首页 | 本学科首页   官方微博 | 高级检索  
     


The Interferon Stimulator Mitochondrial Antiviral Signaling Protein Facilitates Cell Death by Disrupting the Mitochondrial Membrane Potential and by Activating Caspases
Authors:Chia-Yi Yu  Ruei-Lin Chiang  Tsung-Hsien Chang  Ching-Len Liao  Yi-Ling Lin
Affiliation:Institute of Biomedical Sciences,1. Genomics Research Center, Academia Sinica,3. Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan, Republic of China2.
Abstract:Interferon (IFN) signaling is initiated by the recognition of viral components by host pattern recognition receptors. Dengue virus (DEN) triggers IFN-β induction through a molecular mechanism involving the cellular RIG-I/MAVS signaling pathway. Here we report that the MAVS protein level is reduced in DEN-infected cells and that caspase-1 and caspase-3 cleave MAVS at residue D429. In addition to its well-known function in IFN induction, MAVS is also a proapoptotic molecule that triggers disruption of the mitochondrial membrane potential and activation of caspases. Although different domains are required for the induction of cytotoxicity and IFN, caspase cleavage at residue 429 abolished both functions of MAVS. The apoptotic role of MAVS in viral infection and double-stranded RNA (dsRNA) stimulation was demonstrated in cells with reduced endogenous MAVS expression induced by RNA interference. Even though IFN-β promoter activation was largely suppressed, DEN production was not affected greatly in MAVS knockdown cells. Instead, DEN- and dsRNA-induced cell death and caspase activation were delayed and attenuated in the cells with reduced levels of MAVS. These results reveal a new role of MAVS in the regulation of cell death beyond its well-known function of IFN induction in antiviral innate immunity.In the battle of hosts and microbes, the innate immune system uses pathogen recognition receptors (PRRs) to sense pathogen-associated molecular patterns (23). There are several functionally distinct classes of PRRs, such as the transmembrane (TM) Toll-like receptors (TLRs) and the intracellular retinoic acid-inducible gene I (RIG-I)-like helicase (RLH) receptors (15, 23, 25, 38). RLHs, including RIG-I and melanoma differentiation-associated gene 5 (MDA5), comprise an N-terminal caspase recruitment domain (CARD), a middle DEXD/H box RNA helicase domain, and a C-terminal domain. RLHs sense intracellular viral RNA and initiate an antiviral interferon (IFN) response (1, 43). RIG-I binding to viral RNA triggers conformational changes that expose the CARD for subsequent signaling (42). The adaptor molecule providing a link between RIG-I and downstream events was identified independently by four research groups as a mitochondrial CARD-containing protein, which was named mitochondrial antiviral signaling protein (MAVS) (34), IFN-β promoter stimulator 1 (IPS-1) (12), virus-induced signaling adaptor (VISA) (40), and CARD adaptor-inducing IFN-β (Cardif) (24). We refer to this adaptor as MAVS in this paper. MAVS transduces signals from RIG-I through CARD-CARD interactions, which then lead to interferon regulatory factor 3 (IRF-3) and NF-κB activation of IFN-β induction through a signaling cascade involving IKKα/β/γ, IKKɛ, and TBK1 (15). Recently, a protein termed STING (11) or MITA (47) was identified as a mediator that acts downstream of RIG-I and MAVS and upstream of TBK1.MAVS protein contains an N-terminal CARD required for signaling, a proline-rich domain that interacts with TRAF3, and a C-terminal TM region that targets MAVS to the mitochondrial outer membrane (29). Several cellular and viral proteins target MAVS in the attenuation of the IFN induction pathway. Cleavage of MAVS by hepatitis C virus (HCV) and hepatitis A virus (HAV) proteases, at residues C508 (18, 24) and Q428 (41), respectively, results in the loss of MAVS mitochondrial localization, thereby disrupting its function in IFN induction. Another mitochondrial outer membrane protein, NLRX1, can sequester MAVS from its association with RIG-I and act as a negative regulator of the IFN pathway (28). MAVS was recently found to be cleaved and inactivated by caspases during apoptosis (31, 33).The caspases are a well-known family of cysteinyl aspartate-specific proteases. The diverse roles of caspases in the cell cycle, proliferation, differentiation, cytokine production, innate immune regulation, and microbial infection suggest various functions of caspases beyond apoptosis (13, 14). The caspases can be separated into two subfamilies, namely, the cell death and inflammation subfamilies. In response to apoptotic stimuli, the initiators caspase-2, -8, -9, and -10 and effectors caspase-3, -6, and -7 mediate cell death events. Caspase-1, -4, -5, and -12 are known as the inflammatory caspases. Caspase-1 is involved in the cleavage and maturation of cytokines (8, 17). Caspase-8 and -10 were discovered as essential components that mediate antiviral signaling (37). Caspase-1 and -3 are activated in innate immune signaling (32). These findings indicate that caspases are involved in the regulation of innate immunity, in addition to their well-known apoptotic role. However, the details of how caspases are activated, the role of caspase activation, and how caspases manipulate the signaling pathways in innate immunity are still obscure.The family Flaviviridae contains three genera: Hepacivirus, Flavivirus, and Pestivirus. Infections with flaviviruses, such as dengue virus (DEN), Japanese encephalitis virus, and West Nile virus, are emerging worldwide. DEN triggers IFN-β through a molecular mechanism involving the RIG-I/MAVS signaling pathway (5, 20). In this study, we found that MAVS is cleaved during DEN serotype 2 (DEN-2) infection, in a caspase-dependent manner; this contrasts with viral protease-dependent cleavage of MAVS during infection with HCV and HAV. In a cell-free caspase assay system, MAVS was cleaved at residue D429 by caspase-1 and caspase-3. Cleaved MAVS failed to induce IFN production and caspase activation, and overexpression of MAVS also triggered caspase activation, which then negatively regulated its own function. Importantly, the role of MAVS in viral infection was verified by knockdown of MAVS expression. We discuss the possible regulatory mechanisms of MAVS and the biological significance of this cleavage event by caspases in the context of understanding how these apoptosis-related proteases might achieve cross talk with the innate immune pathway during viral infection.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号