Citation: Mei-li LI, Hong GUO, Qiong DING, Chun-fu ZHENG. A Multiple Functional Protein: the Herpes Simplex Virus Type 1 Tegument Protein VP22* .VIROLOGICA SINICA, 2009, 24(3) : 153-161.  http://dx.doi.org/10.1007/s12250-009-3035-2

A Multiple Functional Protein: the Herpes Simplex Virus Type 1 Tegument Protein VP22*

  • Corresponding author: Chun-fu ZHENG, zhengcf@wh.iov.cn
  • Received Date: 19 February 2009
    Accepted Date: 08 April 2009
    Available online: 01 June 2009

    Fund Project: Open Research Fund Program of the State Key Laboratory of Virology of China 2009007The Startup Fund of the Hundred Talents Program of the Chinese Academy of Science 20071010141National Natural Science Foundation of China 30870120Open Research Fund Program of the State Key Laboratory of Virology of China 2007003

  • The herpes simplex virus type 1 (HSV-1) VP22, is one of the most abundant HSV-1 tegument proteins with an average stoichiometry of 2 400 copies per virion and conserved among alphaherpesvirinae. Many functions are attributed to VP22, including nuclear localization, chromatin binding, microtubule binding, induction of microtubule reorganization, intercellular transport, interaction with cellular proteins, such as template activating factor I (TAF-I) and nonmuscle myosin II A (NMIIA), and viral proteins including tegument protein VP16, pUS9 and pUL46, glycoprotein E (gE) and gD. Recently, many novel functions performed by the HSV-1 VP22 protein have been shown, including promotion of protein synthesis at late times in infection, accumulation of a subset of viral mRNAs at early times in infection and possible transcriptional regulation function.

  • 加载中
    1. Aints A, Guven H, Gahrton G, et al. 2001. Mapping of herpes simplex virus-1 vp22 functional domains for inter-and subcellular protein targeting. Gene Ther, 8 (14): 1051-1056.
        doi: 10.1038/sj.gt.3301493

    2. Beerens A M, Rots M G, de Vries E F, et al. 2007. Fusion of herpes simplex virus thymidine kinase to vp22 does not result in intercellular trafficking of the protein. Int J Mol Med, 19 (5): 841-849.

    3. Bian J, Kiedrowski M, Mal N, et al. 2006. Engineered cell therapy for sustained local myocardial delivery of nonsecreted proteins. Cell Transplant, 15 (1): 67-74.
        doi: 10.3727/000000006783982197

    4. Bian J, Popovic Z B, Benejam C, et al. 2007. Effect of cell-based intercellular delivery of transcription factor gata4 on ischemic cardiomyopathy. Circ Res, 100 (11): 1626-1633.
        doi: 10.1161/01.RES.0000269778.75877.68

    5. Brandimarti R, Roizman B. 1997. Us9, a stable lysine-less herpes simplex virus 1 protein, is ubiquitinated before packaging into virions and associates with proteasomes. Proc Natl Acad Sci USA, 94 (25): 13973-13978.
        doi: 10.1073/pnas.94.25.13973

    6. Brignati M J, Loomis J S, Wills J W, et al. 2003. Membrane association of vp 22, a herpes simplex virus type 1 tegument protein. J Virol, 77 (8): 4888-4898.
        doi: 10.1128/JVI.77.8.4888-4898.2003

    7. Duffy C, Mbong E F, Baines J D. 2009. Vp22 of herpes simplex virus 1 promotes protein synthesis at late times in infection and accumulation of a subset of viral mrnas at early times in infection. J Virol, 83 (2): 1009-1017.
        doi: 10.1128/JVI.02245-07

    8. Duffy C, Lavail J H, Tauscher A N, et al. 2006. Characterization of a ul49-null mutant: Vp22 of herpes simplex virus type 1 facilitates viral spread in cultured cells and the mouse cornea. J Virol, 80 (17): 8664-8675.
        doi: 10.1128/JVI.00498-06

    9. Elliott G, O'Hare P. 1997. Intercellular trafficking and protein delivery by a herpesvirus structural protein. Cell, 88 (2): 223-233.
        doi: 10.1016/S0092-8674(00)81843-7

    10. Elliott G, P. Hare O'. 1998. Herpes simplex virus type 1 tegument protein vp22 induces the stabilization and hyperacetylation of microtubules. J Virol, 72 (8): 6448-6455.

    11. Elliott G, O'Hare P. 1999. Intercellular trafficking of vp22-gfp fusion proteins. Gene Ther, 6 (1): 149-151.
        doi: 10.1038/sj.gt.3300850

    12. Elliott G, O'Hare P. 1999. Live-cell analysis of a green fluorescent protein-tagged herpes simplex virus infection. J Virol, 73 (5): 4110-4119.

    13. Elliott G, Mouzakitis G, O'Hare P. 1995. Vp16 interacts via its activation domain with vp22, a tegument protein of herpes simplex virus, and is relocated to a novel macromo-lecular assembly in coexpressing cells. J Virol, 69 (12): 7932-7941.

    14. Elliott G, O'Reilly D, O'Hare P. 1999. Identification of phosphorylation sites within the herpes simplex virus tegument protein vp22. J Virol, 73 (7): 6203-6206.

    15. Elliott G, Hafezi W, Whiteley A, et al. 2005. Deletion of the herpes simplex virus vp22-encoding gene (ul49) alters the expression, localization, and virion incorporation of icp0. J Virol, 79 (15): 9735-9745.
        doi: 10.1128/JVI.79.15.9735-9745.2005

    16. Elliott G D, Meredith D M. 1992. The herpes simplex virus type 1 tegument protein vp22 is encoded by gene ul49. J Gen Virol, 73 (Pt 3): 723-726.

    17. Harms J S, Ren X, Oliveira S C, et al. 2000. Distinctions between bovine herpesvirus 1 and herpes simplex virus type 1 vp22 tegument protein subcellular associations. J Virol, 74 (7): 3301-3312.
        doi: 10.1128/JVI.74.7.3301-3312.2000

    18. Heine J W, Honess R W, Cassai E, et al. 1974. Proteins specified by herpes simplex virus. Xii. The virion poly-peptides of type 1 strains. J Virol, 14 (3): 640-651.

    19. Hutchinson I, Whiteley A, Browne H, et al. 2002. Sequential localization of two herpes simplex virus tegu-ment proteins to punctate nuclear dots adjacent to icp0 domains. J Virol, 76 (20): 10365-10373.
        doi: 10.1128/JVI.76.20.10365-10373.2002

    20. Kim T W, Hung C F, Kim J W, et al. 2004. Vaccination with a DNA vaccine encoding herpes simplex virus type 1 vp22 linked to antigen generates long-term antigen-specific cd8-positive memory t cells and protective immunity. Hum Gene Ther, 15 (2): 167-177.
        doi: 10.1089/104303404772679977

    21. Kotsakis A, Pomeranz L E, Blouin A, et al. 2001. Microtubule reorganization during herpes simplex virus type 1 infection facilitates the nuclear localization of vp22, a major virion tegument protein. J Virol, 75 (18): 8697-8711.
        doi: 10.1128/JVI.75.18.8697-8711.2001

    22. LaVail J H, Tauscher A N, Sucher A, et al. 2007. Viral regulation of the long distance axonal transport of herpes simplex virus nucleocapsid. Neuroscience, 146 (3): 974-985.
        doi: 10.1016/j.neuroscience.2007.02.010

    23. Lee J H, Vittone V, Diefenbach E, et al. 2008. Identification of structural protein-protein interactions of herpes simplex virus type 1. Virology, 378 (2): 347-354.
        doi: 10.1016/j.virol.2008.05.035

    24. Lemken M L, Graepler F, Wolf C, et al. 2007. Fusion of hsv-1 vp22 to a bifunctional chimeric supercd suicide gene compensates for low suicide gene transduction efficiencies. Int J Oncol, 30 (5): 1153-1161.

    25. Lemken M L, Wolf C, Wybranietz W A, et al. 2007. Evidence for intercellular trafficking of vp22 in living cells. Mol Ther, 15 (2): 310-319.
        doi: 10.1038/sj.mt.6300013

    26. Lopez M R, Schlegel E F, Wintersteller S, et al. 2008. The major tegument structural protein vp22 targets areas of dispersed nucleolin and marginalized chromatin during productive herpes simplex virus 1 infection. Virus Res, 136 (1-2): 175-188.
        doi: 10.1016/j.virusres.2008.05.010

    27. Lundberg M, Johansson M. 2001. Is vp22 nuclear homing an artifact? Nat Biotechnol, 19 (8): 713-714.
        doi: 10.1038/90741

    28. Martin A, O'Hare P, McLauchlan J, et al. 2002. Herpes simplex virus tegument protein vp22 contains overlapping domains for cytoplasmic localization, microtubule interaction, and chromatin binding. J Virol, 76 (10): 4961-4970.
        doi: 10.1128/JVI.76.10.4961-4970.2002

    29. Mettenleiter T C. 2002. Herpesvirus assembly and egress. J Virol, 76 (4): 1537-1547.
        doi: 10.1128/JVI.76.4.1537-1547.2002

    30. Miranda-Saksena M, Boadle R A, Armati P, et al. 2002. In rat dorsal root ganglion neurons, herpes simplex virus type 1 tegument forms in the cytoplasm of the cell body. J Virol, 76 (19): 9934-9951.
        doi: 10.1128/JVI.76.19.9934-9951.2002

    31. Miyaji-Yamaguchi M, Okuwaki M, Nagata K. 1999. Coiled-coil structure-mediated dimerization of template activating factor-i is critical for its chromatin remodeling activity. J Mol Biol, 290 (2): 547-557.
        doi: 10.1006/jmbi.1999.2898

    32. Mori T, Mineta Y, Aoyama Y, et al. 2008. Efficient secretion of the herpes simplex virus tegument protein vp22 from living mammalian cells. Arch Virol, 153 (6): 1191-1195.
        doi: 10.1007/s00705-008-0094-x

    33. Mouzakitis G, McLauchlan J, Barreca C, et al. 2005. Characterization of vp22 in herpes simplex virus-infected cells. J Virol, 79 (19): 12185-12198.
        doi: 10.1128/JVI.79.19.12185-12198.2005

    34. Murphy M A, Bucks M A, O'Regan K J, et al. 2008. The hsv-1 tegument protein pul46 associates with cellular membranes and viral capsids. Virology, 376 (2): 279-289.
        doi: 10.1016/j.virol.2008.03.018

    35. Perkins S D, Hartley M G, Lukaszewski R A, et al. 2005. Vp22 enhances antibody responses from DNA vaccines but not by intercellular spread. Vaccine, 23 (16): 1931-1940.
        doi: 10.1016/j.vaccine.2004.10.033

    36. Pomeranz L E, Blaho J A. 1999. Modified vp22 localizes to the cell nucleus during synchronized herpes simplex virus type 1 infection. J Virol, 73 (8): 6769-6781.

    37. Pomeranz L E, Blaho J A. 2000. Assembly of infectious herpes simplex virus type 1 virions in the absence of full-length vp22. J Virol, 74 (21): 10041-10054.
        doi: 10.1128/JVI.74.21.10041-10054.2000

    38. Posnett D N, Engelhorn M E, Lin Y, et al. 2009. Develop-ment of effective vaccines for old mice in a tumor model. Vaccine, 27 (7): 1093-1100.
        doi: 10.1016/j.vaccine.2008.11.112

    39. Potel C, Elliott G. 2005. Phosphorylation of the herpes simplex virus tegument protein vp22 has no effect on incorporation of vp22 into the virus but is involved in optimal expression and virion packaging of icp0. J Virol, 79 (22): 14057-14068.
        doi: 10.1128/JVI.79.22.14057-14068.2005

    40. Rutjes S A, Bosma P J, Rohn J L, et al. 2003. Induction of insolubility by herpes simplex virus vp22 precludes intercellular trafficking of n-terminal apoptin-vp22 fusion proteins. J Mol Med, 81 (9): 558-565.
        doi: 10.1007/s00109-003-0457-4

    41. Saha S, Yoshida S, Ohba K, et al. 2006. A fused gene of nucleoprotein (np) and herpes simplex virus genes (vp22) induces highly protective immunity against different subtypes of influenza virus. Virology, 354 (1): 48-57.
        doi: 10.1016/j.virol.2006.04.015

    42. Schwarze S R, Hruska K A, Dowdy S F. 2000. Protein transduction: Unrestricted delivery into all cells? Trends Cell Biol, 10 (7): 290-295.
        doi: 10.1016/S0962-8924(00)01771-2

    43. Sciortino M T, Taddeo B, Poon A P, et al. 2002. Of the three tegument proteins that package mrna in herpes simplex virions, one (vp22) transports the mrna to unin-fected cells for expression prior to viral infection. Proc Natl Acad Sci U S A, 99 (12): 8318-8323.
        doi: 10.1073/pnas.122231699

    44. Sciortino M T, Taddeo B, Giuffre-Cuculletto M, et al. 2007. Replication-competent herpes simplex virus 1 iso-lates selected from cells transfected with a bacterial artificial chromosome DNA lacking only the ul49 gene vary with respect to the defect in the ul41 gene encoding host shutoff rnase. J Virol, 81 (20): 10924-10932.
        doi: 10.1128/JVI.01239-07

    45. Sellers J R. 2000. Myosins: A diverse superfamily. Biochim Biophys Acta, 1496 (1): 3-22.
        doi: 10.1016/S0167-4889(00)00005-7

    46. Seo S B, McNamara P, Heo S, et al. 2001. Regulation of histone acetylation and transcription by inhat, a human cellular complex containing the set oncoprotein. Cell, 104 (1): 119-130.
        doi: 10.1016/S0092-8674(01)00196-9

    47. Stroh C, Held J, Samraj A K, et al. 2003. Specific inhibition of transcription factor nf-kappab through in-tracellular protein delivery of i kappabalpha by the herpes virus protein vp22. Oncogene, 22 (34): 5367-5373.
        doi: 10.1038/sj.onc.1206544

    48. Taddeo B, Sciortino M T, Zhang W, et al. 2007. Interaction of herpes simplex virus rnase with vp16 and vp22 is required for the accumulation of the protein but not for accumulation of mrna. Proc Natl Acad Sci USA, 104 (29): 12163-12168.
        doi: 10.1073/pnas.0705245104

    49. van Leeuwen H, Elliott G, O'Hare P. 2002. Evidence of a role for nonmuscle myosin ii in herpes simplex virus type 1 egress. J Virol, 76 (7): 3471-3481.
        doi: 10.1128/JVI.76.7.3471-3481.2002

    50. van Leeuwen H, Okuwaki M, Hong R, et al. 2003. Herpes simplex virus type 1 tegument protein vp22 in-teracts with taf-i proteins and inhibits nucleosome assem-bly but not regulation of histone acetylation by inhat. J Gen Virol, 84 (Pt 9): 2501-2510.

    51. Vittone V, Diefenbach E, Triffett D, et al. 2005. Determination of interactions between tegument proteins of herpes simplex virus type 1. J Virol, 79 (15): 9566-9571.
        doi: 10.1128/JVI.79.15.9566-9571.2005

    52. Wybranietz W A, Prinz F, Spiegel M, et al. 1999. Quantification of vp22-gfp spread by direct fluorescence in 15 commonly used cell lines. J Gene Med, 1 (4): 265-274.
        doi: 10.1002/(ISSN)1521-2254

    53. Xiong F, Xiao S, Yu M, et al. 2007. Enhanced effect of microdystrophin gene transfection by hsv-vp22 mediated intercellular protein transport. BMC Neurosci, 8: 50.
        doi: 10.1186/1471-2202-8-50

    54. Xiong F, Xiao S, Peng F, et al. 2007. Herpes simplex virus vp22 enhances adenovirus-mediated microdystrophin gene transfer to skeletal muscles in dystrophin-deficient (mdx) mice. Hum Gene Ther, 18 (6): 490-501.
        doi: 10.1089/hum.2006.155

    55. Yu X, Li W, Liu L, et al. 2008. Functional analysis of transcriptional regulation of herpes simplex virus type 1 tegument protein vp22. Sci China C Life Sci, 51 (11): 966-972.
        doi: 10.1007/s11427-008-0127-4

    56. Zhang Y, Sirko D A, McKnight J L. 1991. Role of herpes simplex virus type 1 ul46 and ul47 in alpha tif-mediated transcriptional induction: Characterization of three viral deletion mutants. J Virol, 65 (2): 829-841.

  • 加载中

Article Metrics

Article views(4601) PDF downloads(16) Cited by()

Related
Proportional views

    A Multiple Functional Protein: the Herpes Simplex Virus Type 1 Tegument Protein VP22*

      Corresponding author: Chun-fu ZHENG, zhengcf@wh.iov.cn
    • State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
    Fund Project:  Open Research Fund Program of the State Key Laboratory of Virology of China 2009007The Startup Fund of the Hundred Talents Program of the Chinese Academy of Science 20071010141National Natural Science Foundation of China 30870120Open Research Fund Program of the State Key Laboratory of Virology of China 2007003

    Abstract: The herpes simplex virus type 1 (HSV-1) VP22, is one of the most abundant HSV-1 tegument proteins with an average stoichiometry of 2 400 copies per virion and conserved among alphaherpesvirinae. Many functions are attributed to VP22, including nuclear localization, chromatin binding, microtubule binding, induction of microtubule reorganization, intercellular transport, interaction with cellular proteins, such as template activating factor I (TAF-I) and nonmuscle myosin II A (NMIIA), and viral proteins including tegument protein VP16, pUS9 and pUL46, glycoprotein E (gE) and gD. Recently, many novel functions performed by the HSV-1 VP22 protein have been shown, including promotion of protein synthesis at late times in infection, accumulation of a subset of viral mRNAs at early times in infection and possible transcriptional regulation function.

    • Human herpes simplex virus type 1 (HSV-1), a nuclear replicating DNA virus, is a widespread human pathogen that causes a lytic infection in the mucosal epithelial cells and a life-long latent infection in neurons. HSV-1 has a distinctive feature of having more than 20 tegument proteins between its capsid shell and envelope (29). Tegument proteins have been shown to play a variety of roles in infection including the regulation of viral and host gene expression and the promotion of virus assembly and egress. An essential tegument protein, VP22, the product of the UL49 gene of HSV-1, has received notable attention for its large number of molecules in each polypeptide per virion (2400 molecules/virion) (18). It is a highly basic, rich in proline residues, nuclear phosphoprotein (14, 16, 19), capable of forming higher-order structures consisting of dimers or tetramers (33). VP22 contains 301 amino acids and has a predicted molecular weight of 32 kDa (16). It is conserved within the alphaherpesvirus family, and undergoes a number of posttranslational modifications during infection, including nucleotidylation, ADP-ribosylation and phosphorylation by at least two cellular kinases, casein kinase Ⅱ and protein kinase C (PKC), and one virion protein kinase UL13 (36).

      A number of functions have been attributed to VP22 and these functions are determined by different regions of this protein (1). VP22 possesses at least two separate determinants of nuclear and nucleolar localization (1, 21) and the protein accumulates to high levels in the nuclei of either infected cells (36) or stable VP22 expression cell lines (37). In cells, VP22 is first localized in the cytoplasm, where it binds to microtubules and induces collapse of the cytoskeleton, creating a large perinuclear lump. After the nuclear membrane has disintegrated in early mitosis, VP22 translocates into the nucleus and binds to chromatin, where it remains until after completion of mitosis and during the next interphase (1, 28). A recent study showed that nuclear VP22 targeted unique subnuclear structures early ( < 6 hpi) during HSV-1 infection and reduced the number of nonring nucleolin patterns that seemed to imply that VP22 might function to inhibit nucleolar disassembly, thus stabilizing these structures (26). A mutant HSV lacking VP22 was able to assemble enveloped virions that reached extracellular compartments. This mutant produced virions with reduced immediate-early proteins (ICP0 and ICP4) and displayed delayed viral protein synthesis (15). Another VP22 null mutant described recently displayed defects in reaching extracellular compartments and in cell-to-cell spread (8). These two VP22 null viruses also decreased packaging of both glycoproteins gD and gE (8, 15). Recently, it was reported that the expressions of ICP0, gD and gE were regulated by the level of phosphate presented on VP22 (39). VP22 interacts with tegument protein VP16, α-trans-inducing factor (α-TIF), and relocates it to a novel macromolecular assembly in coexpressing cells (13). Thus, VP22 plays some role in the final assembly of HSV, although likely in a redundant fashion with other tegument proteins (37). VP22 also can incorporate the mRNA into the virion and transport the mRNA to uninfected cells (43). Another important role of VP22 is that it can interact with template activating factor Ⅰ (TAF-Ⅰ) and impair the nucleosome assembly on entering viral DNAs (50).

      Moreover, VP22 has the capability to spread intercellularly to adjacent cells after infection or transfection solely with a plasmid encoding VP22 (8, 9). This property is maintained in proteins fused with VP22, which can be used to transfer potentially therapeutic proteins into target cells. Furthermore, recent reports showed that VP22 was involved in promotion of protein synthesis, accumulation of a subset of viral mRNAs (7), and in transcriptional regulation (55). All of those studies imply that VP22 is important to virus replication. However, the detailed roles of VP22 during HSV-1 infection remain unclear. Some of its functions are described below.

    • VP22 is an efficient intercellular transporter, able to transfer into neighboring cells and translocate to the nucleus of the target cell (9). After expression in transfection cells, VP22 protein is exported from transfected cells via a non-classical Golgi-independent pathway. After secretion, VP22 can be efficiently internalized into the neighboring non-transfected cells by a poorly understood mechanism. Indeed, VP22 has been used as a cell-penetrating peptide/protein (CPP) (42). Many experiments have demonstrated that VP22 could enhance intercellular trafficking of proteins fused to its N-or C-terminus in vitro and in vivo (17, 52, 53), and trafficking of VP22 was not restricted to particular tissues and species (17, 52). Moreover, VP22-fusion proteins retained both VP22-mediated transport properties and the biophysiological functions of the linked passenger proteins, enhancing the therapeutic effects of transduced genes and proteins. This property has been observed with VP22-linked p27, p53, CD (cytosine deaminase), SuperCD, which is the fusion protein of yeast CD with yeast uracilphosphoribosyltransferase, and human papillomavirus E2 for delivery to tumors (24, 25). The property was also found in the fusion protein of VP22 with green fluorescent protein (VP22-GFP) following adenovirusmediated transgene delivery to the central nervous system (CNS) neurons and others have also reported that VP22-GFP has the property of intercellular trafficking (9, 11, 25). Furthermore, VP22 mediated intercellular trafficking of IkappaB-alpha can efficiently inhibit both constitutive and inducible nuclear factor kappa B (NF-kappaB) activation (47). VP22-mediated intercellular protein delivery is also useful in enhancing the efficacy of myocardial gene therapy (3, 4, 53) and the effects of gene delivery for somatic gene therapy of Duchenne muscular dystrophy (DMD) (53, 54). The fusion gene of VP22–β-galactosidase (βGal) transfection resulted in higher amounts of protein expression compared with βGal transfection, and this study showed that VP22 had a remarkable secretory ability in living cells (32). Preclinical evaluation using the thymidine kinase (TK)-VP22 fusion protein revealed an enhanced cytotoxicity in vitro and in vivo using mixed populations of wildtype and expressing cells (53). However, some authors reported that fusion of HSV TK to VP22 did not result in intercellular trafficking of the protein (2). In fact, others could not detect VP22-GFP trafficking in living cells, only in fixed cells. So it was hypothesized that this difference resulted from a concentration effect or removal of interfering components (11, 17), and it was even suggested that VP22 nuclear homing in living cells was an experimental artifact (27). Therefore, existing studies of VP22 intercellular spread remain confusing and difficult to interpret, but it should be pointed out that some proteins are readily extracted during methanol fixation of cells and that covalently bound polypeptides of a size nearly equivalent to or larger than VP22 could be expected to change the properties of the protein (44). However, Rutjes pointed out that VP22 could have a negative effect on the solubility of some fusion proteins, which consequently precluded intercellular trafficking (40).

      Although it was reported that amplification of the immune response might occur via mechanisms other than VP22-mediated intercellular spread of antigen (35), VP22 could nevertheless be used to augment the potency of DNA vaccines (20, 38, 41). Moreover, it has been shown that VP22 could transport the mRNA to uninfected cells (43). All of these findings suggest that the VP22-mediated protein delivery system can be a useful and potential tool to enhance the efficacy of gene therapy.

    • Previous studies of a UL49 deletion mutant showed that VP22 greatly enhanced plaque size and viral replication at low multiplicities of infection (8, 37). UL49 deletion virions also contained decreased amounts of ICP0, gE and gD (8, 15). A recent investigation, aiming to determine whether the latter phenomenon was due to decreased virion incorporation or decreased synthesis of ICP0, gE and gD in the absence of VP22, showed that VP22 was required for optimal protein synthesis at late times in infection and the accumulation of at least gE, gD and virion host shutoff (vhs) mRNAs at early times in infection (7). Interestingly, VP22 effects on protein synthesis and mRNA levels are distinct and separable with regards to both timing during infection and the genes affected (7). VP22 has also been shown to interact with mRNA and is found in both the nucleus and cytoplasm pointing to a possible direct role for VP22 in mRNA export and/or translation. VP22 interacts with tegument protein VP16, a component of primary enveloped virions, which is also a potent transcriptional activator of viral immediate-early (IE) genes and can recruit various transcriptional coactivators to target gene promoters (13). In addition, the VP16/VP22 complex can interact with the UL41-encoded protein vhs which is a riboendonuclease that specifically cleaves host and viral mRNAs (48). So VP22 may promote protein synthesis and contribute to mRNA accumulation by enhancing the effect of VP16 or modulating the RNase activity of vhs. The details of the mechanisms involved in protein synthesis and accumulation of mRNAs require further investigation.

    • VP22 interacts with VP16 and enhances the effect of VP16 (13). VP22 locates to the cell nucleus where it binds to chromatin (1, 28) and it also specifically inhibits nucleosome assembly by binding to TAF-1 (50). In addition, VP22 has a special relationship with the important viral transcriptional regulatory factor ICP0 (15). Previous studies on VP22 indicated that it was highly phosphorylated during HSV-1 infection (36), it is correlated in its function to certain links during virion assembly (8, 15), and has the capacity to stabilize the microtubule (10, 21). All these studies showed that VP22 might have some transcriptional regulation function. A recent paper demonstrated that VP22 alone in the cellular environment inhibited transcriptional activation of different viral transcriptional regulatory factors on different viral gene promoters (55). Perhaps VP22 performs these functions by interacting with viral or cellular proteins. However whether VP22 has the same transcriptional regulation function during virus infection is still unknown.

    • The exact roles of VP22 in the virus life cycle remain poorly understood. However, it is likely that various dynamic interactions of VP22 with different cellular proteins and/or viral proteins are required.

      VP22 exhibits the properties of a classical microtubuleassociated protein, reorganizing and stabilizing the host cell microtubule network and hyperacetylating these stabilized microtubules (MTs) (10, 21). While the role of the VP22-microtubule interaction during infection has yet to be determined, it would seem that phosphorylation may play an important role in ensuring that this interaction is not dominant over the other activities of VP22 during infection (39) and microtubule reorganization may function as a mecha-nism to regulate VP22 nuclear localization (21). Microtubule reorganization is important for viral exocytosis and for transport of HSV-1 capsids to the nucleus (17). In addition, VP22 interacts with non-muscle myosin IIA (NMIIA) (49), a motor protein which is recruited to the actin cytoskeleton and has been reported to be involved in numerous dynamic cellular processes, including Golgi budding, vesicle secretion, cell spreading, cleavage and migration, and cell adhesion (45). The VP22-myosin interaction provides evidence for an interaction with a network of myosinactin filaments other than the microtubule/ dynein system. NMIIA may be involved in virus maturation (49). The interaction between VP22 and MT/NMIIA could be relevant to assembly and egress, and could play a role in virus transport. Further work will be necessary to identify the exact role of the interactions.

      A previous study showed that VP22 interacted with a cellular histone chaperone TAF-Ⅰ (50), which has been shown to promote more ordered transfer of histones to naked DNA through a direct interaction with histones (31). TAF-Ⅰ, as a subunit of the INHAT (inhibitor of acetyltransferases) protein complex, also binds to histones and masks them from acting as substrates for the acetyltransferases p300 and PCAF (46). The study further showed that interaction with VP22 inhibited the activity of TAF-Ⅰα in chromatin assembly, while, conversely, overexpression of TAF-Ⅰα suppressed HSV infection in vivo. VP22-binding proteins may form an INHAT complex pp32/PHAPI that could be relevant to HSV DNA organization early in infection (50). But other functions such as transcriptional regulation cannot be excluded and further work is necessary to prove this interaction physiologically.

      VP22 interacts with membranes and is also believed to possess the ability to multimerize, and more specifically with membranes of the acidic compart-ments of cells that may include the trans-Golgi network (TGN) (6, 33). A previous study of VP22, which used livecell fluorescence during virus infection, demonstrated a punctate cytoplasmic localization reminiscent of the Golgi apparatus (12). Current evidence supports a model that final tegumentation and envelopment of herpesviruses occur when nucleocapsids bud into cytoplasmic vesicles, which may be derived from the TGN (6). Consistent with this observation, VP22 is packaged into virions during final envelopment as nucleocapsids bud into TGN-derived vesicles (30). VP22 can interact with the cytoplasmic tails of gD and gE, and may facilitate the interaction of viral nucleo-capsids with glycoproteins lining up on the membranes of TGN-derived vesicles, perhaps also through its interaction with VP16 (62). The interaction between VP22 and VP16, gD or gE/gI may promote secondary envelopment.

      A novel interaction between VP22 and the US9 protein (pUS9) has been screened by yeast two-hybrid recently (23). pUS9 of HSV-1 is a tegument protein and the US9 gene is highly conserved among the members of the alpha subfamily of herpesviruses (5). More recent studies indicated that pUS9 is associated with the endoplasmic reticulum and Golgi apparatus, as well as with cytoplasmic, unenveloped capsids. On the other hand, a detailed study of the pseudorabies virus (PRV) demonstrated that pUS9 of PRV was a type Ⅱ membrane protein associated with Golgi or perinuclear membranes (22). Although the US9 gene is dispensable for viral replication in cultured cells, it is believed that pUS9 is a lysineless ubiquitinated protein that interacts with the ubiquitin dependent pathway for degradation of proteins. This function may be initiated at the time of entry of the virus into the cell (5). Arecent observation showed that pUS9 was necessary for long distance anterograde axonal transport of viral nucleocapsids (22). It could represent an additional viral mechanism to regulate specific cellular and virus functions. This interaction is likely to be involved in the process of secondary envelopment during viral assembly. However, whether this is a structural or regulatory interaction requires further investigation.

      pUL46 (VP11/12) is another novel protein which interacts with VP22 (23), it is an abundant virion tegument phosphoprotein with approximately 1200 molecules per virion (18). pUL46 and VP22 have similar localization and both are membrane-associated proteins, however, pUL46 binds the membrane less strongly than VP22 (34). It has been reported that the HSV-1 DNA fragment containing the UL46 gene enhanced the efficiency of VP16-mediated gene expression (56). Also pertinent to this report is the observation that VP22 interacts with VP16 (13); it was reported that pUL46 could also interact with VP16 (51). The interactions among these three tegument proteins indicate that they may simultaneously play some role in the replication of the virus. It seems that pUL46 is an inner layer of the tegument protein which associates directly with capsids, while VP16 and VP22 belong to the peripheral layer composed of proteins that interact with the envelope. The mechanism of interaction between VP22 and pUL46 still needs further study.

    • Herpesviruses are highly disseminated in nature. HSV-1 is one of the most intensively investigated herpesviruses which serves as a model and a tool for a wide range of applications including the study of translocation of proteins, membrane structure, gene regulation, gene therapy and cancer therapy. VP22, the essential tegument protein of HSV-1, is conserved within the alpha-herpesvirus family, and has multiple functions in virus replication and spread.

      VP22 has intercellular trafficking properties. It is necessary for localization of ICP0 to putative sites of viral assembly within the cytoplasm and can also package RNA into the virion. Recently, many novel functions attributed to the HSV-1 VP22 protein were shown, including promotion of protein synthesis at late times in infection, accumulation of a subset of viral mRNAs at early times in infection and a possible transcriptional inhibition function in some genes expression.

      VP22 can accumulate in the nucleus as well as exploit the host cytoskeleton suggesting that this unusual tegument protein may have an important role in herpesvirus infection, replication and pathogenesis. Although the exact molecular mechanisms are still not clearly understood, exploring the mechanism of how VP22 is imported to the nucleus may advance the knowledge of the pathogenesis of the herpesviruses and enable the development of new antiviral approaches.

      VP22 can interact with at least five virus proteins (VP16, gE, gD, pUS9 and pUL46) and at least three cellular proteins (TAF-Ⅰ, NMIIA and MT), and plays different roles according to these different interactions. From these reports we can conclude that the interactions of VP22 with different cellular proteins and/ or virus proteins are important for its functionality. So it is necessary to establish the network of protein (VP22)-protein (host/virus) interactions that facilitate further definition of the potential roles and possible mechanism of VP22. Understanding the mechanism of the multifunctional regulatory protein VP22 may advance our knowledge of the HSV-1 and promote the exploitation of efficient methods for preventing viral infection and antiviral therapy.

    Reference (56) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return