Citation: Lei ZHAO, Wen-bo ZHU, Qiong DING, Gui-qing PENG, Chun-fu ZHENG. The Herpes Simplex Virus Type 1 Multiple Function Protein ICP27 .VIROLOGICA SINICA, 2008, 23(6) : 399-405.  http://dx.doi.org/10.1007/s12250-008-2993-0

The Herpes Simplex Virus Type 1 Multiple Function Protein ICP27

  • Corresponding author: Chun-fu ZHENG, zhengcf@wh.iov.cn
  • Received Date: 18 May 2008
    Accepted Date: 28 September 2008
    Available online: 01 December 2008

    Fund Project: National Natural Science Foundation of China 30870120Start Fund of the Hundred Talents Program of the Chinese Academy of Science 20071010-141Open Research Fund Program of the State Key Laboratory of Virology of China 2007003

  • The herpes simplex virus type 1 (HSV-1) infected-cell protein 27 (ICP27) is an essential, highly conserved protein involved in various steps of HSV-1 gene regulation as well as in the shut-off of host gene expression during infection. It functions primarily at the post-transcriptional level in inhibiting precursor mRNA splicing and in promoting nuclear export of viral transcripts. Recently, many novel functions performed by the HSV-1 ICP27 protein were shown, including leptomycin B resistance, inhibition of the typeⅠinterferon signaling, regulation of the viral mRNA translation and determining the composition of HSV-1 virions.

  • 加载中
    1. Boyne J R, Colgan K J, Whitehouse A. 2008. Herpesvirus saimiri ORF57: a post-transcriptional regulatory protein. Front Biosci, 13: 2928-2938.
        doi: 10.2741/2898

    2. Brown C R, Nakamura M S, Mosca J D, et al. 1995. Herpes simplex virus trans-regulatory protein ICP27 stabilizes and binds to 3' ends of labile mRNA. J Virol, 69 (11): 7187-7195.

    3. Chen I B, Li L, Silva L, et al. 2005. ICP27 recruits Aly/REF but not TAP/NXF1 to herpes simplex virus type 1 transcription sites although TAP/NXF1 is required for ICP27 export. J Virol, 79 (7): 3949-3961.
        doi: 10.1128/JVI.79.7.3949-3961.2005

    4. Chen I B, Sciabica K S, Sandri-Goldin R M. 2002. ICP27 interacts with the RNA export factor Aly/REF to direct herpes simplex virus type 1 intronless mRNAs to the TAP export pathway. J Virol, 76 (24): 12877-12889.
        doi: 10.1128/JVI.76.24.12877-12889.2002

    5. Coller J M, Gray N K, Wickens M P. 1998. mRNA stabilization by poly (A)-binding protein is independent of poly (A) and requires translation. Genes Dev, 12 (20): 3226-3235.
        doi: 10.1101/gad.12.20.3226

    6. Dai-Ju J Q, Li L, Johnson L A, et al.2006. ICP27 Interacts with the C-terminal domain of RNA polymerase Ⅱ and facilitates its recruitment to herpes simplex virus 1 transcription sites, where it undergoes proteasomal deg-radation during infection. J Virol, 80 (7): 3567-3581.
        doi: 10.1128/JVI.80.7.3567-3581.2006

    7. Dai-Ju J Q, Li L, Johnson L A, et al.2006. ICP27 interacts with the C-terminal domain of RNA polymerase Ⅱ and facilitates its recruitment to herpes simplex virus 1 transcription sites, where it undergoes proteasomal degradation during infection. J Virol, 80 (7): 3567-3581.
        doi: 10.1128/JVI.80.7.3567-3581.2006

    8. Dimaano C, Ullman K S. 2004. Nucleocytoplasmic transport: integrating mRNA production and turnover with export through the nuclear pore. Mol Cell Biol, 24 (8): 3069-3076.
        doi: 10.1128/MCB.24.8.3069-3076.2004

    9. Ellison K S, Maranchuk R A, Mottet K L, et al.2005. C Control of VP16 translation by the herpes simplex virus type 1 immediate-early protein ICP27. J Virol, 79 (7): 4120-4131.
        doi: 10.1128/JVI.79.7.4120-4131.2005

    10. Fontaine-Rodriguez E C, Knipe D M. 2008. Herpes simplex virus ICP27 increases translation of a subset of viral late mRNAs. J Virol, 82 (7): 3538-3545.
        doi: 10.1128/JVI.02395-07

    11. Fontaine-Rodriguez E C, Taylor T J, Olesky M, et al. 2004. Proteomics of herpes simplex virus infected cell protein 27: association with translation initiation factors. Virology, 330 (2): 487-492.
        doi: 10.1016/j.virol.2004.10.002

    12. Garcia-Sastre A, Biron C A. 2006. Type 1 interferons and the virus-host relationship: a lesson in detente. Science, 312 (5775): 879-882.
        doi: 10.1126/science.1125676

    13. Gray N K, Coller J M, Dickson K S, et al. 2000. Multiple portions of poly(A)-binding protein stimulate translation in vivo. EMBO J, 19 (17): 4723-4733.
        doi: 10.1093/emboj/19.17.4723

    14. Halford W P, Weisend C, Grace J, et al. 2006. ICP0 antagonizes Stat 1-dependent repression of herpes simplex virus: implications for the regulation of viral latency. Virol J, 3 (1): 44.
        doi: 10.1186/1743-422X-3-44

    15. Hardy W R, Sandri-Goldin R M. 1994. Herpes simplex virus inhibits host cell splicing, and regulatory protein ICP27 is required for this effect. J Virol, 68 (12): 7790-7799.

    16. Hiriart E, Farjot G, Gruffat H, et al.2003. A novel nuclear export signal and a REF interaction domain both promote mRNA export by the Epstein-Barr virus EB2 protein. J Biol Chem, 278 (1): 335-342.
        doi: 10.1074/jbc.M208656200

    17. Jin L, Guzik B W, Bor Y C, et al. 2003. Tap and NXT promote translation of unspliced RNA. Genes Dev, 17 (24): 3075-3086.
        doi: 10.1101/gad.1155703

    18. Johnson K E, Song B, Knipe D M. 2008. Role for herpes simplex virus 1 ICP27 in the inhibition of type Ⅰ interferon signaling. Virology, 374 (2): 487-394.
        doi: 10.1016/j.virol.2008.01.001

    19. Koffa M D, Clements J B, Izaurralde E, et al. 2001. Herpes simplex virus ICP27 protein provides viral mRNAs with access to the cellular mRNA export pathway. EMBO J, 20 (20):5769-5778.
        doi: 10.1093/emboj/20.20.5769

    20. Komatsu T, Takeuchi K, Yokoo J, et al.2000. Sendai virus blocks alpha interferon signaling to signal transducers and activators of transcription. J Virol, 74 (5): 2477-2480.
        doi: 10.1128/JVI.74.5.2477-2480.2000

    21. Larralde O, Smith R W, Wilkie G S, et al. 2006. Direct stimulation of translation by the multifunctional herpes-virus ICP27 Protein. J Virol, 80 (3): 1588-1591.
        doi: 10.1128/JVI.80.3.1588-1591.2006

    22. Lengyel J, Strain A K, Perkins K D, et al. 2006. ICP27-dependent resistance of herpes simplex virus type 1 to leptomycin B is associated with enhanced nuclear localization of ICP4 and ICP0. Virology, 352 (2): 368-379.
        doi: 10.1016/j.virol.2006.04.044

    23. Li L, Johnson L A, Dai-Ju J Q, Sandri-Goldin R M. 2008. Focus formation at the periphery of HSV-1 transcription sites requires ICP27. PLoS ONE, 3 (1): e 1491.
        doi: 10.1371/journal.pone.0001491

    24. McGregor F, Phelan A, Dunlop J et al. 1996. Regulation of herpes simplex virus poly(A) site usage and the action of immediate-early protein IE63 in the early-late switch. J Virol, 70 (3): 1931-1940.

    25. McLauchlan J, Phelan A, Loney C, et al. 1992. Herpes simplex virus IE63 acts at the posttranscriptional level to stimulate viral mRNA 3' processing. J Virol, 66 (12): 6939-6945.

    26. Mears W E, Lam V, Rice S A. 1995. Identification of nuclear and nucleolar localization signals in the herpes simplex virus regulatory protein ICP27. J Virol, 69 (2): 935-947.

    27. Mears W E, Rice S A. 1996. The RGG box motif of the herpes simplex virus ICP27 protein mediates an RNA-binding activity and determines in vivo methylation. J Virol, 70 (11): 7445-7453.

    28. Mears W E, Rice S A. 1998. The herpes simplex virus immediate-early protein ICP27 shuttles between nucleus and cytoplasm. Virology, 242 (1): 128-137.
        doi: 10.1006/viro.1997.9006

    29. Melchjorsen J, Sirén J, Julkunen l, et al. 2006. Induction of cytokine expression by herpes simplex virus in human monocyte-derived macrophages and dendritic cells is dependent on virus replication and is counteracted by ICP27 targeting NF-κB and IRF-3. J Gen Virol, 87 (5): 1099-1108
        doi: 10.1099/vir.0.81541-0

    30. Melroe G, DeLuca N, Knipe D M. 2004. Herpes simplex virus 1 has multiple mechanisms for blocking virus-induced interferon production. J Virol, 78 (16): 8411-8420.
        doi: 10.1128/JVI.78.16.8411-8420.2004

    31. Mossman K. 2005. Analysis of anti-interferon properties of the herpes simplex virus type I ICP0 protein. Methods Mol Med, 116: 195-205.

    32. Park M S, Shaw M L, Munoz-Jordan J, et al.2003. Newcastle disease virus (NDV)-based assay demonstrates interferon-antagonist activity for the NDV V protein and the Nipah virus V, W, and C proteins. J Virol, 77 (2), 1501-1511.
        doi: 10.1128/JVI.77.2.1501-1511.2003

    33. Phelan A, Clements J B. 1997. Herpes simplex virus type 1 immediate early protein IE63 shuttles between nuclear compartments and the cytoplasm. J Gen Virol, 78 (12): 3327-3331.
        doi: 10.1099/0022-1317-78-12-3327

    34. Platanias L C, Fish E N. 1999. Signaling pathways activated by interferons. Exp Hematol, 27 (11): 1583-1592.
        doi: 10.1016/S0301-472X(99)00109-5

    35. Rodrigues J P, Rode M, Gatfield D, et al. 2001. REF proteins mediate the export of spliced and unspliced mRNAs from the nucleus. Proc Natl Acad Sci USA, 98 (3): 1030-1035.
        doi: 10.1073/pnas.98.3.1030

    36. Samuel C E. 2001. Antiviral actions of interferons. Clin Microbiol Rev, 14 (4), 778-809.
        doi: 10.1128/CMR.14.4.778-809.2001

    37. Sandri-Goldin R M. 1998. ICP27 mediates HSV RNA export by shuttling through a leucine-rich nuclear export signal and binding viral intronless RNAs through an RGG motif. Genes Dev, 12 (6): 868-879.
        doi: 10.1101/gad.12.6.868

    38. Sandri-Goldin R M. 2006. The functions and activities of herpes simplex virus protein ICP27, a multifunctional regulator of gene expression In: Alpha herpesviruses: molecular and cellular biology (Sandri-Goldin R M. ed. ), Caister Academic Press: Norfolk, United Kingdom. p65-83.

    39. Sanford J R, Gray N K, Beckmann K, et al. 2004. A novel role for shuttling SR proteins in mRNA translation. Genes Dev, 18 (7): 755-768.
        doi: 10.1101/gad.286404

    40. Sciabica K S, Dai Q J, Sandri-Goldin R M. 2003. ICP27 interacts with SRPK1 to mediate HSV splicing inhibition by altering SR protein phosphorylation. EMBO J, 22 (7): 1608-1619.
        doi: 10.1093/emboj/cdg166

    41. Sedlackova L, Rice S A. 2007. Herpes simplex virus type 1 immediate-early protein ICP27 is required for efficient incorporation of ICP0 and ICP4 into virions. J Virol, 82 (1): 268-277.

    42. Smith R W, Malik P, Clements J B. 2005. The herpes simplex virus ICP27 protein: a multifunctional post-transcriptional regulator of gene expression. Biochem Soc Trans, 33 (3): 499-501.
        doi: 10.1042/BST0330499

    43. Sobol P T, Mossman K L. 2006. ICP0 prevents RNase L-independent rRNA cleavage in herpes simplex virus type 1-infected cells. J Virol, 80 (1): 218-225.
        doi: 10.1128/JVI.80.1.218-225.2006

    44. Sokolowski M, Scott J E, Heaney R P, et al. 2003. Identification of herpes simplex virus RNAs that interact specifically with regulatory protein ICP27 in vivo. J Biol Chem, 278 (35):33540-33549.
        doi: 10.1074/jbc.M302063200

    45. Soliman T M, Sandri-Goldin R M, Silverstein S J. 1997. Shuttling of the herpes simplex virus type 1 regulatory protein ICP27 between the nucleus and cytoplasm mediates the expression of late proteins. J Virol, 71 (12): 9188-9197.

    46. Stutz F, Bachi A, Doerks T, et al. 2000. REF, an evolutionary conserved family of hnRNPlike proteins, interacts with TAP/Mex67p and participates in mRNA nuclear export. RNA, 6 (4): 638-650.
        doi: 10.1017/S1355838200000078

    47. Suganuma H, Kumada M, Omi T, et al. 2005. Aly/REF, a factor for mRNA transport, activates RH gene promoter function. FEBS J, 272 (11): 2696-2704.
        doi: 10.1111/ejb.2005.272.issue-11

    48. Toth Z, Stamminger T. 2008. The human cytome-galovirus regulatory protein UL69 and its effect on mRNA export. Front Biosci, 13: 2939-2949
        doi: 10.2741/2899

    49. Wilkie G S, Gautier P, Lawson D, et al. 2005. Embryonic poly(A)-binding protein stimulates translation in germ cells. Mol Cell Biol, 25 (5): 2060-2071.
        doi: 10.1128/MCB.25.5.2060-2071.2005

    50. Williams B J, Boyne J R, Goodwin D J, et al. 2005. The prototype gamma-2 herpesvirus nucleocytoplasmic shuttling protein, ORF 57, transports viral RNA through the cellular mRNA export pathway. Biochem J, 387 (2): 295-308.
        doi: 10.1042/BJ20041223

    51. Yoshida M, Horinouchi S. 1999. Trichostatin and lep-tomycin. Inhibition of histone deacetylation and signal-dependent nuclear export. Ann N Y Acad Sci, 886: 23-36.
        doi: 10.1111/j.1749-6632.1999.tb09397.x

    52. Young D F, Didcock L, Goodbourn S, et al.2000. Paramyxoviridae use distinct virus-specific mechanisms to circumvent the interferon response. Virology, 269 (2): 383-390.
        doi: 10.1006/viro.2000.0240

    53. Zhou C, Knipe D M. 2002. Association of herpes simplex virus type 1 ICP8 and ICP27 proteins with cellular RNA polymerase Ⅱ holoenzyme. J Virol, 76 (12): 5893-5904.
        doi: 10.1128/JVI.76.12.5893-5904.2002

  • 加载中

Article Metrics

Article views(4474) PDF downloads(19) Cited by()

Related
Proportional views

    The Herpes Simplex Virus Type 1 Multiple Function Protein ICP27

      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:  National Natural Science Foundation of China 30870120Start Fund of the Hundred Talents Program of the Chinese Academy of Science 20071010-141Open Research Fund Program of the State Key Laboratory of Virology of China 2007003

    Abstract: The herpes simplex virus type 1 (HSV-1) infected-cell protein 27 (ICP27) is an essential, highly conserved protein involved in various steps of HSV-1 gene regulation as well as in the shut-off of host gene expression during infection. It functions primarily at the post-transcriptional level in inhibiting precursor mRNA splicing and in promoting nuclear export of viral transcripts. Recently, many novel functions performed by the HSV-1 ICP27 protein were shown, including leptomycin B resistance, inhibition of the typeⅠinterferon signaling, regulation of the viral mRNA translation and determining the composition of HSV-1 virions.

    • 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. During lytic infection, over 80 gene products are expressed in a highly regulated temporal cascade and can be divided into three classes: immediate-early (IE), early and late (18). HSV-1 infected-cell protein 27 (ICP27) is a 63 kDa immediate-early regulatory phosphoprotein homologous to gene products identi-fied in all classes of herpesviruses so far, including Epstein-Barr virus (EBV) transactivator protein Mta encoded by BMLF1, ORF57 of Kaposi's sarcoma-associated herpesvirus (KSHV), ORF57 of Herpesvirus saimiri (HVS), ORF4 of Varicella-Zoster virus and UL69 in human cytomegalovirus (48).

      HSV-1 ICP27 is an essential and multifunctional regulator of gene expression that modulates the synthesis and maturation of viral and cellular mRNAs (9).ICP27 inhibits pre-mRNA splicing (15, 40), stimulates pre-mRNA 3' processing (25), affects mRNA stability (2), and shuttles between the nucleus and the cytoplasm (33, 37, 45), promoting viral RNA nuclear export (3, 19, 37). In a study where use of deletions shows that ICP27 may contribute to HSV-1 leptomycin B (LMB) sensitivity (22). Much recent work in several labo-ratories has demonstrated that ICP27 has a role in promoting translation of viral mRNA (9, 10, 21). ICP27 is also involved in the inhibition of type Ⅰ interferon (IFN) signaling (18, 25) and determining the composition of HSV-1 virions (41).

    • ICP27 is an RNA binding protein that shuttles between the nucleus and the cytoplasm (27, 28, 33, 37, 45). ICP27 contains an arginine and glycine-rich motif, the RGG-box, which allows it to bind to RNA (27). ICP27 also contains two nuclear localization sequences (NLS) (26) and a leucine-rich sequence that bears a strong resemblance to the nuclear export sequence (NES) of the human immunodeficiency virus (HIV) protein Rev (37). Rev mediates the export of HIV-1 mRNA by binding the cellular nuclear transport receptor chromosome region maintenance 1 (CRM1) through its NES, an interaction that is inhibited by LMB, an inhibitor of CRM1 (42). It has been demonstrated that the CRM1 plays a role in ICP27 export (24, 37). CRM1 recognizes its cargo proteins by binding directly to leucine-rich NESs in those polypeptides. However, Lengyel et al. have shown that ICP27 can efficiently exit the nucleus in the presence of LMB, suggesting that CRM1 does not mediate the nuclear export of ICP27 (22). Moreover, Koffa et al. reported that CRM1 is not required for ICP27-induced mRNA export in Xenopus laevis oocytes (19), and Chen et al. provided evidence that the leucine-rich NES of ICP27 does not require CRM1 for its activity (4).

      Most of the recent evidence suggests that ICP27 utilizes the TAP (a non-karyopherin export receptor that mediates transport of mRNAs to the cytoplasm) system, believed to be the major pathway for bulk export of spliced cellular mRNAs (3, 8). It has been demonstrated that at later times, beginning at about 6 h after infection, ICP27 begins to shuttle between the nucleus and cytoplasm (4, 28, 33, 37, 45). ICP27 binds to HSV-1 mRNAs (27, 37, 44) and facilitates their export to the cytoplasm by interacting with the cellular RNA export adaptor protein Aly/REF (RNA and export factor binding proteins) and the cellular export receptor TAP/NXF (3, 4, 19, 27, 45). Aly/REF interacts directly with TAP/NXF1 (46), and excess Aly/REF increased the rate and efficiency of mRNA export in vivo (35). ICP27 binds to the cellular mRNA export factors REF and TAP in an RNase sensitive manner suggesting that RNA may be a stabilizing component of the ICP27 viral mRNA export complex (16). A similar observation has also been reported for EB2 and HVS ORF57 (16, 50).

      It may be that the interactions of ICP27 with Aly/ REF and TAP facilitate the export of viral mRNAs during wild-type infection, since ICP27 stimulates the export of viral intronless mRNAs, but not cellular mRNAs, after microinjection into X. laevis oocytes, although these interactions are not required for late mRNAs to be exported, the interaction of ICP27 with TAP/NXF1 occurs after ICP27 leaves viral transcri-ption sites (19). However, Chen et al. reported that ICP27 interacts directly with TAP/NXF1 and does not require Aly/REF to bridge the interaction (3).

      An earlier study shows that, ICP27 was required for efficient nuclear export of viral mRNAs (37). However, more recent studies have shown that ICP27 is not required in HSV-infected cells for efficient cytop-lasmic accumulation of at least certain HSV-1 mRNAs (10), such as HSV-1 late mRNAs VP16, ICP5, gB, gC, and gD are all exported into the cytoplasm in the absence of ICP27 (9, 10). It is possible that viral late mRNAs are exported via a different pathway in the absence of ICP27 or that viral late mRNAs continue to be exported via the REF/TAP pathway even in the absence of ICP27. Meanwhile, it has been reported that, TAP plays a role in promoting the translation of unspliced mRNAs (17), and Aly/REF enhances transcriptional promoter activity (47). It may be that the interaction of ICP27 with Aly/REF and TAP facilitates late mRNAs transcription and translation, respectively, and that the role in late mRNAs export is minimal (10).

      ICP27 has been demonstrated to interact with RNA polymerase Ⅱ (RNAPⅡ) and facilitates its recruitment to viral transcription sites (6, 53), but it is not known whether, once RNAPⅡ is localized to the viral trans-cription sites, ICP27-binding to RNAⅡ is required for ICP27-mediated viral mRNA export. Although the exact molecular mechanisms are still not clearly under-stood, exploring the mechanism of how the herpes viral mRNA export to the cytoplasm may advance the knowledge of the pathogenesis of the herpes viruses and may enable the development of new antiviral approaches (48).

    • It was previously shown that HSV-1 infection is sensitive to LMB (51). Because LMB has no known targets other than CRM1, it is presumed that the LMB sensitivity of HSV-1 infection reflects a need by the virus to utilize CRM1 during its lytic infection. However, Lengyel et al. reported that the amino N-terminus of ICP27 is involved in the LMB resistance, and an unexpected linkage between LMB resistance and the nucleocytoplasmic distribution of two other HSV-1 IE regulatory proteins, ICP0 and ICP4. The same amino (N)-terminal region of ICP27 that determines sensitivity to LMB also enhances ICP27's previously documented ability to promote the cytoplasmic accumulation of ICP4 and ICP0. There-fore they speculate that ICP27's effects on ICP4 and ICP0 may contribute to HSV-1 LMB sensitivity (22).

    • Recent studies have shown that HSV-1 ICP27 is involved in the innate immune response (18, 29). ICP27 has been identified as an important viral protein counteracting the early innate immune response (29).

      Because the innate immune response promotes viral clearance, it is important for HSV-1 to have processes that inhibit these pathways in order to productively infect the epithelial cells, establish latent infection, and persist in neurons throughout the life of the host (18). Several viruses have evolved strategies to evade type Ⅰ IFN signaling at different stages of the pathway (20, 32, 52). Herpes viruses also have evolved anti-IFN signaling activities (18). HSV-1 has several mechanisms to inhibit type Ⅰ IFN production, the activities of IFN-stimulated genes (ISGs), and the IFN signaling pathway itself (18).

      Type Ⅰ IFN (IFN-α/β) signaling is an important antiviral response that results in the expression of antiviral, antiproliferative, and immunomodulatory proteins (34). IFN-α/β binds the type Ⅰ IFN receptor subunits (IFNAR-1 and-2), causing heterodimeri-zation and phosphorylation of the subunits. This activation leads to the phosphorylation of the Janus kinases, Jak-1 and Tyk-2, which in turn phosphorylate signal transducers and activators of transcription (STATs) 1 and 2. The STATs heterodimerize and translocate into the nucleus, where they associate with p48 (also known as IRF-9), bind a cis-acting DNA element called the IFN stimulated response element, or ISRE (34) for the transactivation of ISGs (12, 36).

      In a study, Johnson et al. reported that the inhibition of the Jak/STAT pathway by HSV-1 requires viral gene expression and that viral immediate-early protein ICP27 plays a role in downregulating STAT-1 phosp-horylation and in preventing the accumulation of STAT-1 in the nucleus (18). The reduced STAT-1 phosphorylation response to IFN-α treatment in cells infected with the ICP0 mutant could indicate that ICP0 is also involved in the inhibition of the Jak/ STAT pathway. ICP0 has been shown to have many activities to counteract various stages of IFN signaling (14, 30, 31, 43), but because ICP27 expression is sufficient to inhibit IFN-α-induced nuclear accumu-lation of STAT-1, it seems that any role ICP0 might play in this process is secondary to that of ICP27 (18).

    • HSV-1 ICP27 protein is an essential regulator of viral gene expression with roles at various levels of RNA metabolism in the nucleus. More recently, intensive studies have shown that ICP27 can interact with host translation factors (10, 21) and enhance the translation of certain viral mRNAs (9, 10). The ICP27 C terminus is required for stimulation of translation (21).

      The widely used tethered function assay of Xenopus oocytes was employed to test the effect of ICP27 on translation in vivo (5, 13, 39, 49). The data show that ICP27 can directly stimulate the translation of mRNAs to which it is bound and establish that ICP27 is sufficient to stimulate translation in the absence of any other viral factors (21). Moreover, Fontaine-Rodriguez et al. has showed that the interaction of ICP27 with translation factors are required for efficient translation of specific viral mRNAs (10), and they also confirmed that ICP27 stimulates translation of VP16 mRNA as well as an additional HSV-1 late ICP5 mRNA, and this function requires the C terminus of ICP27 (10). Ellison et al. also showed that ICP27 enhances VP16 expression primarily by increasing the translational efficiency of VP16 mRNA (9).

    • HSV-1 virions are composed of four morphologi-cally distinct structures: core, capsid, tegument, and envelope. Viral proteins that are present in the cell at the very earliest stages of infection have critical regu-latory functions, e.g., to counteract host immunity or to transactivate viral gene expression. Recently, Sed-lackova and Rice have showed that ICP27 plays a previously unrecognized role in determining the composition of HSV-1 virions (41).

      ICP27 is a nucleocytoplasmic shuttling protein that performs a number of functions during infection, most notably the induction of several E and L viral genes (38, 42). ICP27 also determines the intracellular locali-zation of ICP0 and ICP4. Furthermore, the mechanism by which ICP27 promotes ICP0/4 cytoplasmic locali-zation does not require the environment of the in-fected cell or other viral factors (41). As both ICP0 and ICP4 are known to be minor virion components, Sedlackova and Rice used a viral ICP27 deletion mutant d3-4 to test the hypothesis that the cytoplasmic localization of these proteins is required for their incorporation into viral particles (41). Consistent with this conjecture, d3-4 virions were found to lack ICP0 in their tegument and to have greatly reduced levels of ICP4. Thus, the cytoplasmic localization of ICP0 and ICP4 appears to be a prerequisite for the assembly of these important transcriptional regulatory proteins into viral particles. Moreover, their results show that ICP27 plays a previously unrecognized role in determining the composition of HSV-1 virions (41). Namely, the IE protein ICP27, itself not a virion protein, is a required cofactor for the efficient virion incorporation of ICP0 and ICP4. Given that ICP27 can promote the cytoplasmic accumulation of ICP0/4 even in the absence of infection, it is likely that ICP27's role in virion assembly is indirect, though the mechanism by which ICP27 promotes the cytoplasmic accumulation of ICP0/4 is unknown (41). Thus, it appears that ICP27 can affect the composition of both the tegument and envelope, but by apparently distinct mechanisms (41).

    • Herpes viruses are highly disseminated in nature. Herpes simplex virus type 1 is one of the most in-tensively investigated viruses. HSV-1 serves as models and tools for the study of translocation of proteins, membrane structure, gene regulation, gene therapy, cancer therapy and so on. ICP27, conserved in all mammalian and avian herpes viruses, is a multifunctional regulatory protein that is required for HSV-1 productive infection. It is responsible for both repression and trans-activation of viral gene expression, depending on the target gene, at a post-transcriptional level. ICP27 protein is involved in various steps of HSV-1 gene regulation and functions to shut off host protein synthesis by inhibiting the splicing of host pre-mRNAs, and promoting nuclear export of viral mRNAs. Recently, many novel functions performed by the ICP27 protein, including LMB resistance, inhibition of the typeⅠIFN signaling, regulation of the viral mRNA translation and determining the composition of HSV-1 virions, were reported. Under-standing the mechanism of the multifunctional regulatory protein ICP27 may advance our knowledge of the HSV-1 and promote the exploitation of efficient methods for prevent viral infection and antiviral therapy.

    Reference (53) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return