread disorders worldwide. They refer to many different pathological conditions affecting the pulmonary system in acute or chronic forms, such as asthma, chronic obstructive pulmonary disease, infections, cystic fibrosis, lung cancer and many other breath complications. Environmental, epigenetic and genetic co-factors are responsible for these pathologies that can lead to respiratory failure, and, even, ultimately death. Increasing evidences have highlighted the implication of the autophagic pathways in the pathogenesis of lung diseases and, in some cases, the deregulated molecular mechanisms underlying autophagy may be considered as potential new therapeutic targets. This chapter summarizes recent advances in understanding the pathophysiological functions of autophagy and its possible roles in the causation and/or prevention of human lung diseases.
Collapse
|
236
|
Kimura T, Jain A, Choi SW, Mandell MA, Schroder K, Johansen T, Deretic V. TRIM-mediated precision autophagy targets cytoplasmic regulators of innate immunity. J Cell Biol 2015; 210:973-89. [PMID: 26347139 PMCID: PMC4576868 DOI: 10.1083/jcb.201503023] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
TRIM20 and TRIM21 are mediators of IFN-γ–induced autophagy, which act as autophagic receptor regulators that target specific inflammasome components and type I interferon response regulators for degradation by precision autophagy. The present paradigms of selective autophagy in mammalian cells cannot fully explain the specificity and selectivity of autophagic degradation. In this paper, we report that a subset of tripartite motif (TRIM) proteins act as specialized receptors for highly specific autophagy (precision autophagy) of key components of the inflammasome and type I interferon response systems. TRIM20 targets the inflammasome components, including NLRP3, NLRP1, and pro–caspase 1, for autophagic degradation, whereas TRIM21 targets IRF3. TRIM20 and TRIM21 directly bind their respective cargo and recruit autophagic machinery to execute degradation. The autophagic function of TRIM20 is affected by mutations associated with familial Mediterranean fever. These findings broaden the concept of TRIMs acting as autophagic receptor regulators executing precision autophagy of specific cytoplasmic targets. In the case of TRIM20 and TRIM21, precision autophagy controls the hub signaling machineries and key factors, inflammasome and type I interferon, directing cardinal innate immunity response systems in humans.
Collapse
|
237
|
Kimmey JM, Huynh JP, Weiss LA, Park S, Kambal A, Debnath J, Virgin HW, Stallings CL. Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection. Nature 2015; 528:565-9. [PMID: 26649827 PMCID: PMC4842313 DOI: 10.1038/nature16451] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 11/16/2015] [Indexed: 02/08/2023]
Abstract
Mycobacterium tuberculosis (Mtb), a major global health threat, replicates in macrophages (MΦ) in part by inhibiting phagosome-lysosome fusion, until IFN-γ activates the MΦ to traffic Mtb to the lysosome. How IFN-γ elicits this effect is unknown, but many studies suggest a role for macroautophagy (autophagy herein), a cellular process by which cytoplasmic contents are sequestered into an autophagosome and targeted for lysosomal degradation1. The involvement of autophagy has been defined based on studies in cultured MΦ or dendritic cells (DC) where Mtb colocalizes with autophagy (ATG) factors ATG5, ATG12, ATG16L1, p62, NDP52, Beclin1 and LC32–6, stimulation of autophagy increases bacterial killing6–8, and inhibition of autophagy allows for increased bacterial survival1,2,4,6,7. Notably, these studies reveal modest (e.g. 1.5- to 3-fold change) effects on Mtb replication. In contrast, Atg5fl/fl-LysM-Cre mice lacking ATG5 in monocyte-derived cells and neutrophils (polymorphic mononuclear cells, PMN) succumb to Mtb within 30 days4,9, an extremely severe phenotype similar to mice lacking IFN-γ signaling10,11. Importantly, ATG5 is the only ATG factor that has been studied during Mtb infection in vivo and autophagy-independent functions of ATG5 have been described12–18. For this reason, we used a genetic approach to elucidate the role for multiple ATG genes and the requirement for autophagy in resistance to Mtb infection in vivo. We have discovered that, contrary to expectation, autophagic capacity does not correlate with the outcome of Mtb infection. Instead, ATG5 plays a unique role in protection against Mtb by preventing PMN-mediated immunopathology. Furthermore, while ATG5 is dispensable in alveolar MΦ during Mtb infection, loss of Atg5 in PMN can sensitize mice to Mtb. These findings shift our understanding of the role of ATG5 during Mtb infection, reveal a new outcome of ATG5 activity, and shed light on early events in innate immunity that are required to regulate tuberculosis disease pathology and Mtb replication.
Collapse
Affiliation(s)
- Jacqueline M Kimmey
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Jeremy P Huynh
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Leslie A Weiss
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Sunmin Park
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Amal Kambal
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| |
Collapse
|
238
|
Burada F, Nicoli ER, Ciurea ME, Uscatu DC, Ioana M, Gheonea DI. Autophagy in colorectal cancer: An important switch from physiology to pathology. World J Gastrointest Oncol 2015; 7:271-284. [PMID: 26600927 PMCID: PMC4644850 DOI: 10.4251/wjgo.v7.i11.271] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/20/2015] [Accepted: 10/09/2015] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death in both men and women worldwide. Among the factors and mechanisms that are involved in the multifactorial etiology of CRC, autophagy is an important transformational switch that occurs when a cell shifts from normal to malignant. In recent years, multiple hypotheses have been considered regarding the autophagy mechanisms that are involved in cancer. The currently accepted hypothesis is that autophagy has dual and contradictory roles in carcinogenesis, but the precise mechanisms leading to autophagy in cancer are not yet fully defined and seem to be context dependent. Autophagy is a surveillance mechanism used by normal cells that protects them from the transformation to malignancy by removing damaged organelles and aggregated proteins and by reducing reactive oxygen species, mitochondrial abnormalities and DNA damage. However, autophagy also supports tumor formation by promoting access to nutrients that are critical to the metabolism and growth of tumor cells and by inhibiting cellular death and increasing drug resistance. Autophagy studies in CRC have focused on several molecules, mainly microtubule-associated protein 1 light chain 3, beclin 1, and autophagy related 5, with conflicting results. Beneficial effects were observed for some agents that modulate autophagy in CRC either alone or, more often, in combination with other agents. More extensive studies are needed in the future to clarify the roles of autophagy-related genes and modulators in colorectal carcinogenesis, and to develop potential beneficial agents for the prognosis and treatment of CRC.
Collapse
|
239
|
Rey-Jurado E, Riedel CA, González PA, Bueno SM, Kalergis AM. Contribution of autophagy to antiviral immunity. FEBS Lett 2015; 589:3461-70. [PMID: 26297829 PMCID: PMC7094639 DOI: 10.1016/j.febslet.2015.07.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 07/20/2015] [Accepted: 07/29/2015] [Indexed: 12/22/2022]
Abstract
Although identified in the 1960's, interest in autophagy has significantly increased in the past decade with notable research efforts oriented at understanding as to how this multi-protein complex operates and is regulated. Autophagy is commonly defined as a "self-eating" process evolved by eukaryotic cells to recycle senescent organelles and expired proteins, which is significantly increased during cellular stress responses. In addition, autophagy can also play important roles during human diseases, such as cancer, neurodegenerative and autoimmune disorders. Furthermore, novel findings suggest that autophagy contributes to the host defense against microbial infections. In this article, we review the role of macroautophagy in antiviral immune responses and discuss molecular mechanisms evolved by viral pathogens to evade this process. A role for autophagy as an effector mechanism used both, by innate and adaptive immunity is also discussed.
Collapse
Affiliation(s)
- Emma Rey-Jurado
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM U1064, Nantes, France.
| |
Collapse
|
240
|
Cui J, Chen Y, Wang HY, Wang RF. Mechanisms and pathways of innate immune activation and regulation in health and cancer. Hum Vaccin Immunother 2015; 10:3270-85. [PMID: 25625930 DOI: 10.4161/21645515.2014.979640] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Research on innate immune signaling and regulation has recently focused on pathogen recognition receptors (PRRs) and their signaling pathways. Members of PRRs sense diverse microbial invasions or danger signals, and initiate innate immune signaling pathways, leading to proinflammatory cytokines production, which, in turn, instructs adaptive immune response development. Despite the diverse functions employed by innate immune signaling to respond to a variety of different pathogens, the innate immune response must be tightly regulated. Otherwise, aberrant, uncontrolled immune responses will lead to harmful, or even fatal, consequences. Therefore, it is essential to better discern innate immune signaling and many regulators, controlling various signaling pathways, have been identified. In this review, we focus on the recent advances in our understanding of the activation and regulation of innate immune signaling in the host response to pathogens and cancer.
Collapse
Key Words
- AIM2, absent in melanoma 2
- ALRs, AIM2-like receptors
- AMPK, AMP activated protein kinase
- ASC, apoptosis-associated speck-like protein containing a CARD
- Atg16L, autophagy related 16-like
- BMM, bone marrow-derived macrophage
- CARD, caspase recruitment domain
- CDNs, cyclic dinucleotides
- CLRs, C-type lectin receptors
- CMV, cytomegalovirus
- CYLD, the familial cylindromatosis tumor suppressor gene
- DAMPs, danger-associated molecular patterns
- DCs, dendritic cells
- DDX41, DEAD (Asp-Glu-Ala-Asp) box polypeptide 41
- ER, endoplasmic reticulum
- GBP5, guanylate-binding protein 5
- GSK3β, Glycogen synthase kinase 3β
- HCC, hepatocellular carcinoma
- IFI16, interferon, gamma-inducible protein 16
- IFN, interferon
- IKK, IkB kinase
- IKKi, inducible IkB kinase
- IRAK, interleukin-1 receptor-associated kinase
- IRF, interferon regulatory factor
- KSHV, Kaposi's sarcoma-associated herpesvirus
- LBP, LPS-binding protein
- LGP 2, laboratory of genetics and physiology 2
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- LT, lethal toxin
- LUBAC, linear ubiquitin assembly complex
- MAVS, mitochondrial antiviral signaling protein
- MDA5, melanoma differentiation-associated protein 5
- MDP, muramyl dipeptide
- MIB, mind bomb
- MyD88, myeloid differentiation factor 88
- NAIPs, neuronal apoptosis inhibitory proteins
- NEMO, NF-kB essential modulator
- NLRs, Nod- like receptors
- NOD, nucleotide-binding oligomerization domain
- Nrdp1, neuregulin receptor degradation protein 1
- PAMPs, pathogen-associated molecular patterns
- PKC-d, protein kinase C delta
- PKR, dsRNA-dependent protein kinase
- PRRs
- PRRs, pathogen recognition receptors
- RACK1, receptor for activated C kinase 1
- RAUL, RTA-associated E3 ligase
- RIG-I, retinoic acid-inducible gene 1
- RIP, receptor-interacting protein
- RLRs, RIG-I-like receptors
- ROS, reactive oxygen species
- SARM, sterile a- and armadillo motif-containing protein
- SIGIRR, single Ig IL-1-related receptor
- SOCS, suppressor of cytokine signaling
- STING, stimulator of interferon gene
- TAK1, TGF-b-activating kinase 1
- TANK, TRAF family-member-associated NF-kB activator
- TBK1, TANK binding kinase 1
- TIR, Toll IL-1 receptor
- TIRAP, TIR domain-containing adapter protein
- TLRs, Toll-like receptors
- TRAF, TNFR-associated factor
- TRAILR, tumor-necrosis factor-related apoptosis-inducing ligand receptor
- TRAM, TRIF-related adaptor molecule
- TRIF, TIR domain-containing adaptor inducing IFN-b
- TRIMs, tripartite motif containing proteins
- TRIP, TRAF-interacting protein
- ULK1, autophagy related serine threonine UNC-51- like kinase
- cDC, conventional dendritic cell
- cGAS, cyclic GMP-AMP synthase
- cIAP, cellular inhibitor of apoptosis protein
- cancer
- iE-DAP, g-D-glutamyl-meso-diaminopimelic acid
- inflammation
- innate immunity
- pDC, plasmacytoid dendritic cell
- type I interferon
Collapse
Affiliation(s)
- Jun Cui
- a Key Laboratory of Gene Engineering of the Ministry of Education; State Key Laboratory of Biocontrol; School of Life Sciences ; Sun Yat-sen University ; Guangzhou , P. R. China
| | | | | | | |
Collapse
|
241
|
Wang L, Law HKW. The Role of Autophagy in Lupus Nephritis. Int J Mol Sci 2015; 16:25154-67. [PMID: 26506346 PMCID: PMC4632796 DOI: 10.3390/ijms161025154] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/05/2015] [Accepted: 10/19/2015] [Indexed: 12/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease characterized by the generation of immune responses to self-antigens. Lupus nephritis is one of the most common and severe complications in SLE patients. Though the pathogenesis of lupus nephritis has been studied extensively, unresolved questions are still left and new therapeutic methods are needed for disease control. Autophagy is a conserved catabolic process through which cytoplasmic constituents can be degraded in lysosome and reused. Autophagy plays vital roles in maintaining cell homeostasis and is involved in the pathogenesis of many diseases. In particular, autophagy can affect almost all parts of the immune system and is involved in autoimmune diseases. Based on genetic analysis, cell biology, and mechanism studies of the classic and innovative therapeutic drugs, there are growing lines of evidence suggesting the relationship between autophagy and lupus nephritis. In the present review, we summarize the recent publications investigating the relationship between autophagy and lupus nephritis and provide a new perspective towards the pathogenesis of lupus nephritis.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hunghom, Hong Kong, China.
| | - Helen Ka Wai Law
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hunghom, Hong Kong, China.
| |
Collapse
|
242
|
Autophagy and autophagy-related proteins in the immune system. Nat Immunol 2015; 16:1014-24. [DOI: 10.1038/ni.3273] [Citation(s) in RCA: 366] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/17/2015] [Indexed: 02/06/2023]
|
243
|
Nalpas NC, Magee DA, Conlon KM, Browne JA, Healy C, McLoughlin KE, Rue-Albrecht K, McGettigan PA, Killick KE, Gormley E, Gordon SV, MacHugh DE. RNA sequencing provides exquisite insight into the manipulation of the alveolar macrophage by tubercle bacilli. Sci Rep 2015; 5:13629. [PMID: 26346536 PMCID: PMC4642568 DOI: 10.1038/srep13629] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/31/2015] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium bovis, the agent of bovine tuberculosis, causes an estimated $3 billion annual losses to global agriculture due, in part, to the limitations of current diagnostics. Development of next-generation diagnostics requires a greater understanding of the interaction between the pathogen and the bovine host. Therefore, to explore the early response of the alveolar macrophage to infection, we report the first application of RNA-sequencing to define, in exquisite detail, the transcriptomes of M. bovis-infected and non-infected alveolar macrophages from ten calves at 2, 6, 24 and 48 hours post-infection. Differentially expressed sense genes were detected at these time points that revealed enrichment of innate immune signalling functions, and transcriptional suppression of host defence mechanisms (e.g., lysosome maturation). We also detected differentially expressed natural antisense transcripts, which may play a role in subverting innate immune mechanisms following infection. Furthermore, we report differential expression of novel bovine genes, some of which have immune-related functions based on orthology with human proteins. This is the first in-depth transcriptomics investigation of the alveolar macrophage response to the early stages of M. bovis infection and reveals complex patterns of gene expression and regulation that underlie the immunomodulatory mechanisms used by M. bovis to evade host defence mechanisms.
Collapse
Affiliation(s)
- Nicolas C Nalpas
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - David A Magee
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kevin M Conlon
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - John A Browne
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Claire Healy
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kirsten E McLoughlin
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kévin Rue-Albrecht
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland.,UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Paul A McGettigan
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kate E Killick
- Systems Biology Ireland, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Eamonn Gormley
- Tuberculosis Diagnostics and Immunology Research Centre, UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Stephen V Gordon
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland.,UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - David E MacHugh
- Animal Genomics Laboratory, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
244
|
Yakoub AM, Shukla D. Basal Autophagy Is Required for Herpes simplex Virus-2 Infection. Sci Rep 2015; 5:12985. [PMID: 26248741 PMCID: PMC4528227 DOI: 10.1038/srep12985] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a conserved catabolic process of the cell, which plays an important role in regulating plethora of infections. The role of autophagy in Herpes simplex virus-2 (HSV-2) infection is unknown. Here, we found that HSV-2 does not allow induction of an autophagic response to infection, but maintains basal autophagy levels mostly unchanged during productive infection. Thus, we investigated the importance of basal autophagy for HSV-2 infection, using pharmacological autophagy suppression or cells genetically deficient in an autophagy-essential gene (ATG5). Interference with basal autophagy flux in cells significantly reduced viral replication and diminished the infection. These results indicate that basal autophagy plays an indispensable role required for a productive infection. Importantly, this study draws a sharp distinction between induced and basal autophagy, where the former acts as a viral clearance mechanism abrogating infection, while the latter supports infection.
Collapse
Affiliation(s)
- Abraam M Yakoub
- 1] Department of Microbiology and Immunology, University of Illinois, Chicago, IL USA, 60612 [2] Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL USA, 60612
| | - Deepak Shukla
- 1] Department of Microbiology and Immunology, University of Illinois, Chicago, IL USA, 60612 [2] Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL USA, 60612
| |
Collapse
|
245
|
RNase L Cleavage Products Promote Switch from Autophagy to Apoptosis by Caspase-Mediated Cleavage of Beclin-1. Int J Mol Sci 2015; 16:17611-36. [PMID: 26263979 PMCID: PMC4581211 DOI: 10.3390/ijms160817611] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/17/2015] [Accepted: 07/27/2015] [Indexed: 01/03/2023] Open
Abstract
Autophagy and apoptosis share regulatory molecules enabling crosstalk in pathways that affect cellular homeostasis including response to viral infections and survival of tumor cells. Ribonuclease L (RNase L) is an antiviral endonuclease that is activated in virus-infected cells and cleaves viral and cellular single-stranded RNAs to produce small double-stranded RNAs with roles in amplifying host responses. Activation of RNase L induces autophagy and apoptosis in many cell types. However, the mechanism by which RNase L mediates crosstalk between these two pathways remains unclear. Here we show that small dsRNAs produced by RNase L promote a switch from autophagy to apoptosis by caspase-mediated cleavage of Beclin-1, terminating autophagy. The caspase 3-cleaved C-terminal fragment of Beclin-1 enhances apoptosis by translocating to the mitochondria along with proapoptotic protein, Bax, and inducing release of cytochrome C to the cytosol. Cleavage of Beclin-1 determines switch to apoptosis since expression of caspase-resistant Beclin-1 inhibits apoptosis and sustains autophagy. Moreover, inhibiting RNase L-induced autophagy promotes cell death and inhibiting apoptosis prolongs autophagy in a cross-inhibitory mechanism. Our results demonstrate a novel role of RNase L generated small RNAs in cross-talk between autophagy and apoptosis that impacts the fate of cells during viral infections and cancer.
Collapse
|
246
|
Netea-Maier RT, Plantinga TS, van de Veerdonk FL, Smit JW, Netea MG. Modulation of inflammation by autophagy: Consequences for human disease. Autophagy 2015. [PMID: 26222012 PMCID: PMC4836004 DOI: 10.1080/15548627.2015.1071759] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Autophagy and inflammation are 2 fundamental biological processes involved in both physiological and pathological conditions. Through its crucial role in maintaining cellular homeostasis, autophagy is involved in modulation of cell metabolism, cell survival, and host defense. Defective autophagy is associated with pathological conditions such as cancer, autoimmune disease, neurodegenerative disease, and senescence. Inflammation represents a crucial line of defense against microorganisms and other pathogens, and there is increasing evidence that autophagy has important effects on the induction and modulation of the inflammatory reaction; understanding the balance between these 2 processes may point to important possibilities for therapeutic targeting. This review focuses on the crosstalk between autophagy and inflammation as an emerging field with major implications for understanding the host defense on the one hand, and for the pathogenesis and treatment of immune-mediated diseases on the other hand.
Collapse
Affiliation(s)
- Romana T Netea-Maier
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,b Division of Endocrinology, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Theo S Plantinga
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Frank L van de Veerdonk
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,c Radboud Center for Infectious Diseases, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Johannes W Smit
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,b Division of Endocrinology, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Mihai G Netea
- a Department of Internal Medicine , Radboud University Medical Center , Nijmegen , The Netherlands.,c Radboud Center for Infectious Diseases, Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|
247
|
Boyle JP, Parkhouse R, Monie TP. Insights into the molecular basis of the NOD2 signalling pathway. Open Biol 2015; 4:rsob.140178. [PMID: 25520185 PMCID: PMC4281710 DOI: 10.1098/rsob.140178] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The cytosolic pattern recognition receptor NOD2 is activated by the peptidoglycan fragment muramyl dipeptide to generate a proinflammatory immune response. Downstream effects include the secretion of cytokines such as interleukin 8, the upregulation of pro-interleukin 1β, the induction of autophagy, the production of antimicrobial peptides and defensins, and contributions to the maintenance of the composition of the intestinal microbiota. Polymorphisms in NOD2 are the cause of the inflammatory disorder Blau syndrome and act as susceptibility factors for the inflammatory bowel condition Crohn's disease. The complexity of NOD2 signalling is highlighted by the observation that over 30 cellular proteins interact with NOD2 directly and influence or regulate its functional activity. Previously, the majority of reviews on NOD2 function have focused upon the role of NOD2 in inflammatory disease or in its interaction with and response to microbes. However, the functionality of NOD2 is underpinned by its biochemical interactions. Consequently, in this review, we have taken the opportunity to address the more ‘basic’ elements of NOD2 signalling. In particular, we have focused upon the core interactions of NOD2 with protein factors that influence and modulate the signal transduction pathways involved in NOD2 signalling. Further, where information exists, such as in relation to the role of RIP2, we have drawn comparison with the closely related, but functionally discrete, pattern recognition receptor NOD1. Overall, we provide a comprehensive resource targeted at understanding the complexities of NOD2 signalling.
Collapse
Affiliation(s)
- Joseph P Boyle
- Department of Biochemistry, University of Cambridge, Cambridge, UK Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | | | - Tom P Monie
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK MRC Human Nutrition Research, Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge, UK
| |
Collapse
|
248
|
Rubinsztein DC, Bento CF, Deretic V. Therapeutic targeting of autophagy in neurodegenerative and infectious diseases. J Exp Med 2015; 212:979-90. [PMID: 26101267 PMCID: PMC4493419 DOI: 10.1084/jem.20150956] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a conserved process that uses double-membrane vesicles to deliver cytoplasmic contents to lysosomes for degradation. Although autophagy may impact many facets of human biology and disease, in this review we focus on the ability of autophagy to protect against certain neurodegenerative and infectious diseases. Autophagy enhances the clearance of toxic, cytoplasmic, aggregate-prone proteins and infectious agents. The beneficial roles of autophagy can now be extended to supporting cell survival and regulating inflammation. Autophagic control of inflammation is one area where autophagy may have similar benefits for both infectious and neurodegenerative diseases beyond direct removal of the pathogenic agents. Preclinical data supporting the potential therapeutic utility of autophagy modulation in such conditions is accumulating.
Collapse
Affiliation(s)
- David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge CB2 OSP, England, UK
| | - Carla F Bento
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge CB2 OSP, England, UK
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology and Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131 Department of Molecular Genetics and Microbiology and Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| |
Collapse
|
249
|
Abstract
Autophagy, a lysosomal degradative pathway that maintains cellular homeostasis, has emerged as an innate immune defense against pathogens. The role of autophagy in the deregulated HIV-infected central nervous system (CNS) is unclear. We have found that HIV-1-induced neuro-glial (neurons and astrocytes) damage involves modulation of the autophagy pathway. Neuro-glial stress induced by HIV-1 led to biochemical and morphological dysfunctions. X4 HIV-1 produced neuro-glial toxicity coupled with suppression of autophagy, while R5 HIV-1-induced toxicity was restricted to neurons. Rapamycin, a specific mTOR inhibitor (autophagy inducer) relieved the blockage of the autophagy pathway caused by HIV-1 and resulted in neuro-glial protection. Further understanding of the regulation of autophagy by cytokines and chemokines or other signaling events may lead to recognition of therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajeev Mehla
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Ashok Chauhan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
250
|
Chan YK, Gack MU. RIG-I-like receptor regulation in virus infection and immunity. Curr Opin Virol 2015; 12:7-14. [PMID: 25644461 PMCID: PMC5076476 DOI: 10.1016/j.coviro.2015.01.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/07/2015] [Indexed: 02/07/2023]
Abstract
Mammalian cells have the intrinsic capacity to detect viral pathogens and to initiate an antiviral response that is characterized by the induction of interferons (IFNs) and proinflammatory cytokines. A delicate regulation of the signaling pathways that lead to cytokine production is needed to ensure effective clearance of the virus, while preventing tissue damage caused by excessive cytokine release. Here, we focus on the mechanisms that modulate the signal transduction triggered by RIG-I-like receptors (RLRs) and their adaptor protein MAVS, key components of the host machinery for sensing foreign RNA. Specifically, we summarize recent advances in understanding how RLR signaling is regulated by posttranslational and posttranscriptional mechanisms, microRNAs (miRNAs) and autophagy. We further discuss how viruses target these regulatory mechanisms for immune evasion.
Collapse
Affiliation(s)
- Ying Kai Chan
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Michaela U Gack
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|