Báo cáo khoa học: Weapons of STAT destruction Interferon evasion by paramyxovirus V proteins ppt

8 186 0
Báo cáo khoa học: Weapons of STAT destruction Interferon evasion by paramyxovirus V proteins ppt

Đang tải... (xem toàn văn)

Thông tin tài liệu

MINIREVIEW Weapons of STAT destruction Interferon evasion by paramyxovirus V proteins Curt M. Horvath Department of Medicine, Evanston Northwestern Healthcare Research Institute, and Departments of Medicine and Biochemistry, Molecular Biology & Cell Biology, Northwestern University, Evanston, IL, USA The signal transducer and activator of transcription ( STAT) family of proteins fun ction to activate gene transcription downstream of myriad cytokine and growth factor signals. The prototype STAT proteins, STAT1 and STAT2, are required for innate and adaptive antimicrobial immune responses that result from interferon signal transduction. While many viruses have evolved the ability to avoid these antiviral cytokines, t he Paramyxoviruses are dist inct in their abilities to i nterfere directly with STAT proteins. Individual paramyxovirus species diffe r greatly in their precise mech- anism of STAT signaling evasion, but a virus-encoded pro- tein called V plays a central role in this process. The theme of V-dependent interferon evasion and its variations provide significant insights into virus–host interactions and viral immune evasion that c an help define tar gets for antiviral drug design. Exposure of the viral weapons of STAT destruction may also be instructive for application to STAT-directed therapeutics for diseases characterized by STAT hyperactivity. Keywords: antiviral; interferon; paramyxovirus; STAT; viral evasion. Interferons: the antiviral cytokines The interferon (IFN) family, including type I (IFNa,IFNb) and type II (IFNc), refers to a group of cytokines that a re capable of modulating diverse biological responses such as immune regulation, tumor inhibition, cell growth arrest, innate antimicrobial responses, and promotion of a daptive immunity. Type I IFNs have long been associated w ith the ability t o diminish v irus replication [ 1], and this antiviral activity is the result of IFN-induced changes in cellular gene expression (reviewed in [2–4]). Cellular response to IFN leads to th e establishment of an antiviral state, a process that requires new mRNA and protein synthesis of many IFN- stimulated gene (ISG) products that contribute to the antiviral responses required to limit diverse virus f amilies. Immediate responses to virus infection result in rapid transcriptional activation of type I IFN, typified by the single human IFNb gene (Fig. 1). This IFN induction is initiated by diverse virus replication in termediates, including dsRNA as well as other Toll-like receptor (TLR) ligands [5,6]. In response to these signals, serine/threonine kinases activate immediate-responding transcription factors inclu- ding interferon regulatory facto r (IRF) 3, AP1(ATF2/ c-Jun), and NFjB, which rapidly mobilize to the IFNb enhancer where they collaborate to recruit a series of transcriptional coactivators that remodel the enhancer chromatin a nd enable RNA pol II transcription [7]. The newly synthesized IFNb is secreted from the primary infected cell and signals to adjacent cells through direct binding to a transmembrane type I IFN receptor on the cell surface. The receptors are phosphorylated by associated Janus family tyrosine kinases, leading t o receptor tyrosine phosphorylation. Latent STAT2 in a ssociation with IRF9 [8,9] binds to these docking sites, and becomes phosphor- ylated, followed by the recruitment and tyrosine phosphory- lation of latent STAT1 [ 10]. The STATs heterodimerize via SRC homology 2 (SH2) domain–phosphotyrosine inter- actions, and together with the STAT-associated IRF9, assemble into a heterotrimeric complex known as the IFN- stimulated gene factor 3, ISGF3 [11–15]. ISGF3 rapidly accumulates in the nucleus, binds to conserved IFN- stimulated response e lement (ISRE) sequences on IFN a/ b-stimulated gene promoters, and increases their transcrip- tion rates. One ISG target is IRF7, which combines with IRF3 to amplify the IFN response by inducing the expression of the numerous IFNa genes [16–18]. DespitethenegativeselectivepressureexertedbyIFN signaling on viruses, the very existence of successful infectious and pathogenic viruses in IFN-competent hosts demonstrates their ability t o r esist host defenses. In fact, many well-characterized virus adaptations allow t hem to Correspondence to C. M. Horvath, Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Pan- coe Pavillion, Room 4401, 2200 Campus Drive, Evanston, IL 60208, USA. Tel.: +1 847 491 5530, E-mail: horvath@northwestern.edu Abbreviations: CTD, C-terminal domain; CRM1, chromosomal region maintenance 1; DDB1, UV-damaged DNA binding protein 1; E1, Ub-activating enzyme; E2, Ub-conjugating enzyme; E3, Ub- ligating enzyme; IFN, interferon; ISG, IFN-stimulated gene; ISRE, IFN-stimulated response element; NES, nuclear export signal; NDV, Newcastle disease virus; SH2, SRC homology 2; STAT, signal trans- ducer and activator of transcription; SV5, simian virus 5; TLR, Toll- like receptor; Ub, ubiquitin; VDC, V protein-dependent degradation complex; VIP, V interaction protein. (Received 7 January 2 004, revised 6 February 2004, accepted 7 October 2004) Eur. J. Biochem. 271, 4621–4628 (2004) Ó FEBS 2004 doi:10.1111/j.1432-1033.2004.04425.x evade IFN-induced innate antiviral responses through a number of access points vulnerable to viral invaders [3,19] (Fig. 2). For s ome viruses, the common early steps of IFN induction are targeted by general inhibitory mechanisms that can occur via dsRNA sequestration or signaling interference to antagonize IRF3 and NFjB pathways, protein kinase inhibition by protein–protein interaction, or TLR signaling interference by viral products. In other cases, virus-encoded soluble IFN receptors or receptor antagonists block cytokine signalling. Viruses can also block specific antiviral effectors to preserve key cellular machinery needed for their replication. Recent investigations of paramyxo- virus IFN evasion strategies have revealed unique abilitie s to directly target STAT components of IFN signal trans- duction. While phenotypically similar, the molecular mech- anisms of STAT-directed IFN evasion are as diverse as the viruses themselves. These similarities and differences are highlighted in the descriptions below. Paramyxoviruses and their V protein Paramyxoviruses e ncompass a large family of enveloped, negative strand RNA viruses that cause zoonotic diseases including significant hu man pathogens like measles virus Fig. 2. Targets for virus evasion of IFN antiviral responses. Vulnerable access points for virus evasion include: (1) Induction of IFN biosynthesis, (2) Interaction between IFN and receptor, (3) IF N signal transduction and (4) Activities of antiviral effectors. Fig. 1. IFN biosynthesis and antiviral signal transduction induced b y virus infection. Left cell illustrates IFNb biosynthesis. In r esponse to virus infection, Toll-like receptor (TLR) signal transduction, or intracellular dsRNA, pathogens activate IRF3 and NFjB transcription factors which combine wit h ATF2/ c-jun to init iate IF Nb tran scription. IjB, inhibitor of NFjB. Right cell illustrates the cellular response to the released IFNb. Interaction with specific cell surface receptor initiates a tyrosine phosphorylation signaling program. Upon tyrosine phosphorylation, latent STAT1 and STAT2 (which can b e p reassociated with IRF9 [8,9]) heter odimerize to induce the trimeric IRF9–STAT1–STAT2 complex, ISGF3. ISGF3 binds to response elements in the promoters of target antiviral genes, and increases the rate of tran scription. JAK1, Janus kinase 1; Tyk2, tyrosine kinase 2. 4622 C. M. Horvath (Eur. J. Biochem. 271) Ó FEBS 2004 and mumps virus, or the more treacherous Nipah virus and Hendra virus. The large family Paramyxoviridae is subdi- vided into several genera, including the Rubulavirus, Hen- ipavirus, Morbillivirus,andRespirovirus groups. All of these viruses s hare common structural, biochemical, and genetic elements including the s ingle-stranded RNA genome that encodes a small number of proteins, including surface glycoproteins and several subunits of an RNA-dependent RNA polymerase (reviewed in [20]). One locus contains a polycistronic gene that encodes two or more viral proteins from overlapping ORFs that code for the phosphopro tein, P, a s econd protein named V (Fig. 3), and in some species additional overprinted proteins called C, W, X or Y. Due to a unique coding strategy involving generation of alternate mRNAs via cotranscriptional insertion of nontemplated guanine nucleotides [21], the paramyxovirus P and V proteins are amino coterminal but have unique C-termini. Paramyxovirus V proteins ar e readily identifiable by a highly conserved cysteine-rich domain at their C-termini derived from the overlapping ORF [20–22]. This conserved C-terminal domain (CTD) is approximately 50% identical among all p aramyxovirus V proteins and contains seven invariant cysteine residues. This domain enables the V protein to bind two atoms of zinc, a stoichiometry similar to that found in some cellular zinc-binding proteins [22,23]. Aside from this outward resemblance, it is important to note that V proteins have no cellular homologues and that the spacing of CTD cysteine residues is not consistent with known cellular zinc-binding domains including the RING, PHD, or LIM motifs [ 24]. Paramyxovirus host evasion has been ascribed to this locus, and a diverse r ange o f host evasion activities, including IFN signaling i nhibition [25], prevention of apoptosis [26,27], cell cycle alterations [28], inhibition of double-stranded RNA signaling [27,29], and prevention of IFN biosynthesis [26,27,29] have been ascribed to paramyxovirus V proteins. A number of recent findings demonstrate that a fundamentally important activity associated with a variety of paramyxovirus V proteins is direct interference with STAT p rotein function, but individual genera within the family exhibit remarkably diverse mechanisms of STAT inhibition. Rubulavirus V proteins: STAT ubiquitin ligases The STAT proteins are well known to cycle between active and inactive states as the result of reversible post-transla- tional modification, namely tyrosine phosphorylation and dephosphorylation [30–32]. The estimated half-lives of STAT1 and STAT2 are in the o rder of days rather than hours [32–34], but this long half-life can be greatly reduced upon infection with Rubulavirus species or following expression of the Rubulavirus V protein. In the prototype example, STAT1 protein accumulation was found to be dramatically reduced by infection of cells with simian virus 5 (SV5) [25]. This STAT1 targeting w as conferred by the sole expression of the SV5 V protein, and similar STAT degradation properties were soon found to be shared by V proteins from a variety of Rubulaviruses [2,25,34–40]. Chemical proteasome inhibitors can prevent STAT degra- dation by Rubulavirus V proteins [25,34]. Moreover, expression of the Rubulavirus V proteins induces polyubi- quitylation of specific target STATs [34,39–41]. Character- ization of bacterially expressed SV5 and type II human parainfluenza virus (HPIV2) V proteins in vitro revealed an intrinsic ability to catalyze t he transfer of u biquitin (Ub) in a reaction that required ATP, Ub-activating enzyme (E1), and Ub-conjugating enzyme (E2). This intrinsic enzymatic activity meets the definition of a Ub -ligating e nzyme ( E3), but the in vitro reaction failed to fully recapitulate the native reaction because it w as substrate-independent and g ener- ated only mono-Ub transfer rather than a poly-Ub chain [41]. In intact cells that express V protein, the complete Fig. 3. Paramyxovirus c oding s trategies f or accessing the conserved V protein CTD. Diagrams illustrate the open reading frames generated in alternate mRNAs that encode C-terminally unique P and V proteins. Coloring ind icates common translational reading frames. C TD, cysteine-rich, V-specific C-terminal domain. (A) Coding st rategy used by the Rubulavirus genus. The c olinea r mRNA e ncod es the V protein from a single translational reading frame , but site-sp ecific addition of two nontemplated guanine nucleotides (+2G) ge nerates a second ÔeditedÕ mRNA encoding the P protein from two overlapping reading frames. (B) Coding strategy used by th e Henipavirus, Morbillivirus,andRespirovirus genera. The colinear mRNA encodes the P protein, but sit e-specific addition o f a single nontemplated guanine nu cleotide (+1G) generates a second ÔeditedÕ mRNA encoding the V protein from two overlapping reading frames. In Sendai virus, both transcripts encode a third overlapping open reading frame that encodes a nested set of C proteins (C¢, C, Y1, and Y2). ( C) Comparison o f the am ino acid se quen ces in the V-specific C-te rminal domain of several paramyxoviruses. Boxes highlight conserved amino acids. Ó FEBS 2004 Weapons of STAT destruction (Eur. J. Biochem. 271) 4623 STAT targeting b y polyubiquitylation results i n protea- some-dependent degradation. It is striking that, in spite of the high amino acid sequence identity between V proteins a nd similar abilities to target STAT pr oteins for proteasomal degradation, Rubulavirus species differ in their specificity. While the SV5 V protein can t arget STAT1 for polyubiquitylation and proteasomal degradation, HPIV2 V protein targets STAT2 [34], and mumps v irus V protein can eliminate both STAT1 [42] and STAT3 [40]. Affinity purification of Rubulavirus Vproteins from host cells identified strikingly s imilar p atterns o f V interaction protein (VIP) partners (Fig. 4) but in detail each species also exhibited unique superim posed VIP p atterns. These differences in the VIP composition have been suggested to account for differential V protein activities and target specificity [40,41]. The STAT-targeting machinery consists of V protein- dependent degradation co mplexes (VDCs) that contain the V protein and VIPs including STAT1 and STAT2 (and STAT3 in the case of mumps virus). A number of additional cellular proteins including DDB1, a UV-damaged DNA binding protein [40,41,43,44], and members of the C ullin family of ubiquitin ligase subunits including Cullin 4A are also required [40,41] (Fig. 4). RNA interference experiments demonstrate that DDB1 a nd Cullin 4A are required f or STAT1 degradation by SV5, lending support t o the model that the VDC is a coalition of virus-encoded and host factors that together function as a STAT-directed E3 ubiquitin ligase enzyme [41]. Somatic cell genetics and biochemical analysis has revealed that all of the Rubulavirus V proteins require the participation of a nontarget STAT for their in vivo E3 Ub ligase activity [36]. SV5 can only target STAT1 in cells that express S TAT2, w hile HPIV2-mediated STAT2 degrada- tion fails in the absence of STAT1. For mumps virus, STAT1 targeting requires cellular STAT2, but STAT3 targeting is STAT2-independent [40]. A powerful c onfirma- tion for the role of STAT2 in STAT1 destruction by SV5 was provided by the discovery that STAT2 acts as a host range determinant for this virus [35]. SV5 does not replicate efficiently or cause STAT1 degradation in the mouse [45,46], where the murine STAT2 protein is unusually divergent in amino acid sequence [ 47–49]. Expression of human STAT2 in mouse c ells Ôrescues Õ the defective STAT1 targeting and provides the virus wit h a replication advantage [35]. The Newcastle disease virus (NDV, a member of the Avulovirus genus that is restricted to avian species) also encodes a V protein that can antagonize the avian IFN system [50]. Like SV5, the NDV IFN inhibition is species-restricted. The molecular b asis underlying this NDV species-specificity has not yet b een revealed, but it is interesting to speculate that avian STAT2 might be involved. Henipavirus V proteins: STAT sequestration in high molecular mass cytoplasmic complexes Nipah virus and Hendra virus are the two known species of a recently emerged and deadly paramyxovirus genus, Henipavirus, that was responsible for outbreaks of res- piratory disease and fatal encephalitis in humans and livestock in Malaysia and Australia [51,52]. Both Henipa- virus species were demonstrated to share V-dependent IFN signaling evasion p roperties with other paramyxoviru ses [50,53–55]. Nucleotide sequencing of the Henipavirus genomes revealed many similarities with other paramyxoviruses, including a polycistronic gene encoding a V protein CTD [52,56]. In comparison to the STAT-degrading Rubulavirus V proteins, the V proteins of Nipah virus and Hend ra virus share  50% amino acid identity within the CTD (Fig. 5). The Henipavirus V protein N-terminus is larger and entirely unique compared to other paramyxovirus proteomes and has no obvious homology to any cellular protein. This sequence divergence between Henipavirus and Rubulavirus V proteins indicates an alternate mechanism of IFN signaling inhibition. The Henipavirus V proteins are overall  58% identical in amino acid sequence, with  83% identity between amino acids 1–140,  44% identity Fig. 4. Schematic diagram of the Rubulavirus VDC ubiquitin ligase c omplex. In this model, the box representation of the SV5 V protein serves as a nucleation site for protein inter- actions that coordinate the transfer of ubiqu- itin (Ub) via Ub-conjugating enzymes (E2) to the specific STAT protein target (in this case, STAT1). Colored ovals represent the cellular V i nteraction protein com ponents required f or complete E3 Ub ligase a ctivity to g enerate polyubiquitylation of STATs, some of which are identified as DDB1, Cullin 4A, STAT1, STAT2. Also depicted are the Ub-activating enzyme, E1, and polyubiquitylation leading to degradat ion via the prote asome . This model is illustrative only, is not drawn to scale, and does not accurately p ortray protein inter- action sites or stoichiometry. 4624 C. M. Horvath (Eur. J. Biochem. 271) Ó FEBS 2004 between amino acids 141–405, and  80% identity within the CTD (amino acids 406–457). This sequence conserva- tion accounts for the functional similarity in the IFN evasion activities of the Nipah virus and Hendra v irus V proteins. Both Henipavirus V p roteins have b een demon- strated to subvert I FN responses by sequester ing STAT1 and STAT2 in high molecular mass cytoplasmic complexes without inducing their degradation [53,54]. This c omplex formation prevents IFN-induced STAT tyrosine phos- phorylation [53]. In addition to the ability to bind t o both S TAT1 and STAT2, the Henipavirus V proteins e xhibit nuclear–cyto- plasmic shuttling behavior that depends on chromosomal region maintenance 1 (CRM1)-dependent nuclear export signals. Not only does this shuttling affect the steady-state subcellular distribution of t he V protein, but it also alters the distribution of the latent STAT1. STAT1 is typically observed in both the cytoplasm and nucleus of unstimulated cells, and expression of the Henipavirus V p rotein efficiently relocalizes the latent STAT1 protein to the cytoplasm [53,54]. Unexpectedly, despite the high degree of sequence conservation within the cysteine-rich CTD, it is dispensable for IFN signaling inhibition [50]. Dissection of Nipah V protein functional domains revealed insights into the molecular mechanisms underlyin g Henipavirus IFN evasion and explained the dispensable role of the CTD [55]. Three V protein activities, nuclear export, STAT protein interaction, and IFN signaling i nhibition, all map to the N-terminal portion (Fig. 5 B). A no vel nuclear export signal (NES) was i dentified within Nipah V amino acids 174–192. Deletion or substitution within the N ES prevents V p rotein cytoplasmic accumulation and also prevents redistribution of latent STAT1 to the cytoplasm. However, the ability to thwart IFN-dependent STAT1 and STAT2 nuclear translocation r emains intact r egardless of NES mutation, suggesting that the shuttling behavior of the V protein has a distinct role in Henipavirus biology. Dissection of the V protein domains involved in IFN evasion activity and STAT protein interactions revealed that these functions also map within Nipah V amino acid residues 100–300. STAT1 binds independently to residues 100–160, and this interaction site is the primary evasion motif, sufficient t o block IFN signaling responses. The amino coterminal P and W proteins [57], or artificial fusion proteins that share t his sequence motif [55] can also prevent IFN signaling. Moreover, STAT1 binding is a prerequisite for S TAT2 binding, a nd association with STAT2 conse- quently requires a large overlapping binding s ite between residues 100–300. Evidence from site-directed mutagenesis suggests that contact between STAT2 and Nipah V requires a conserved peptide including amino acids 230–237. Hence, in intact cells, a coordinately assembled trimeric V–STAT1– STAT2 complex forms that inhibits IFN signal transduc- tion. As these p rotein interaction domains are absolutely required for V protein IFN evasion activity, they are prime candidates for therapeutic intervention with Henipavirus outbreaks. The region of STAT1 bound by Nipah V was also determined. Nipah V binds to STAT1 but not to STAT3, and only binds to chimeric STAT1–STAT3 fusion proteins when the C-terminal region was derived from S TAT1. The results indicate that a STAT1 fragment containing the linker domain and SH2 domain is the target site for Henipavirus V protein interaction, two regions important for STAT activation, dimerization, and DNA binding. Morbillivirus V proteins: inhibition of STAT nuclear translocation Measles virus, a prototype species of the Morbillivirus genus, encodes a V protein distinct from both the Rubulavirus and Henipavirus genera, sharing only  20% overall amino acid sequence identity. Despite the divergence, measles virus V protein is an efficient inhibitor of IFN signal transduction but acts via a mechanism distinct from either Rubulavirus or Henipavirus V proteins [58]. Measles virus V protein expression effe ctively prevents both IFNa/b and IFNc- induced transcriptional responses. The measles virus V protein does not degrade STATs or prevent IFN-induced STAT protein activating tyrosine phosphorylation, but effectively prevents IFN-induced STAT1 and STAT2 nuclear import. Unlike the Henipaviruses, measles V does not shuttle between nucleus and cytoplasm, and conse- quently does not alter the distribution pattern of latent STAT1. Affinity chromatography demonstrated that the measles V protein copurifies STAT1, STAT2, STAT3, and IRF9, but not the cellular c omponents required f or Rubulavirus VDC ubiquitin ligase function, in agreemen t with its distinct mechanism of action. In addition, measles V binds to an IFN receptor subunit (IFNAR2.2 or b Long ; H. Palosaari and C. M. Horvath, unpublished observations) and a signaling Fig. 5. Henipavirus V protein sequence conservation and domain structure. (A) Comparison of Nipah virus and Hendra virus V proteins to SV5 V and HPIV2 V. Percent sequence identities were d etermine d using NCBI BLAST algorithms. (B) Illustration of functional domains mapped in the Nipah virus V protein. (Adapted f rom [55], see text for details.) Ó FEBS 2004 Weapons of STAT destruction (Eur. J. Biochem. 271) 4625 adaptor, RACK1 [59], possibly indicating multivalent receptor i nteractions. The measles V-dependent binding of STAT3 partially inhibits signaling by IL6 and v-Src, which, in conjunction with the mumps virus S TAT3 degradation, further suggests a role for STAT3 in antiviral responses. The ability to prevent STAT nuclear import is also observed in measles virus-infected cells, where a dramatic redistribution of cellular STAT proteins is also observed. In measles-infected cells, a portion of the STAT1 and STAT2 proteins are redistributed to cytoplasmic aggregates that also stain for the viral nucleocapsid protein and nucleic acids [58]. Similar intracellular a ggregates are observed for many viruses, including mumps virus, where STAT2 is condensed to cytoplasmic bodies. It is enticing to speculate that these bodies represent intracellular sites of virus replication or assembly, possibly i ndicating that the STATs may also play some role in measle s virus replication. Respirovirus V and C proteins Sendai v irus, the most studied member of the Respirovirus genus, h as also evolved strategies t o e vade the host IFN response. Lik e other paramyxoviruses, Sendai virus contains a polycistronic P /V gene encoding multiple proteins, c om- plicated by the presence of several additional polypeptides derived from alternative nested translational initiation sites that encode the collective C proteins (C, C¢, Y1 and Y2 [20]) from a third overlapping translational reading frame (Fig. 3 B). Investigation of V-deficient recombinant viruses indicates that the V protein of Sendai appears to p lay a role in pathogenesis in animal infections, but its exact function has not been well characterized [60]. However, the four Sendai virus C proteins have been found to block IFN signaling [61–64]. Several studies of C protein functions in IFN evasion have been conducted, and a broad range of activities are documented. C p roteins have b een shown to bind STAT1 and induce its shift to a high molecular mass complex [65]. Other studies report that C proteins inhibit both STAT1 and STAT2 tyrosine phosphorylation [61,66]. It has also been reported that the C proteins cau se prolonged tyrosine phosphorylation of STAT1, and can impair S TAT1 serine phosphorylation [67]. Remarkably, C proteins have been described to cause mono-ubiquitylation and degrada- tion of the STAT1 protein in certain mou se cell lines, but with a concomitant increase in and preservation of tyrosine phosphorylated STAT1 [66,68]. The mechanistic basis for C protein effects on the IFN re sponse will need to be examined carefully to clarify the role of this protein in host evasion. Furthermore, independent studies of the Sendai virus V protein’s potential for STAT inhibition are required to clarify the molecular basis for Respirovirus IF N antagonism . Conclusions The diverse mechanisms that have evolved for V protein- dependent IFN e vasion provide many insights into STAT protein inhibition that might not be easily discerned by laboratory investigations. Discovery of new paramyxovi- ruses and their IFN evasion properties will almost certainly reveal novel mechanisms of STAT protein antagonism, and may also uncover new functions for STAT proteins. Probing the molecular details of virus-designed STAT inhibitors will not only y ield new t herapeutic targets a nd vaccination strategies for the control of the infectious diseases themselves, but will also undoubtedly provide insights into new ways to regulate h yperactive cytokine– JAK-STAT signaling that is characteristic of neoplastic and inflammatory diseases. Acknowledgements The author is grateful to all the members of the Horvath Laboratory, and wishes t o acknowle dge the contributions in the study of V proteins made by Jean-Patrick Parisien, Cristian Cruz, Christina Ulane, Jason Rodriguez, Heidi Palosaari, and Tom Kraus. P aramyxovirus research in the Horvath Laboratory is supported by NIH grants AI-50707 and AI-55733. References 1. Isaacs, A. & Lindemann, J. ( 1957) Viru s i nterference. I. The interferon. Proc. R. Soc. Lond. B. 147, 258–267. 2. Goodbourn, S., Didcock, L. & Randall, R.E. (2000) Interferons: cell signalling, immune modulation, antiviral response and virus countermeasures. J. Gen. Virol. 81 , 2341–2364. 3. Samuel, C.E. (2001) Antiviral actions of interferons. Clin. Microbiol. Rev. 14, 778–809. 4. Pestka, S .A., Langer, J.A., Zoon,K.C.&Samuel,C.E.(1987) Interferons and their a ctions. Annu. Rev. Biochem. 56, 727–777. 5. Doyle, S., Vaidya, S., O’Connell, R., D adgostar, H., Dempsey, P., Wu,T.,Rao,G.,Sun,R.,Haberland,M.,Modlin,R.&Cheng, G. (2002) IRF3 mediates a TLR3/TLR4-specific antiviral gene program. Immunity 17, 251–263. 6. Vaidya, S.A. & Cheng, G. (2003) Toll-like receptors and innate antiviral responses. Curr. Opin. Immunol. 15, 402–407. 7.Munshi,N.,Yie,Y.,Merika,M.,Senger,K.,Lomvardas,S., Agalioti, T. & Thanos, D. (1999) The IFN-beta e nhancer: a paradigm for u nderstandin g activation and repressio n of i nducible gene expression. Cold Spring Harb. Symp. Quant. Biol. 64, 149– 159. 8. Lau, J.F., Parisien, J P. & Horvath, C.M. (2000) Interferon reg- ulatory factor subcellular localizationisdeterminedbyabipartite nuclear localization signal in the DNA-binding domain and interaction with cytoplasmic retention factors. Proc. N atl Acad. Sci. USA 97, 7278–7283. 9. Horvath, C.M., Stark, G.R., Kerr, I.M. & Darnell, J.E. (1996) Interactions between STAT and n on-STAT prote ins in the Interferon stimulated ge ne factor 3 transcription c omplex. Mol. Cell. Biol. 16, 6957–6964. 10. Li, X., Leung, S., Kerr, I.M. & Stark, G.R. ( 1997) Functional subdomains of STAT2 required for preassociation with the alpha interferon rec eptor and f or signaling. Mol. Cell. Biol. 17, 2048– 2056. 11. Horv at h , C.M . (20 00 )STAT proteins and transcriptional responses to extracellular signals. Trends Biochem. Sci. 25, 496–502. 12. Darnell, J.E. Jr, K err, I.M. & Stark, G .M. (1994) Jak-STAT pathways and transcriptional activation in resp on se to IF Ns and other extracellular sign aling proteins. Science 264, 1415–1421. 13. Kessler, D.S., Veals, S.A., Fu, X Y. & L evy, D.E. (1990) Inter- feron-a regulates nuclear translocation and DNA-binding affinity of ISGF-3, a multimeric transcriptional activator. Genes Dev. 4, 1753–1765. 14. Aaronson, D.S. & Horvath, C.M. (2002) A road map for those who don’t know JAK-STAT. Science 296, 1653–1655. 15. Lau, J.F. & Horvath, C.M. (2002) Mechanisms of Type I Inter- feron Cell Signaling and STAT-Mediated Transcriptional Responses. Mt Sinai J . Med. 69, 156–168. 4626 C. M. Horvath (Eur. J. Biochem. 271) Ó FEBS 2004 16. Levy, D.E., Marie, I., Smith, E. & Prakash, A. (2002) Enhance- ment and diversification of IFN induction by IRF-7-mediated positive feedback. J. Interferon Cytokine Res. 22, 87–93. 17. Grandvaux, N., t enOever, B.R., Servant, M.J. & Hiscott, J. (2002) The interferon antiviral response: from viral invasion to evasion. Curr.Opin.Infect.Dis.15, 259–267. 18. Barnes, B., Lubyova, B. & Pitha, P.M. (2002) On the role of IRF in host defense. J. Interferon Cytokine Res. 22, 59–71. 19. Levy, D.E. & Garcia-Sastre, A. (2001) The virus battles: IFN induction of t he antiviral state and mechanisms of viral evasion. Cytokine Growth Factor Rev. 12 , 143–156. 20. Lamb, R.A. & Kolakofsky, D. (2001) Paramyxoviridae: The viruses a nd their replication. In Field s Virology (Fields, B.N., Knipe, D.M., Howley, P.M. & Griffin, D.E., eds), pp. 1305–1340. Lippincott-Raven Publishers, Philadelphia. 21. Thomas, S.M., Lamb, R.A. & Paterson, R.G. (1988) Two mRNAs th at differ by two nontemplated nucleotides encode the amino coterminal proteins P and V of the paramyxovirus SV5. Cell 54, 891–902. 22. Paterson, R.G., Leser, G.P., Shaughnessy, M.A. & Lamb, R.A. (1995) The paramyxovirus SV5 V protein binds two atoms of zinc and is a structural component of virions. Virology 208,121– 131. 23. Liston, P. & Briedis, D.J. (1994) Measles virus V prote in binds zinc. Virology 198, 399–404. 24. Capili, A.D., Schultz, D.C., Rauscher, I.F. & Borden, K.L. (2001) Solution structure of t h e PHD domain from the K AP-1 cor- epressor: structural determinants for PHD, RING and LIM zinc- binding domains. EMBO J. 20, 165–177. 25. Didcock, L., Young, D .F., Goodbourn, S . & Randall, R.E. (1999) The V protein of simian virus 5 inhibits i nterferon signalling by targeting STAT1 for p roteasome-mediated degradation. J. Virol. 73, 9928–9933. 26. Wansley, E.K. & Parks, G.D. (2002) Naturally occurring sub- stitutions in the P/V gene convert the noncytopathic paramyxo- virus simian virus 5 into a virus that induces alph a/bet a interferon synthesis and cell de ath. J. Virol. 76, 10109–10121. 27. He, B., Pat erson, R.G., Stock, N., Durbin, J.E., Durbin, R.K., Goodbourn, S., Randall, R.E. & Lamb, R.A. (2002) Recovery of paramyxovir us sim ian virus 5 with a V protein lacking the con- served cysteine-rich domain: the multifunctional V protein blocks both inte rferon-be ta induction and interferon signaling. Virology 303, 15–32. 28. Lin, G.Y. & Lamb, R.A. (2000) The paramyxovirus simian virus 5 V protein slows progression of the cell cycle. J. Virol. 74, 9152– 9166. 29. Poole, E., H e, B ., Lamb, R.A., Randall, R.E. & Goodb ourn, S. (2002) The V proteins of simian virus 5 and other Para- myxoviruses inhibit induction of interferon-beta. Virology 30 3, 33–46. 30. ten Hoeve, J., de Jesus Ibarra-Sanchez, M., Fu, Y., Zhu, W., Tremblay, M., D avid, M. & Shuai, K. (2002) Identification of a nuclear Stat1 protein tyrosine p hosp hatase. Mol. Cell. Biol. 22, 5662–5668. 31. Haspel, R.L. & Darnell, J.E. Jr ( 1999) A nuclear protein tyrosine phosphatase is required for the inactivatio n of Stat1. Proc. Natl Acad. Sci. USA 96, 10188–10193. 32. Haspel, R.L., Salditt-Georgieff, M . & Darnell, J.E. J r ( 1996) The rapid inactivation of nuclear tyrosine phosphorylated Stat1 depends on a protein tyrosine phosphatase. EMBO J. 15, 6262– 6268. 33. Lee, C.K., Bluyssen, H.A. & Levy, D.E. (1997) Regulation of interferon-alpha responsiveness by th e duration of J anus kinase activity. J. Biol. Chem. 272, 21872–21877. 34. Parisien, J P., Lau, J.F., Rodriguez, J.J., Sullivan, B.M., Mosc- ona, A., Parks, G.D., Lamb, R.A. & Horvath, C.M. (2001) The V protein o f human parainfluenza virus 2 antagonizes type I in ter- feron responses by destabilizing signal transducer and activator of transcription 2. Virology 283, 230–239. 35. Parisien, J.P., Lau, J.F. & Horvath, C.M. (2002) STAT2 acts as a host range determinant for spec ies-specific paramyxovirus interferon antagonism and simian virus 5 replication. J. Virol. 76, 6435–6441. 36. Parisien, J P., Lau, J.F., Rodriguez, J.J., Ulane, C.M. & Horvath, C.M. (2002) S elective STAT Prote in Degradation Induced b y Paramyxoviruses Requires both STAT1 and STAT2, but is Independent of alpha/beta Interferon Signal Transduction. J. Virol. 76, 4190–4198. 37. Kubota, T., Yokosawa, N., Yokota, S. & Fujii, N. (2001) C t erminal CYS-RICH region of mumps virus structural V pro- tein correlates with block of interferon alpha and gamma signal transduction pathway throu gh decrease o f STAT 1-alpha. Bio- chem. Biophys. Res. Commun. 283, 255–259. 38. Young, D.F., Didcock, L., Goodbourn, S . & Randall, R.E. (2000) Paramyxoviridae use distinct virus-specific mechanisms to cir- cumvent the interferon response. Virology 269, 383–390. 39. Yokosawa, N., Yokota, S., Kubota, T. & Fujii, N. (2002) C-Terminal Region of STAT-1al pha Is Not N e cessary for Its Ubiquitination and Degradation Caused by Mumps Virus V Protein. J. Virol. 76, 1 2683–12690. 40. Ulane, C.M., Rodriguez, J .J., Parisien,J.P.&Horvath,C.M. (2003) STAT3 ubiquitylation and degradation by mumps virus suppress c ytok ine and oncogene signaling. J. Virol. 77 , 6385–6393. 41. Ulane, C.M. & Horvath, C .M. (2002) Paramyxoviruses SV5 and HPIV2 Assemble STAT Protein Ubiquitin Ligase Complexes from Cellular Components. Virology 304, 1 60–166. 42. Nishio, M., Garcin, D., Simonet, V. & Kolakofsky, D. (2002) The carboxyl segment of the mumps virus V protein associates with Stat proteins in vitro via a tryptophan-rich motif. Virology 300, 92–99. 43. Andrejeva, J., Poole, E., Young, D.F., Goodbourn, S. & Randall, R.E. (2002) The p127 subunit (DDB1) of the UV-DNA damage repair binding protein is essential for the targete d degradation of STAT1 by the V p rotein of the paramyxovirus simian virus 5. J. Virol. 76, 11379–11386. 44. Lin, G.Y., Paterson, R.G., Richardson, C.D. & Lamb, R.A. (1998) The V protein o f the param yxovirus SV5 in teracts wit h damage-specific DNA binding protein. Virology 249, 189–200. 45. Didcock, L., Young, D .F., Goodbourn, S . & Randall, R.E. (1999) Sendai virus and sim ian virus 5 block a ctivation of i nterferon- responsive genes: importance for viru s pathogenesis. J. Virol. 73, 3125–3133. 46. Young, D.F., Chatziandreou, N., He, B., Go odbou rn, S., Lamb, R.A. & Randall, R.E. (2001) Single amino acid substitution in the V protein of simian virus 5 differentiates i ts ability to block interferon signaling in human and m urine cells. J. Virol. 75, 3363– 3370. 47. Farrar, J.D., S mith, J.D., Murphy, T .L., Leung, S., Stark, G.R. & Murphy, K.M. (2000) Selective loss of type 1 interferon induced STAT4 activation caused by a minisatellite insertion in mouse Stat2. Nat. Immunol. 1, 65–69. 48. Paulson, M., Pisharody, S., Pan, L., Guadagno, S., Mui, A.L. & Levy, D.E. (1999) Stat protein transactivation domains recruit p300/CBP through widely d ivergent se quences. J. Biol. Chem. 274, 25343–25349. 49. Park, C., Lecomte, M.J. & Schindler, C. (1999) Murine Stat2 is uncharacteristically divergent. Nucleic Acids Res. 27 , 4191–4199. 50. Park, M.S., Shaw, M.L., Munoz-Jordan, J ., Cros, J.F., N akaya, T.,Bouvier,N.,Palese,P.,Garcia-Sastre, A. & Basler, C.F. (2003) Newcastle Disease Virus (NDV)-Based Assay Demonstrates Interferon-Antagonist A ctivity f or th e NDV V Protein and the Nipah Virus V, W, and C Proteins. J. Virol. 77, 1501–1511. Ó FEBS 2004 Weapons of STAT destruction (Eur. J. Biochem. 271) 4627 51. Lam, S.K. (2003) Nipah virus – a potential agent of bioterrorism? Antiviral Res. 57, 113–119. 52. Chua, K.B., Bellini, W.J., Rota, P.A., Harcourt, B.H., Tamin, A., Lam, S.K., Ksiazek, T.G., Rollin, P.E., Zaki, S.R., Shieh, W., Goldsmith, C.S., Gubler, D.J., Roehrig, J.T., Eaton, B., Gould, A.R., Olson, J., Field, H., Daniels, P ., Ling, A.E., Peters, C.J., Anderson, L.J. & Mahy, B.W. (2000) Nipah virus: a recently emergent deadly paramyxovirus. Science 288, 1432–1435. 53. Rodriguez, J.J., Parisien, J.P. & Horvath, C.M. (2002) Nipah v irus V protein evades alpha and g amma interferons by p reventing STAT1 and STAT2 activation and nu clear accumulation. J. Virol. 76, 11476–11483. 54. Rodriguez, J.J., Wang, L.F. & Horvath, C.M. (2003) Hendra virus V protein inhibits interferon signalingbypreventingSTAT1and STAT2 nuclear accumulation. J. Virol. 77, 11842–11845. 55. Rodriguez, J.J., Cruz, C.D. & Horvath, C.M. (2004) Identification of the nuclear export signal and STAT-binding domains of the Nipah virus V protein reveals mechanisms underlying interfero n evasion. J. Virol. 78, 5358–5367. 56. Wang, L. & Harcourt, B.H., YuM., Tamin, A., Rota, P.A., Bel- lini, W.J. & Eaton, B.T. (2001) Molecular biology of Hendra and Nipah viruses. Microbes Infect. 3, 279–287. 57. Shaw, M.L., Garcia-Sastre, A., Palese, P. & Basler, C.F. (2004) Nipah virus V and W proteins have a common STAT1- binding d omain y et in hibit S TAT1 activation from t he c yto- plasmic and nuclear compartments, respectively. J. Virol. 78 , 5633–5641. 58. Palosaari, H., Parisien, J.P., Rodriguez, J.J., Ulane, C.M. & Horvath, C.M. (2003) STAT protein interference and suppression of cytokine sign al transduction by measle s virus V protein. J. Virol. 77, 7635–7644. 59. Yokota, S., Saito, H., K ubota, T., Y okosawa, N., Amano , K. & Fujii, N . (2003) Measles virus suppresses interferon-alpha signal- ing pathway: suppression of Jak1 phosphorylation and association of viral accessory proteins, C and V, w ith interferon-alpha receptor complex. Virology 306, 135–146. 60. Kato,A.,Kiyotani,K.,Sakai,Y.,Yoshida,T.&Nagai,Y.(1997) The paramyxovirus, Sendai virus, V p rotein encodes a l uxury function required for viral pathogenesis. EMBO J. 16, 578–587. 61. Gotoh, B., Takeuchi, K., Komatsu, T. & Yokoo, J. (2003) The STAT2 activation process is a crucial target of Sendai virus C protein for the blockade o f alpha in terferon signaling. J. Virol. 77 , 3360–3370. 62.Garcin,D.,Curran,J.,Itoh,M.&Kolakofsky,D.(2001) Longer and shorter forms of sendai virus c proteins play different roles in modulating the cellular antiviral response. J. Virol. 75, 6800–6807. 63. Garcin, D., Curran, J. & K olakofsk y, D. (2000) Sendai virus C proteins must interact directly with cellular co mponents to in ter- fere with interferon action. J. Virol. 74 , 8823–8830. 64. Garcin, D., Latorre, P . & Kolakofsky, D. (1999) Sendai virus C proteins counteract the i nterferon-me diated induction of a n anti- viral state. J. Virol. 73 , 6559–6565. 65. Takeuchi, K., Komatsu, T., Yokoo, J., Kato, A., Shioda, T., Nagai, Y. & Gotoh, B. (2001) Sendai virus C protein physically associates with Stat1. Genes Cells 6, 545–557. 66. Garcin,D.,Marq,J.B.,Goodbourn,S.&Kolakofsky,D.(2003) The a mino-terminal extensions of the longer Sendai virus C pro- teins modulate pY701-Stat1 and bulk Stat1 levels independently of interferon signaling. J. Virol. 77, 2321–2329. 67. Saito, S., Ogino, T., Miyajima, N., Kato, A. & Kohase, M . (2002) Dephosphorylation failure of tyrosine-phosphorylated STAT1 in IFN-stimulated Se ndai virus C protein-expressing cells. Viro logy 293, 205–209. 68. Garcin, D., M arq, J.B., Strahle, L., le Mercier, P. & Kolakofsky, D. (2002) All four Sendai Virus C proteins bind S tat1, but on ly the larger forms also i nduce its mono -ubiquitination and degradation. Virology 295, 256–265. 4628 C. M. Horvath (Eur. J. Biochem. 271) Ó FEBS 2004 . MINIREVIEW Weapons of STAT destruction Interferon evasion by paramyxovirus V proteins Curt M. Horvath Department of Medicine, Evanston Northwestern Healthcare Research Institute, and Departments of. to STAT- directed therapeutics for diseases characterized by STAT hyperactivity. Keywords: antiviral; interferon; paramyxovirus; STAT; viral evasion. Interferons: the antiviral cytokines The interferon. Disease Virus (NDV)-Based Assay Demonstrates Interferon- Antagonist A ctivity f or th e NDV V Protein and the Nipah Virus V, W, and C Proteins. J. Virol. 77, 1501–1511. Ó FEBS 2004 Weapons of STAT destruction

Ngày đăng: 30/03/2014, 15:20

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan