首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 62 毫秒
1.
2.
3.
4.
5.
6.
7.
8.
9.
Alpha interferon (IFN-α) is an approved medication for chronic hepatitis B. Gamma interferon (IFN-γ) is a key mediator of host antiviral immunity against hepatitis B virus (HBV) infection in vivo. However, the molecular mechanism by which these antiviral cytokines suppress HBV replication remains elusive. Using an immortalized murine hepatocyte (AML12)-derived cell line supporting tetracycline-inducible HBV replication, we show in this report that both IFN-α and IFN-γ efficiently reduce the amount of intracellular HBV nucleocapsids. Furthermore, we provide evidence suggesting that the IFN-induced cellular antiviral response is able to distinguish and selectively accelerate the decay of HBV replication-competent nucleocapsids but not empty capsids in a proteasome-dependent manner. Our findings thus reveal a novel antiviral mechanism of IFNs and provide a basis for a better understanding of HBV pathobiology.Hepatitis B virus (HBV) is a noncytopathic hepatotropic DNA virus which belongs to the family Hepadnaviridae (11, 44). Despite the fact that most adulthood HBV infections are transient, approximately 5 to 10% of infected adults and more than 90% of infected neonates fail to clear the virus and develop a lifelong persistent infection, which may progress to chronic hepatitis, cirrhosis, and primary hepatocellular carcinoma (4, 33, 34). It has been shown by several research groups that resolution of HBV and other animal hepadnavirus infection in vivo depends on both killing of infected hepatocytes by viral antigen-specific cytotoxic T lymphocytes and noncytolytic suppression of viral replication, which is most likely mediated by inflammatory cytokines, such as gamma interferon (IFN-γ) and tumor necrosis factor α (TNF-α) (10, 12, 15, 20, 26, 27, 48). Moreover, together with five nucleoside or nucleotide analogs that inhibit HBV DNA polymerase, alpha IFN (IFN-α) and pegylated IFN-α are currently available antiviral medications for the management of chronic hepatitis B. Compared to the viral DNA polymerase inhibitors, the advantages of IFN-α therapy include a lack of drug resistance, a finite and defined treatment course, and an increased likelihood for hepatitis B virus surface antigen (HBsAg) clearance (8, 39). However, only approximately 30% of treated patients achieve a sustained virological response to a standard 48-month pegylated IFN-α therapy (6, 32). Thus far, the antiviral mechanism of IFN-α and IFN-γ and the parameters determining the success or failure of IFN-α therapy in chronic hepatitis B remain elusive. Elucidation of the mechanism by which the cytokines suppress HBV replication represents an important step toward understanding the pathobiology of HBV infection and the molecular basis of IFN-α therapy of chronic hepatitis B.Considering the mechanism by which IFNs noncytolytically control HBV infection in vivo, it is possible that the cytokines either induce an antiviral response in hepatocytes to directly limit HBV replication or modulate the host antiviral immune response to indirectly inhibit the virus infection. However, due to the fact that IFN-α and -γ do not inhibit or only modestly inhibit HBV replication in human hepatoma-derived cell lines (5, 22, 23, 30), the direct antiviral effects of the cytokines and their antiviral mechanism against HBV have been studied with either an immortalized hepatocyte cell line derived from HBV transgenic mice or duck hepatitis B virus (DHBV) infection of primary duck hepatocytes (37, 53). While these studies revealed that IFN treatment significantly reduced the amount of encapsidated viral pregenomic RNA (pgRNA) in both mouse and duck hepatocytes, further mechanistic analyses suggested that IFN-α inhibited the formation of pgRNA-containing nucleocapsids in murine hepatocytes (52) but shortened the half-life of encapsidated pgRNA in DHBV-replicating chicken hepatoma cells (21). Moreover, the fate of viral DNA replication intermediates or nucleocapsids in the IFN-treated hepatocytes was not investigated in the previous studies.To further define the target(s) of IFN-α and -γ in the HBV life cycle and to create a robust cell culture system for the identification of IFN-stimulated genes (ISGs) that mediate the antiviral response of the cytokines (25), we established an immortalized murine hepatocyte (AML-12)-derived stable cell line that supported a high level of HBV replication in a tetracycline-inducible manner. Consistent with previous reports, we show that both IFN-α and IFN-γ potently inhibited HBV replication in murine hepatocytes (37, 40). With the help of small molecules that inhibit HBV capsid assembly (Bay-4109) (7, 47) and prevent the incorporation of pgRNA into nucleocapsids (AT-61) (9, 29), we obtained evidence suggesting that the IFN-induced cellular antiviral response is able to distinguish and selectively accelerate the decay of HBV replication-competent nucleocapsids but not empty capsids in a proteasome-dependent manner. Our findings provide a basis for further studies toward better understanding of IFN′s antiviral mechanism, which might ultimately lead to the development of strategies to improve the efficacy of IFN therapy of chronic hepatitis B.  相似文献   

10.
Enterovirus 71 (EV71) is a human pathogen that induces hand, foot, and mouth disease and fatal neurological diseases. Immature or impaired immunity is thought to associate with increased morbidity and mortality. In a murine model, EV71 does not facilitate the production of type I interferon (IFN) that plays a critical role in the first-line defense against viral infection. Administration of a neutralizing antibody to IFN-α/β exacerbates the virus-induced disease. However, the molecular events governing this process remain elusive. Here, we report that EV71 suppresses the induction of antiviral immunity by targeting the cytosolic receptor retinoid acid-inducible gene I (RIG-I). In infected cells, EV71 inhibits the expression of IFN-β, IFN-stimulated gene 54 (ISG54), ISG56, and tumor necrosis factor alpha. Among structural and nonstructural proteins encoded by EV71, the 3C protein is capable of inhibiting IFN-β activation by virus and RIG-I. Nevertheless, EV71 3C exhibits no inhibitory activity on MDA5. Remarkably, when expressed in mammalian cells, EV71 3C associates with RIG-I via the caspase recruitment domain. This precludes the recruitment of an adaptor IPS-1 by RIG-I and subsequent nuclear translocation of interferon regulatory factor 3. An R84Q or V154S substitution in the RNA binding motifs has no effect. An H40D substitution is detrimental, but the protease activity associated with 3C is dispensable. Together, these results suggest that inhibition of RIG-I-mediated type I IFN responses by the 3C protein may contribute to the pathogenesis of EV71 infection.Enterovirus 71 (EV71) is a single-stranded, positive-sense RNA virus belonging to the Picornaviridae family. The viral genome is approximately 7,500 nucleotides in length, with a single open reading frame that encodes a large precursor protein. Upon infection, this protein precursor is processed into four structural (VP1, VP2, VP3, and VP4) and seven nonstructural (2A, 2B, 2C, 3A, 3B, 3C, and 3D) proteins (32). EV71 infection manifests most frequently as the childhood exanthema known as hand, foot, and mouth disease (HFMD). Additionally, EV71 infection may cause neurological diseases, which include aseptic meningitis, brain stem and/or cerebellar encephalitis, and acute flaccid paralysis (32). Young children and infants are especially susceptible to EV71 infection. Since the initial recognition of EV71 in the United States, outbreaks have been reported in Southeast Asia, Europe, and Australia (1-3, 11, 14, 24, 30-32). Recently, large epidemics of HFMD occurred in the mainland of China (26, 42, 52).The mechanism of EV71 pathogenesis remains obscure. It is believed that immature or impaired immunity, upon EV71 infection, is associated with increased morbidity and mortality (7, 14, 17). In a murine infection model, lymphocyte as well as antibody responses reduce tissue viral loads and EV71 lethality (28). Notably, EV71 induces skin rash at the early stage and hind limb paralysis or death at the late stage. Oral infection leads to initial replication in the intestine and subsequent spread to various organs such as the spinal cord and the brain stem (8). Intriguingly, EV71 does not facilitate the production of type I interferon (IFN), a family of cytokines involved in first-line defense against virus infection. Indeed, administration of neutralizing antibody to IFN-α/β increases tissue viral loads and exacerbates the virus-induced disease (29).Type I IFN is produced in response to viral infections (22). For example, Toll-like receptor 3 (TLR3) in the endosome recognizes double-stranded RNA (dsRNA), where it recruits the adaptor Toll/interleukin-1 receptor (TIR) domain-containing adaptor inducing IFN-β (TRIF) (22). TRIF, together with tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3), then activates the two IKK-related kinases, TANK-binding kinase 1 (TBK1) and inducible IκB kinase (IKKi), both of which phosphorylate interferon regulatory factor 3/7 (IRF3/7) (10, 13, 36, 45). IRF3 or IRF7, in turn, stimulates the expression of target genes, such as IFN-α/β (33, 37, 39, 51). In parallel, TRIF also induces NF-κB activation via TRAF6 (18, 19). In addition, alternative mechanisms exist in host cells to detect cytosolic nucleic acids. Two RNA helicases, retinoid acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5), recognize viral RNA present in the cytoplasm and subsequently recruit the adaptor IFN promoter-stimulating factor 1 (IPS-1; also called Cardif, MAVS, and VISA) (22, 23, 54). The interaction of IPS-1, TRAF3, and TBK1/IKKi activates IRF3/IRF7 and induces the expression of IFN-α/β while the interaction of IPS-1 with the Fas-associated protein-containing death domain (FADD) leads to NF-κB activation. It has been shown that MDA5 recognizes long double-stranded RNAs, such as in cells infected with picornaviruses, whereas RIG-I senses 5′ triphosphate single-stranded RNA with poly(U/A) motifs and short dsRNA in cells infected with a variety of RNA viruses (16, 20, 40, 43).The objective of this study was to investigate the interaction of EV71 with the type I IFN system. We demonstrate that, unlike Sendai virus or double-stranded RNA, EV71 does not stimulate the expression of antiviral genes in mammalian cells. Among structural and nonstructural proteins encoded by EV71, the 3C protein is able to inhibit virus-induced activation of the IFN-β promoter. We provide evidence that when expressed in mammalian cells, the 3C protein suppresses RIG-I signaling by disruption of the RIG-I-IPS-1 complex and IRF3 nuclear translocation. While H40, KFRDI, and VGK motifs are involved, the protease and RNA binding activities are dispensable. Collectively, these results suggest that control of RIG-I by the 3C protein impairs type I IFN responses, which may contribute to the pathogenesis of EV71 infection.  相似文献   

11.
12.
Type I interferons (IFNs) play a critical role in the host defense against viruses. Lymphocytic choriomeningitis virus (LCMV) infection induces robust type I IFN production in its natural host, the mouse. However, the mechanisms underlying the induction of type I IFNs in response to LCMV infection have not yet been clearly defined. In the present study, we demonstrate that IRF7 is required for both the early phase (day 1 postinfection) and the late phase (day 2 postinfection) of the type I IFN response to LCMV, and melanoma differentiation-associated gene 5 (MDA5)/mitochondrial antiviral signaling protein (MAVS) signaling is crucial for the late phase of the type I IFN response to LCMV. We further demonstrate that LCMV genomic RNA itself (without other LCMV components) is able to induce type I IFN responses in various cell types by activation of the RNA helicases retinoic acid-inducible gene I (RIG-I) and MDA5. We also show that expression of the LCMV nucleoprotein (NP) inhibits the type I IFN response induced by LCMV RNA and other RIG-I/MDA5 ligands. These virus-host interactions may play important roles in the pathogeneses of LCMV and other human arenavirus diseases.Type I interferons (IFNs), namely, alpha interferon (IFN-α) and IFN-β, are not only essential for host innate defense against viral pathogens but also critically modulate the development of virus-specific adaptive immune responses (6, 8, 28, 30, 36, 50, 61). The importance of type I IFNs in host defense has been demonstrated by studying mice deficient in the type I IFN receptor, which are highly susceptible to most viral pathogens (2, 47, 62).Recent studies have suggested that the production of type I IFNs is controlled by different innate pattern recognition receptors (PRRs) (19, 32, 55, 60). There are three major classes of PRRs, including Toll-like receptors (TLRs) (3, 40), retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) (25, 48, 51), and nucleotide oligomerization domain (NOD)-like receptors (9, 22). TLRs are a group of transmembrane proteins expressed on either cell surfaces or endosomal compartments. RLRs localize in the cytosol. Both TLRs and RLRs are involved in detecting viral pathogens and controlling the production of type I IFNs (52, 60). In particular, the endosome-localized TLRs (TLR3, TLR7/8, and TLR9) play important roles in detecting virus-derived double-stranded RNA (dsRNA), single-stranded RNA (ssRNA), and DNA-containing unmethylated CpG motifs, respectively. In contrast, RIG-I detects virus-derived ssRNA with 5′-triphosphates (5′-PPPs) or short dsRNA (<1 kb), whereas melanoma differentiation-associated gene 5 (MDA5) is responsible for recognizing virus-derived long dsRNA as well as a synthetic mimic of viral dsRNA poly(I):poly(C) [poly(I·C)] (24, 60). Recognition of viral pathogen-associated molecular patterns (PAMPs) ultimately leads to the activation and nuclear translocation of interferon regulatory factors (IRFs) and nuclear factor κB (NF-κB), which, in turn, switches on a cascade of genes controlling the production of both type I IFNs and other proinflammatory cytokines (10, 11, 60).Lymphocytic choriomeningitis virus (LCMV) infection in its natural host, the mouse, is an excellent system to study the impact of virus-host interactions on viral pathogenesis and to address important issues related to human viral diseases (1, 45, 49, 67). LCMV infection induces type I IFNs as well as other proinflammatory chemokines and cytokines (6, 41). Our previous studies have demonstrated that TLR2, TLR6, and CD14 are involved in LCMV-induced proinflammatory chemokines and cytokines (66). The mechanism by which LCMV induces type I IFN responses, however, has not been clearly defined (7, 8, 31, 44). The role of the helicase family members RIG-I and MDA5 in virus-induced type I IFN responses has been recently established. RIG-I has been found to be critical in controlling the production of type I IFN in response to a number of RNA viruses, including influenza virus, rabies virus, Hantaan virus, vesicular stomatitis virus (VSV), Sendai virus (SeV), etc. In contrast, MDA5 is required for responses to picornaviruses (15, 25, 63).In the present study, we demonstrated that LCMV genomic RNA strongly activates type I IFNs through a RIG-I/MDA5-dependent signaling pathway. Our present study further demonstrated that the LCMV nucleoprotein (NP) blocks LCMV RNA- and other viral ligand-induced type I IFN responses.  相似文献   

13.
Intracranial (i.c.) infection of mice with lymphocytic choriomeningitis virus (LCMV) results in anorexic weight loss, mediated by T cells and gamma interferon (IFN-γ). Here, we assessed the role of CD4+ T cells and IFN-γ on immune cell recruitment and proinflammatory cytokine/chemokine production in the central nervous system (CNS) after i.c. LCMV infection. We found that T-cell-depleted mice had decreased recruitment of hematopoietic cells to the CNS and diminished levels of IFN-γ, CCL2 (MCP-1), CCL3 (MIP-1α), and CCL5 (RANTES) in the cerebrospinal fluid (CSF). Mice deficient in IFN-γ had decreased CSF levels of CCL3, CCL5, and CXCL10 (IP-10), and decreased activation of both resident CNS and infiltrating antigen-presenting cells (APCs). The effects of IFN-γ signaling on macrophage lineage cells was assessed using transgenic mice, called “macrophages insensitive to interferon gamma” (MIIG) mice, that express a dominant-negative IFN-γ receptor under the control of the CD68 promoter. MIIG mice had decreased levels of CCL2, CCL3, CCL5, and CXCL10 compared to controls despite having normal numbers of LCMV-specific CD4+ T cells in the CNS. MIIG mice also had decreased recruitment of infiltrating macrophages and decreased activation of both resident CNS and infiltrating APCs. Finally, MIIG mice were significantly protected from LCMV-induced anorexia and weight loss. Thus, these data suggest that CD4+ T-cell production of IFN-γ promotes signaling in macrophage lineage cells, which control (i) the production of proinflammatory cytokines and chemokines, (ii) the recruitment of macrophages to the CNS, (iii) the activation of resident CNS and infiltrating APC populations, and (iv) anorexic weight loss.Immune cell recruitment to and infiltration of the central nervous system (CNS) is central to the pathology of a variety of inflammatory neurological diseases, including infectious meningoencephalitis, multiple sclerosis, and cerebral ischemia (59, 60). Chemokines have been shown to be highly upregulated in both human diseases and animal models of neuroinflammation and are thought to be important mediators of immune cell entry into the CNS (59, 60). For example, during experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS), the chemokines CCL2 (monocyte chemoattractant protein 1 [MCP-1α]), CCL3 (macrophage inflammatory protein 1α [MIP-1α]), CCL5 (regulated upon activation, T-cell expressed and secreted [RANTES]), and CXCL10 (gamma interferon [IFN-γ]-inducible protein 10 [IP-10]) are produced by either resident CNS cells or infiltrating cells (27) and serve to amplify the ongoing inflammatory response (25, 28). However, in some EAE studies, neither CCL3 nor CXCL10 were required for disease (72, 73). During CNS viral infection, CXCL10 and CCL5 are highly produced in several models (2, 41, 48, 82). In addition, mice deficient in CCR5, which binds (among others) CCL3 and CCL5, do not display impaired CNS inflammation after certain viral infections (13). Thus, the role of chemokines in CNS inflammation is likely complex and dissimilar between autoimmune and viral infection models.IFN-γ is present in the CNS during autoimmunity and infection (7, 54, 69). Several studies suggest that IFN-γ can be a potent inducer of CNS chemokine expression. Adenoviral expression of IFN-γ in the CNS strongly induced CCL5 and CXCL10 mRNA and protein, and this induction was dependent on the presence of the IFN-γ receptor (50). In EAE and Toxoplasma infection, mice deficient in IFN-γ or the IFN-γ receptor demonstrated reduced expression of several chemokines, including CCL2, CCL3, CCL5, and CXCL10 (26, 69). However, given the near-ubiquitous expression of the IFN-γ receptor (44), the mechanisms by which IFN-γ regulates CNS chemokine production remain to be elucidated.We studied neuroinflammation and immune-mediated disease using a well-studied mouse model of infection with lymphocytic choriomeningitis virus (LCMV). Intracranial (i.c.) injection of mice with LCMV results in seizures and death 6 to 8 days after inoculation. The onset of symptoms is associated with a massive influx of mononuclear cells into the cerebrospinal fluid (CSF), meninges, choroid plexus, and ependymal membranes (6, 8, 18), as well as the presence of proinflammatory cytokines (7, 38). The immune response is critical for disease, since infection of irradiated or T-cell-depleted mice leads to persistent infection with very high levels of virus in multiple tissues without the development of lethal meningitis (18, 34, 64). i.c. LCMV infection of β2-microglobulin-deficient mice (β2m−/− mice) also results in meningitis and production of proinflammatory cytokines and chemokines; however, meningitis occurs with a later onset and lower severity compared to wild-type mice (17, 24, 53, 57). Interestingly, i.c. LCMV infection of these mice also causes severe anorexia and weight loss (33, 38, 46, 52, 57) that is mediated by major histocompatibility complex (MHC) class II-restricted, CD4+ T cells (17, 46, 53, 57). Anorexia and weight loss are also observed in wild-type mice, but they succumb to lethal meningitis shortly thereafter (33), making study of this particular aspect of disease difficult. LCMV-induced weight loss, similar to what we have observed in β2m−/− mice also occurs in perforin-deficient mice, which possess CD8+ T cells (37). Although some reports have observed weight loss after peripheral LCMV infection (11, 45), we note that these studies used high doses of the clone 13 strain of LCMV, in contrast to our studies which have used the Armstrong strain of LCMV and orders of magnitude less virus (33, 38, 46, 52, 57). Although we cannot exclude a contribution of peripheral cells to weight loss in our i.c. Armstrong infection model, we previously showed that this weight loss does not occur with peripheral infection with LCMV Armstrong (33, 38), indicating that interactions between the CNS and the immune system are contribute substantially to disease.During LCMV infection, there is biphasic production of IFN-γ: a small, early peak of IFN-γ (most likely produced by NK or NKT cells), followed by T-cell-mediated production of IFN-γ (23, 75). Further, both CD4+ T cells and CD8+ T cells produce large amounts of IFN-γ after LCMV infection and T-cell production of IFN-γ is critical for LCMV-induced weight loss (35). Chemokines, especially CXCL10, CCL5, and CCL2, and their receptors, are upregulated in the brain after i.c. LCMV infection (2, 13). Brain chemokine mRNA expression after i.c. LCMV infection is reduced in IFN-γ-deficient mice and relatively absent in athymic mice (2). However, the mechanism(s) by which T cells and IFN-γ mediate the effects on CNS chemokine expression, cellular infiltration into the CNS, and LCMV-induced anorexic weight loss remain unclear.In the present study, we focused on two major questions. The first question concerned the role of IFN-γ on immune cell recruitment to and chemokine/cytokine production within the CNS? We found that macrophages and myeloid dendritic cells (DCs) require IFN-γ for their accumulation within the CNS. Second, since macrophages and myeloid DCs are the predominant cellular infiltrate, we sought to determine whether IFN-γ signaling on these cells was direct with regard to their recruitment and to chemokine/cytokine production. We found that IFN-γ signaling in macrophage lineage cells contributes significantly to their recruitment, to chemokine production in the CNS, and to anorexic weight loss. Together, these data suggest that much of the proinflammatory effects of IFN-γ in the CNS are mediated by the effects of IFN-γ on CD68-bearing cells.  相似文献   

14.
15.
16.
17.
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.  相似文献   

18.
19.
20.
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