Citation: Yijie Ma, Dustin Verpooten, Bin He. Herpes Simplex Viruses and Induction of Interferon Responses .VIROLOGICA SINICA, 2008, 23(6) : 416-428.  http://dx.doi.org/10.1007/s12250-008-2999-7

Herpes Simplex Viruses and Induction of Interferon Responses

  • Corresponding author: Bin He, tshuo@uic.edu
  • Received Date: 01 September 2008
    Accepted Date: 03 September 2008
    Available online: 01 December 2008
  • Herpes simplex viruses (HSV) are human pathogens responsible for a variety of diseases, including localized mucocutaneous lesions, encephalitis, and disseminated diseases. HSV infection leads to rapid induction of innate immune responses. A critical part of this host response is the type I IFN system including the induction of type I IFNs, IFN-mediated signaling and amplification of IFN response. This provides the host with immediate countermeasure during acute infection to limit initial viral replication and to facilitate an appropriate adaptive immune response. However, HSV has devised multiple strategies to evade and interfere with innate immunity. This review will focus on the induction of type I IFN response by HSV during acute infection and current knowledge of mechanisms by which HSV interferes with this induction process.

  • 加载中
    1. Alexopoulou L, Holt A C, Medzhitov R, et al. 2001. Recognition of double-stranded RNA and activation of NF-[kappa]B by Toll-like receptor 3. Nature, 413 (6857):732.
        doi: 10.1038/35099560

    2. Ankel H, Westra D F, Welling-Wester S, et al. 1998. Induction of Interferon-[alpha] by Glycoprotein D of Herpes Simplex Virus: A Possible Role of Chemokine Receptors. Virology, 251 (2):317.
        doi: 10.1006/viro.1998.9432

    3. Aravalli R, Peterson P, Lokensgard J. 2007. Toll-like Receptors in Defense and Damage of the Central Nervous System. J Neuroimmune Pharmacol, 2 (4):297.
        doi: 10.1007/s11481-007-9071-5

    4. Au W, Moore P A, LaFleur D W, et al. 1998. Characterization of the Interferon Regulatory Factor-7 and Its Potential Role in the Transcription Activation of Interferon A Genes. J Biol Chem, 273 (44):29210-29217.
        doi: 10.1074/jbc.273.44.29210

    5. Au W, Moore P A, Lowther W, et al. 1995. Identification of a Member of the Interferon Regulatory Factor Family that Binds to the Interferon-Stimulated Response Element and Activates Expression of Interferon-Induced Genes. Proc Natl Acad Scie USA, 92 (25):11657-11661.
        doi: 10.1073/pnas.92.25.11657

    6. Bosnjak L, Jones C A, Abendroth A, et al. 2005. Dendritic Cell Biology in Herpesvirus Infections. Viral Immunol, 18 (3):419-433.
        doi: 10.1089/vim.2005.18.419

    7. Casrouge A, Zhang S Y, Eidenschenk C, et al. 2006. Herpes Simplex Virus Encephalitis in Human UNC-93B Deficiency. Science, 314 (5797):308-312.
        doi: 10.1126/science.1128346

    8. Cassady K A, Gross M, Roizman B. 1998. The Second-Site Mutation in the Herpes Simplex Virus Recombinants Lacking the gamma 134.5 Genes Precludes Shutoff of Protein Synthesis by Blocking the Phosphory-lation of eIF-2alpha. J Virol, 72 (9):7005-7011.

    9. Cassady K A, Gross M, Roizman B. 1998. The Herpes Simplex Virus Us11 Protein Effectively Compensates for the g134.5 Gene if Present before Activation of Protein Kinase R by Precluding Its Phosphorylation and That of the alpha subunit of Eukaryotic Translation Initiation Factor 2. J Virol, 72 (11):8620-8626.

    10. Cerveny M, Hessefort S, Yang K, et al. 2003. Amino acid substitutions in the effector domain of the g134.5 protein of herpes simplex virus 1 have differential effects on viral response to interferon-a. Virology, 307 (2):290.
        doi: 10.1016/S0042-6822(02)00075-2

    11. Chee A V, Roizman B. 2004. Herpes Simplex Virus 1 Gene Products Occlude the Interferon Signaling Pathway at Multiple Sites. J Virol, 78 (8):4185-4196.
        doi: 10.1128/JVI.78.8.4185-4196.2004

    12. Cheng G, Brett M E, He B. 2001. Val193 and Phe195 of the g134.5 Protein of Herpes Simplex Virus 1 Are Required for Viral Resistance to Interferon-α/β. Virology, 290 (1):115.
        doi: 10.1006/viro.2001.1148

    13. Cheng G, Gross M, Brett M E, et al. 2001. AlaArg Motif in the Carboxyl Terminus of the g134.5 Protein of Herpes Simplex Virus Type 1 Is Required for the Formation of a High-Molecular-Weight Complex That Dephosphorylates eIF-2a. J Virol, 75 (8):3666-3674.
        doi: 10.1128/JVI.75.8.3666-3674.2001

    14. Cheng G, Yang K, He B. 2003. Dephosphorylation of eIF-2a Mediated by the g134.5 Protein of Herpes Simplex Virus Type 1 Is Required for Viral Response to Interferon but Is Not Sufficient for Efficient Viral Replication. J Virol, 77 (18):10154-10161.
        doi: 10.1128/JVI.77.18.10154-10161.2003

    15. Cheng G, Zhong J, Chung J, et al. 2007. Double-stranded DNA and double-stranded RNA induce a common antiviral signaling pathway in human cells. Proc Natl Acad Scie USA, 104 (21):9035-9040.
        doi: 10.1073/pnas.0703285104

    16. Collins S E, Noyce R S, Mossman K L. 2004. Innate Cellular Response to Virus Particle Entry Requires IRF3 but Not Virus Replication. J Virol, 78 (4):1706-1717.
        doi: 10.1128/JVI.78.4.1706-1717.2004

    17. Diebold S S, Kaisho T, Hemmi H, et al. 2004. Innate Antiviral Responses by Means of TLR7-Mediated Recognition of Single-Stranded RNA. Science, 303 (5663):1529-1531.
        doi: 10.1126/science.1093616

    18. Duerst R J, Morrison L A. 2004. Herpes simplex virus 2 virion host shutoff protein interferes with type Ⅰ interferon production and responsiveness. Virology, 322 (1):158.
        doi: 10.1016/j.virol.2004.01.019

    19. Eidson K M, Hobbs W E, Manning B J, et al. 2002. Expression of Herpes Simplex Virus ICP0 Inhibits the Induction of Interferon-Stimulated Genes by Viral In-fection.J Virol, 76 (5):2180-2191.
        doi: 10.1128/jvi.76.5.2180-2191.2002

    20. Elain G, Romero P, Segal D, et al. 2007. TLR3 Deficiency in Patients with Herpes Simplex Encephalitis. Science, 317 (5844): 1522-1527.
        doi: 10.1126/science.1139522

    21. Fitzgerald K A, McWhirter S M, Faia K L, et al. 2003. IKKε and TBK1 are essential components of the IRF3 signaling pathway. Nat Immunol, 4 (5):491.
        doi: 10.1038/ni921

    22. 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

    23. Garcia M A, Meurs E F, Esteban M. 2007. The dsRNA protein kinase PKR: Virus and cell control. Biochimie, 89 (6-7):799.
        doi: 10.1016/j.biochi.2007.03.001

    24. Gitlin L, Barchet W, Gilfillan S, et al. 2006. Essential role of mda-5 in type Ⅰ IFN responses to polyriboinosinic: polyribocytidylic acid and encephalomyocarditis picor-navirus. Proc Natl Acad Scie USA, 103 (22):8459-8464.
        doi: 10.1073/pnas.0603082103

    25. Guo J, Peters K L, Sen G C. 2000. Induction of the Human Protein P56 by Interferon, Double-Stranded RNA, or Virus Infection. Virology, 267 (2):209.
        doi: 10.1006/viro.1999.0135

    26. Hacker H, Redecke V, Blagoev B, et al. 2006. Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6. Nature, 439 (7073):204.
        doi: 10.1038/nature04369

    27. He B, Chou J, Brandimarti R, et al. 1997. Suppression of the phenotype of g134.5- herpes simplex virus 1: failure of activated RNA-dependent protein kinase to shut off protein synthesis is associated with a deletion in the domain of the α 47 gene. J Virol, 71 (8):6049-6054.

    28. He B, Gross M, Roizman B. 1997. The g134.5 protein of herpes simplex virus 1 complexes with protein phosphatase 1α to dephosphorylate the alpha subunit of the eukaryotic translation initiation factor 2 and preclude the shutoff of protein synthesis by double-stranded RNA-activated pro-tein kinase. PNAS, 94 (3):843-848.
        doi: 10.1073/pnas.94.3.843

    29. Heil F, Hemmi H, Hochrein H, et al. 2004. Species-Specific Recognition of Single-Stranded RNA via Toll-like Receptor 7 and 8. Science, 303 (5663):1526-1529.
        doi: 10.1126/science.1093620

    30. Hemmi H, Takeuchi O, Kawai T, et al. 2000. A Toll-like receptor recognizes bacterial DNA. Nature, 408 (6813):740.
        doi: 10.1038/35047123

    31. Herbst-Kralovetz M M, Pyles R B. 2006. Toll-like Receptors, Innate Immunity and HSV Pathogenesis. Herpes, 13 (2):37-41.

    32. Hochrein H, Schlatter B, O'Keeffe M, et al. 2004. Herpes simplex virus type-1 induces IFN-{alpha} produc-tion via Toll-like receptor 9-dependent and -independent pathways. Proc Natl Acad Scie USA, 101 (31):11416-11421.
        doi: 10.1073/pnas.0403555101

    33. Hornung V, Ellegast J, Kim S, et al. 2006. 5'-Triphosp-hate RNA Is the Ligand for RIG-Ⅰ. Science, 314 (5801):994-997.
        doi: 10.1126/science.1132505

    34. Ishii K J, Akira S. 2006. Innate immune recognition of, and regulation by, DNA. Trends Immunol, 27 (11):525.
        doi: 10.1016/j.it.2006.09.002

    35. Ishii K J, Coban C, Kato H, et al. 2006. A Toll-like receptor-independent antiviral response induced by double-stranded B-form DNA. Nat Immunol, 7 (1):40.
        doi: 10.1038/ni1282

    36. Ishii K J, Kawagoe T, Koyama S, et al. 2008. TANK-binding kinase-1 delineates innate and adaptive immune responses to DNA vaccines. Nature, 451 (7179):725.
        doi: 10.1038/nature06537

    37. Jacquemont B, Roizman B. 1975. RNA synthesis in cells infected with herpes simplex virus. X. Properties of viral symmetric transcripts and of double-stranded RNA prepared from them. J Virol, 15 (4):707-713.

    38. Jiang Z, Mak T W, Sen G, et al. 2004. Toll-like receptor 3-mediated activation of NF-{kappa}B and IRF3 diverges at Toll-IL-1 receptor domain-containing adapter inducing IFN-{beta}. Proc Natl Acad Scie USA, 101 (10):3533-3538.
        doi: 10.1073/pnas.0308496101

    39. 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.
        doi: 10.1016/j.virol.2008.01.001

    40. Kato H, Takeuchi O, Sato S, et al. 2006. Differential roles of MDA5 and RIG-Ⅰ helicases in the recognition of RNA viruses. Nature, 441 (7089):101.
        doi: 10.1038/nature04734

    41. Kawai T, Takahashi K, Sato S, et al. 2005. IPS-1, an adaptor triggering RIG-Ⅰ-and Mda5-mediated type Ⅰ in-terferon induction. Nat Immunol, 6 (10):981.
        doi: 10.1038/ni1243

    42. Kawai T, Akira S. 2006. Innate immune recognition of viral infection. Nat Immunol, 7 (2):131.
        doi: 10.1038/ni1303

    43. Kim J C, Lee S Y, Kim S Y, et al. 2008. HSV-1 ICP27 suppresses NF-[kappa]B activity by stabilizing Ⅰ[kappa]B [alpha]. FEBS Letters, 582 (16):2371.
        doi: 10.1016/j.febslet.2008.05.044

    44. Konat G W, Kielian T, Marriott I. 2006. The role of Toll-like receptors in CNS response to microbial challenge. J Neurochem, 99 (1):1-12.
        doi: 10.1111/jnc.2006.99.issue-1

    45. Korom M, Wylie K M, Morrison L A. 2008. Selective Ablation of Virion Host Shutoff Protein RNase Activity Attenuates Herpes Simplex Virus 2 in Mice. J Virol, 82 (7):3642-3653.
        doi: 10.1128/JVI.02409-07

    46. Krug A, Luker G D, Barchet W, et al. 2004. Herpes simplex virus type 1 activates murine natural interferon-producing cells through toll-like receptor 9. Blood, 103 (4):1433-1437.

    47. Ku C L, von Bernuth H, Picard C, et al. 2007. Selective predisposition to bacterial infections in IRAK-4 deficient children: IRAK-4 dependent TLRs are otherwise redundant in protective immunity. J Exp Med, 204 (10):2407-2422.
        doi: 10.1084/jem.20070628

    48. Kumar-Sinha C, Varambally S, Sreekumar A, et al. 2002. Molecular Cross-talk between the TRAIL and Interferon Signaling Pathways. J Biol Chem, 277 (1):575-585.
        doi: 10.1074/jbc.M107795200

    49. Kumar H, Zheng M, Atherton S S, et al. 2006. Herpes simplex virus 1 infection induces the expression of proin-flammatory cytokines, interferons and TLR7 in human corneal epithelial cells. Immunology, 117 (2):167-176.
        doi: 10.1111/imm.2006.117.issue-2

    50. Kurt-Jones E A, Chan M, Zhou S, et al. 2004. Herpes simplex virus 1 interaction with Toll-like receptor 2 contri-butes to lethal encephalitis. Proc Natl Acad Scie USA, 101 (5):1315-1320.
        doi: 10.1073/pnas.0308057100

    51. Lebon P. 1985. Inhibition of Herpes Simplex Virus Type 1-induced Interferon Synthesis by Monoclonal Antibodies against Viral Glycoprotein D and by Lysosomotropic Drugs.J Gen Virol, 66 (12):2781-2786.
        doi: 10.1099/0022-1317-66-12-2781

    52. Leib D A, Harrison T E, Laslo K M, et al. 1999. Interferons Regulate the Phenotype of Wild-type and Mutant Herpes Simplex Viruses In Vivo. J Exp Med, 189 (4):663-672.
        doi: 10.1084/jem.189.4.663

    53. Lin R, Noyce R S, Collins S E, et al. 2004. The Herpes Simplex Virus ICP0 RING Finger Domain Inhibits IRF3-and IRF7-Mediated Activation of Interferon-Stimulated Genes. J Virol, 78 (4):1675-1684.
        doi: 10.1128/JVI.78.4.1675-1684.2004

    54. Lund J, Sato A, Akira S, et al. 2003. Toll-like Receptor 9-mediated Recognition of Herpes Simplex Virus-2 by Plasmacytoid Dendritic Cells. J Exp Med, 198 (3):513-520.
        doi: 10.1084/jem.20030162

    55. Malmgaard L, Paludan S. R. 2003. Interferon (IFN)-{alpha}/{beta}, interleukin (IL)-12 and IL-18 coordinately induce production of IFN-{gamma} during infection with herpes simplex virus type 2. J Gen Virol, 84 (9):2497-2500.
        doi: 10.1099/vir.0.19251-0

    56. Malmgaard L, Melchjorsen J, Bowie A G, et al. 2004. Viral Activation of Macrophages through TLR-Dependent and -Independent Pathways. J Immunol, 173 (11):6890-6898.
        doi: 10.4049/jimmunol.173.11.6890

    57. Medzhitov R, Preston-Hurlburt P, Kopp E, et al. 1998. MyD88 Is an Adaptor Protein in the hToll/IL-1 Receptor Family Signaling Pathways. Mol Cell, 2 (2):253-258.
        doi: 10.1016/S1097-2765(00)80136-7

    58. Melchjorsen J, Siren J, Julkunen I, 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-{kappa}B and IRF-3. J Gen Virol, 87 (5):1099-1108.
        doi: 10.1099/vir.0.81541-0

    59. Melroe G T, DeLuca N A, 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

    60. Melroe G T, Silva L, Schaffer P A, et al. 2007. Recruit-ment of activated IRF-3 and CBP/p300 to herpes simplex virus ICP0 nuclear foci: Potential role in blocking IFN-[beta] induction. Virology, 360 (2):305.
        doi: 10.1016/j.virol.2006.10.028

    61. Mercurio F, Zhu H, Murray B W, et al. 1997. IKK-1 and IKK-2: Cytokine-Activated Ⅰ{kappa}B Kinases Essential for NF-B Activation. Science, 278 (5339):860-866.
        doi: 10.1126/science.278.5339.860

    62. Meylan E, Curran J, Hofmann K, et al. 2005. Cardif is an adaptor protein in the RIG-Ⅰ antiviral pathway and is targeted by hepatitis C virus. Nature, 437 (7062):1167.
        doi: 10.1038/nature04193

    63. Mohr I, Sternberg D, Ward S, et al. 2001. A Herpes Simplex Virus Type 1 g134.5 Second-Site Suppressor Mutant That Exhibits Enhanced Growth in Cultured Glioblastoma Cells Is Severely Attenuated in Animals. J Virol, 75 (11):5189-5196.
        doi: 10.1128/JVI.75.11.5189-5196.2001

    64. Mohrl I, Gluzman Y. 1996. A herpesvirus genetic element which affects translation in the absence of the viral GADD34 function. EMBO J, 15 (17):4759-4766.

    65. Mossman K L, Saffran H A, Smiley J R. 2000. Herpes Simplex Virus ICP0 Mutants Are Hypersensitive to Interferon. J Virol, 74 (4):2052-2056.
        doi: 10.1128/JVI.74.4.2052-2056.2000

    66. Mossman K L, Macgregor P F, Rozmus J J, et al. 2001. Herpes Simplex Virus Triggers and Then Disarms a Host Antiviral Response. J Virol, 75 (2):750-758.
        doi: 10.1128/JVI.75.2.750-758.2001

    67. Mossman K L, Ashkar A A. 2005. Herpesviruses and the Innate Immune Response.Viral Immunol, 18 (2):267-281.
        doi: 10.1089/vim.2005.18.267

    68. Murphy J A, Duerst R J, Smith T J, et al. 2003. Herpes Simplex Virus Type 2 Virion Host Shutoff Protein Regulates Alpha/Beta Interferon but Not Adaptive Immune Responses during Primary Infection In Vivo. J Virol, 77 (17):9337-9345.
        doi: 10.1128/JVI.77.17.9337-9345.2003

    69. Narita M, Ando Y, Soushi S, et al. 1998. The BglII-N fragment of herpes simplex virus type 2 contains a region responsible for resistance to antiviral effects of interferon. J Gen Virol, 79 (3):565-572.
        doi: 10.1099/0022-1317-79-3-565

    70. Nicholl M J, Robinson L H, Preston C M. 2000. Activation of cellular interferon-responsive genes after infection of human cells with herpes simplex virus type 1. J Gen Virol, 81 (9):2215-2218.
        doi: 10.1099/0022-1317-81-9-2215

    71. Ninomiya-Tsuji J, Kishimoto K, Hiyama A, et al. 1999. The kinase TAK1 can activate the NIK-Ⅰ[kappa]B as well as the MAP kinase cascade in the IL-1 signalling pathway. Nature, 398 (6724):252.
        doi: 10.1038/18465

    72. Noyce R S, Collins S E, Mossman K L. 2006. Identification of a Novel Pathway Essential for the Immediate-Early, Interferon-Independent Antiviral Res-ponse to Enveloped Virions. J Virol, 80 (1):226-235.
        doi: 10.1128/JVI.80.1.226-235.2006

    73. Oganesyan G, Saha S K, Guo B, et al. 2006. Critical role of TRAF3 in the Toll-like receptor-dependent and -independent antiviral response. Nature, 439 (7073):208.
        doi: 10.1038/nature04374

    74. Okabe Y, Kawane K, Akira S, et al. 2005. Toll-like receptor-independent gene induction program activated by mammalian DNA escaped from apoptotic DNA degra-dation. J Exp Med, 202 (10):1333-1339.
        doi: 10.1084/jem.20051654

    75. Oshiumi H, Matsumoto M, Funami K, et al. 2003. TICAM-1, an adaptor molecule that participates in Toll-like receptor 3-mediated interferon-[beta] induction. Nat Immunol, 4 (2):161.
        doi: 10.1038/ni886

    76. Overton H, McMillan D, Hope L, et al. 1994. Production of Host Shutoff-Defective Mutants of Herpes Simplex Virus Type 1 by Inactivation of the UL13 Gene. Virology, 202 (1):97.
        doi: 10.1006/viro.1994.1326

    77. Paladino P, Cummings D T, Noyce R S, et al. 2006. The IFN-Independent Response to Virus Particle Entry Provides a First Line of Antiviral Defense That Is Independent of TLRs and Retinoic Acid-Inducible Gene Ⅰ. J Immunol, 177 (11):8008-8016.
        doi: 10.4049/jimmunol.177.11.8008

    78. Pasieka T J, Baas T, Carter V S, et al. 2006. Functional Genomic Analysis of Herpes Simplex Virus Type 1 Coun-teraction of the Host Innate Response. J Virol, 80 (15):7600-7612.
        doi: 10.1128/JVI.00333-06

    79. Pasieka T J, Lu B, Crosby S D, et al. 2008. Herpes Simplex Virus Virion Host Shutoff Attenuates Establish-ment of the Antiviral State. J Virol, 82 (11):5527-5535.
        doi: 10.1128/JVI.02047-07

    80. Pedersen E B, Haahr S, Mogensen S C. 1983. X-linked resistance of mice to high doses of herpes simplex virus type 2 correlates with early interferon production. Infect Immun, 42 (2):740-746.

    81. Perry A K, Chen G, Zheng D, et al. 2005. The host type Ⅰ interferon response to viral and bacterial infections. Cell Res, 15 (6):407.
        doi: 10.1038/sj.cr.7290309

    82. Pichlmair A, Schulz O, Tan C P, et al. 2006. RIG-Ⅰ-Mediated Antiviral Responses to Single-Stranded RNA Bearing 5'-Phosphates. Science, 314 (5801):997-1001.
        doi: 10.1126/science.1132998

    83. Platanias L C. 2005. Mechanisms of type-Ⅰ-and type-Ⅱ-interferon-mediated signalling. Nat Rev Immunol, 5 (5):375.
        doi: 10.1038/nri1604

    84. Preston C M, Harman A N, Nicholl M J. 2001. Activation of Interferon Response Factor-3 in Human Cells Infected with Herpes Simplex Virus Type 1 or Human Cytomegalovirus. J Virol, 75 (19):8909-8916.
        doi: 10.1128/JVI.75.19.8909-8916.2001

    85. Rasmussen S B, Sorensen L N, Malmgaard L, et al. 2007. Type Ⅰ Interferon Production during Herpes Simplex Virus Infection Is Controlled by Cell-Type-Specific Viral Recognition through Toll-Like Receptor 9, the Mito-chondrial Antiviral Signaling Protein Pathway, and Novel Recognition Systems. J Virol, 81 (24):13315-13324.
        doi: 10.1128/JVI.01167-07

    86. Reske A, Pollara G, Krummenacher C, et al. 2008. Glycoprotein-Dependent and TLR2-Independent Innate Immune Recognition of Herpes Simplex Virus-1 by Dendritic Cells. J Immunol, 180 (11):7525-7536.
        doi: 10.4049/jimmunol.180.11.7525

    87. Roizman B. 1999. HSV gene functions: what have we learned that could be generally applicable to its near and distant cousins? Acta Virol, 43 (2-3):75-80.

    88. Roizman B, Knipe D M. 2001. Herpes simplex viruses and their replication. In: Fields Virology (Knipe D M, Howley P, Griffin D, et al ed. ), 4th ed, vol. 2. Lippincott Williams & Wilkins, p 2399-2459.

    89. Rong Q, Alexander T S, Koski G K, et al. 2003. Multiple mechanisms for HSV-1 induction of interferon α produc-tion by peripheral blood mononuclear cells. Arch Virol, 148 (2):329.
        doi: 10.1007/s00705-002-0912-5

    90. Sato A, Linehan M M, Iwasaki A. 2006. Dual recognition of herpes simplex viruses by TLR2 and TLR9 in dendritic cells. Proc Natl Acad Scie USA, 103 (46):17343-17348.
        doi: 10.1073/pnas.0605102103

    91. Sato M, Suemori H, Hata N, et al. 2000. Distinct and Essential Roles of Transcription Factors IRF-3 and IRF-7 in Response to Viruses for IFN-[alpha]/[beta] Gene In-duction. Immunity, 13 (4):539.
        doi: 10.1016/S1074-7613(00)00053-4

    92. Sato M, Taniguchi T, Tanaka N. 2001. The interferon system and interferon regulatory factor transcription factors-studies from gene knockout mice. Cytokine & Growth Factor Rev, 12 (2-3):133.

    93. Sato S, Sugiyama M, Yamamoto M, et al. 2003. Toll/IL-1 Receptor Domain-Containing Adaptor Inducing IFN-{beta} (TRIF) Associates with TNF Receptor-Associated Factor 6 and TANK-Binding Kinase 1, and Activates Two Distinct Transcription Factors, NF-{kappa}B and IFN-Regulatory Factor-3, in the Toll-Like Receptor Signaling. J Immunol, 171 (8):4304-4310.
        doi: 10.4049/jimmunol.171.8.4304

    94. Seth R B, Sun L, Ea C K, et al. 2005. Identification and Characterization of MAVS, a Mitochondrial Antiviral Signaling Protein that Activates NF-κB and IRF3. Cell, 122 (5):669-682.
        doi: 10.1016/j.cell.2005.08.012

    95. Sharma S, tenOever B R, Grandvaux N, et al. 2003. Triggering the Interferon Antiviral Response Through an IKK-Related Pathway. Science, 300 (5622):1148-1151.
        doi: 10.1126/science.1081315

    96. Shimada T, Kawai T, Takeda K, et al. 1999. IKK-i, a novel lipopolysaccharide-inducible kinase that is related to Ⅰ{kappa}B kinases. Int Immunol, 11 (8):1357-1362.
        doi: 10.1093/intimm/11.8.1357

    97. Shirota H, Ishii K J, Takakuwa H, et al. 2006. Contri-bution of interferon-β to the immune activation induced by double-stranded DNA. Immunology, 118 (3):302-310.
        doi: 10.1111/imm.2006.118.issue-3

    98. Smiley J R. 2004. Herpes Simplex Virus Virion Host Shutoff Protein: Immune Evasion Mediated by a Viral RNase? J Virol, 78 (3):1063-1068.
        doi: 10.1128/JVI.78.3.1063-1068.2004

    99. Stetson1 D B, Medzhitov R. 2006. Type Ⅰ Interferons in Host Defense. Immunity, 25 (3):373-381.
        doi: 10.1016/j.immuni.2006.08.007

    100. Stetson D B, Medzhitov R. 2006. Recognition of Cytosolic DNA Activates an IRF3-Dependent Innate Immune Response. Immunity, 24 (1):93-103.
        doi: 10.1016/j.immuni.2005.12.003

    101. Su Y H, Oakes J E, Lausch R N. 1993. Mapping the genetic region coding for herpes simplex virus resistance to mouse interferon {alpha}/beta. J Gen Virol, 74 (11):2325-2332.
        doi: 10.1099/0022-1317-74-11-2325

    102. Sun Q, Sun L, Liu H H, et al. 2006. The Specific and Essential Role of MAVS in Antiviral Innate Immune Responses. Immunity, 24 (5):633.
        doi: 10.1016/j.immuni.2006.04.004

    103. Suzutani T, Nagamine M, Shibaki T, et al. 2000. The role of the UL41 gene of herpes simplex virus type 1 invasion of non-specific host defence mechanisms during primary infection. J Gen Virol, 81 (7):1763-1771.
        doi: 10.1099/0022-1317-81-7-1763

    104. Takaoka A, Wang Z, Choi M K, et al. 2007. DAI (DLM -1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature, 448 (7152):501.
        doi: 10.1038/nature06013

    105. Takeda K, Akira S. 2004. TLR signaling pathways Semin Immunol, 16 (1): 3-9.

    106. Wang Z, Choi M K, Ban T, et al. 2008. Regulation of innate immune responses by DAI (DLM-1/ZBP1) and other DNA-sensing molecules. Proc Natl Acad Scie USA, 105 (14):5477-5482.
        doi: 10.1073/pnas.0801295105

    107. Wathelet M G, Lin C H, Parekh B S, et al. 1998. Virus Infection Induces the Assembly of Coordinately Activated Transcription Factors on the IFN-[beta] Enhancer In Vivo. Mol Cell, 1 (4):507.
        doi: 10.1016/S1097-2765(00)80051-9

    108. Weber F, Wagner V, Rasmussen S B, et al. 2006. Double-Stranded RNA Is Produced by Positive-Strand RNA Viruses and DNA Viruses but Not in Detectable Amounts by Negative-Strand RNA Viruses. J Virol, 80 (10):5059-5064.
        doi: 10.1128/JVI.80.10.5059-5064.2006

    109. Xu L G, Wang Z, Han K J, et al. 2005. VISA Is an Adapter Protein Required for Virus-Triggered IFN-β Signaling. Mol Cell, 19 (6):727-740.
        doi: 10.1016/j.molcel.2005.08.014

    110. Yamamoto M, Sato S, Mori K, et al. 2002. Cutting Edge: A Novel Toll/IL-1 Receptor Domain-Containing Adapter That Preferentially Activates the IFN-{beta} Promoter in the Toll-Like Receptor Signaling. J Immunol, 169 (12):6668-6672.
        doi: 10.4049/jimmunol.169.12.6668

    111. Yang H, Lin C H, Ma G, et al. 2002. Transcriptional activity of interferon regulatory factor (IRF)-3 depends on multiple protein-protein interactions. Eur J Biochem, 269 (24):6142-6151.
        doi: 10.1046/j.1432-1033.2002.03330.x

    112. Yang K, Puel A, Zhang S, et al. 2005. Human TLR-7-, -8-, and -9-mediated induction of IFN-[alpha]/[beta] and -[lambda] is IRAK-4 dependent and redundant for protective immunity to viruses. Immunity, 23 (5):465.
        doi: 10.1016/j.immuni.2005.09.016

    113. Yoneyama M, Suhara W, Fukuhara Y, et al. 1998. Direct triggering of the type Ⅰ interferon system by virus infection: activation of a transcription factor complex containing IRF-3 and CBP/p300. EMBO J, 17 (4):1087-1095.
        doi: 10.1093/emboj/17.4.1087

    114. Yoneyama M, Kikuchi M, Natsukawa T, et al. 2004. The RNA helicase RIG-Ⅰ has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol, 5 (7):730.
        doi: 10.1038/ni1087

    115. Zandi E, Rothwarf D M, Delhase M, et al. 1997. The ⅠκB Kinase Complex (IKK) Contains Two Kinase Subunits, IKKα and IKKβ, Necessary for ⅠκB Phosphorylation and NF-κB Activation. cell, 91 (2):243-252.
        doi: 10.1016/S0092-8674(00)80406-7

    116. Zawatzky R, Kirchner H, DeMaeyer-Guignard J, et al. 1982. An X-linked Locus Influences the Amount of Circulating Interferon Induced in the Mouse by Herpes Simplex Virus Type 1. J Gen Virol, 63 (2):325-332.
        doi: 10.1099/0022-1317-63-2-325

    117. Zhang S Y, Jouanguy E, Sancho-Shimizu V, et al. 2007. Human Toll-like receptor-dependent induction of inter-ferons in protective immunity to viruses. Immunol Rev, 220 (1):225-236.
        doi: 10.1111/imr.2007.220.issue-1

    118. Zheng M, Klinman D M, Gierynska M, et al. 2002. DNA containing CpG motifs induces angiogenesis. Proc Natl Acad Scie USA, 99 (13):8944-8949.
        doi: 10.1073/pnas.132605599

  • 加载中

Article Metrics

Article views(4172) PDF downloads(14) Cited by()

Related
Proportional views

    Herpes Simplex Viruses and Induction of Interferon Responses

      Corresponding author: Bin He, tshuo@uic.edu
    • Department of Microbiology and Immunology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA

    Abstract: Herpes simplex viruses (HSV) are human pathogens responsible for a variety of diseases, including localized mucocutaneous lesions, encephalitis, and disseminated diseases. HSV infection leads to rapid induction of innate immune responses. A critical part of this host response is the type I IFN system including the induction of type I IFNs, IFN-mediated signaling and amplification of IFN response. This provides the host with immediate countermeasure during acute infection to limit initial viral replication and to facilitate an appropriate adaptive immune response. However, HSV has devised multiple strategies to evade and interfere with innate immunity. This review will focus on the induction of type I IFN response by HSV during acute infection and current knowledge of mechanisms by which HSV interferes with this induction process.

    • Herpes simplex viruses (HSV), belonging to the family of Herpesviridae, are large DNA viruses that encode more than 80 gene products (87). A prominent feature of HSV is their ability to establish latent or lytic infection, in which viral replication is regulated in a temporal cascade fashion. During HSV infection, the viruses interfere with innate immunity, particularly the type Ⅰ interferon (IFN) system (22, 42, 99). Type Ⅰ IFN responses are initiated by pattern recognition receptors (PRRs) that detect invariant molecular structures shared by pathogens of various origins (pathogen-associated molecular patterns, PAMPs).

      Toll-like Receptors (TLRs) are a group of PRRs residing on the cell plasma and/or endosomal mem-branes. Among them, TLR 3, 7, 8 and 9 recognize distinct types of virally-derived nucleic acids and activate signaling cascades that result in the induction of type Ⅰ IFNs (22, 42, 99, 105). Their expression patterns and magnitudes vary among tissues, with preferential expressions in immune cells. Of note, these TLRs are expressed, albeit differentially, in the epithelial cells from HSV-targeted oral, ocular and genital mucosa (31) as well as in the central nervous system (CNS) resident cells (3, 44). TLR7 and TLR8 recognize single-stranded RNA (ssRNA) (17, 29), whereas TLR9 senses unmethylated deoxycytidylate-phosphate-deoxyguanylate (CpG) DNA(30). TLR7/8/ 9 induction of type Ⅰ IFNs are mediated by an adaptor protein called myeloid differentiation primary response protein 88 (MyD88). Interacting with the Toll/ interleukin-1 receptor (TIR) domain of TLRs, MyD88 then recruits other adaptor proteins such as inter-leukin-1 receptor-associated kinases 1 and 4 (IRAK1 and IRAK4), and the tumor necrosis factor receptor-associated factor 6 (TRAF6), leading to the activation of IRF7, canonical ⅠκB kinase (IKK) α/β/γ complex and the MAPK cascade (57). IKKs result in activation of NF-κB while MAPK cascade activates the activating protein 1 (AP-1) (42, 61, 115). Consequently, activated IRF7, coordinated with activated transcription factors NF-κB and AP-1, results in the transcription of both IFN-α/β genes (57) as well as inflammatory cytokines (42). In contrast to TLR7/8/9, TLR3 is activated by double-stranded RNA (dsRNA) produced during viral replication (1). The induction of type Ⅰ IFNs by TLR3 is mediated through TIR domain-containing adaptor inducing IFN-β (TRIF) (75, 110). TRIF either interacts with two noncanonical IKKs, TANK-binding kinase 1 (TBK1) and IKKε (also known as IKK-i (96)) which activate IRF3, or recruits the TRAF6-TAK1-TAB 2 complex which activates NF-κB and AP-1 (38, 71, 93). Consequently, these transcription factors coordinate the transcriptional regulation of the IFN-β and IFN-α 4 gene specifically (107). Activated IRF3 is also able to induce a subset of IFN-stimulated genes (ISGs), in the absence of IFN production (5, 25, 66, 113), which contributes to antiviral responses.

      TLR-independent induction of type Ⅰ IFNs is mediated by cytoplasmic PRRs including retinoic acid-inducible gene Ⅰ (RIG-Ⅰ)/melanoma differentiation-associated gene 5 (MDA5) and DNA-dependent activator of IFN-regulatory factors (DAI). RIG-Ⅰ/MDA5 sense dsRNA during viral replication and transcription in infected cells (24, 114). Specifically, RIG-Ⅰ recognizes uncapped 5'-triphosphate ssRNA (33, 82), whereas MDA5 is the primary responder to polyinosine-polycytidylic acid (poly (I:C)), a synthetic dsRNA analogue (24, 40). RIG-Ⅰ/MDA5 associates with an adaptor protein, IFN-promoter stimulator 1 (IPS-1; also known as MAVS, VISA or CARDIF) (41, 62, 94, 109) which resides on the mitochondrial outer mem-brane and interacts with TRAF3 (26, 73). TRAF3 recruits and activates TBK1/IKKε (26), leading to activation of IRF3 and IRF7 (21, 95) and thus induction of type Ⅰ IFNs. A recent study shows that RIG-Ⅰ/IPS-1 is also involved in dsDNA-induced type Ⅰ IFN response (15). DAI is activated by binding to either B-form or Z-form DNA from a variety of sources, leading to type Ⅰ IFN gene expression through the activation of IRF3 and, probably, IRF7 (104). The signal transduction from DAI to IRFs seems to be mediated by direct interaction between DAI and the TBK1/IRF complex (104, 106). This pathway thereby provides a way for mounting type Ⅰ IFN responses against DNA viruses.

      It is notable that secreted type Ⅰ IFNs then initiate a positive feedback loop (91) by binding to type Ⅰ IFN receptors on the surface of neighboring cells in a paracrine and autocrine manner and trigger the Janus kinase-signal transducer and activator of transcription (Jak-STAT) pathway, leading to the expression of an array of ISGs. These ISGs, together with various cytokines and chemokines secreted by the infected cells, exhibit anti-viral, anti-proliferative and immuno-modulatory functions (22, 83, 99). Emerging evidence suggests that HSV interacts with the innate immune system in a complex way. Although HSV triggers type Ⅰ IFN responses, the large coding capacity of HSV enables it to express multiple viral products to interfere with these responses and thus to establish a successful infection. This review will focus on the induction and interference of innate immunity in terms of type Ⅰ IFNs by HSV.

    • The induction of IFN-α/β by HSV during early infection has been reported in mouse models early in the 1980's and mouse resistance to generalized infection was for both HSV-1 and-2 attributed to a genetically determined difference in the capacity for IFN-α/β production, which was shown to segregate with the X-chromosome (80, 116). Studies with gene knock-out mice and cells have demonstrated the essential roles of IRF3 in the induction of IFN-β/α4 and IRF7 in the activation of IFNα-non4 subtypes by viruses in fibroblasts and epithelial cells (4, 91, 92). Later, it was reported that infection of human fibrob-lasts with both HSV-1 and human cytomegalovirus (HCMV) activated IRF3 and induced type Ⅰ IFNs (84). HSV-1 induction of IFN-α/β was also observed in infected human corneal epithelial cells (HCECs) (49), a clinically and physiologically relevant cell type. In addition, infection of murine peritoneal cells by HSV-2 induces a rapid production of IFN-α/β within 4 h of infection (55). It has been shown that HSV entry alone was sufficient to induce innate antiviral responses in human fibroblasts (16, 66). Consistent with this, UV-inactivated HSV-1 that is competent for entry but not for replication is able to induce IFN-α production in peripheral blood mononuclear cells (PBMCs) (89). This induction process also requires IRF3 (16, 72). However, the receptor and precise mechanism for the induction of innate antiviral response by HSV virus entry is still unclear, although phosphoinositide 3-kinase (PI3 kinase) family mem-bers have been implicated to play a role during this event (72). The envelope glycoproteins of HSV-1, particularly glycoprotein D (gD), has been shown to induce IFN-α production in PBMCs (2, 51) through a mechanism involving unknown intracellular signaling and engagement of chemokine receptors CCR3 and CXCR4 (2). The mechanism by which these chemo-kine receptors mediate HSV induction of type Ⅰ IFNs remains unclear. In light of this, HSV gD is shown to be a strong inducer of IFN-α/β in PBMC-derived dendritic cells and mannose receptors on target cells are indicated to be involved (89). In contrast to the stimulating role of gD in PBMCs, HSV-1 induction of IFN-α/β in macrophages does not rely on gD, sug-gesting that other components of HSV may function as the inducer (56).

      A recent study demonstrated that HSV activation of TLR2 contributes to lethal encephalitis (50). TLR2 is an extracellular PRR sensing microbial lipopeptides and mainly induces inflammatory cytokine production in a MyD88-mediated NF-κB-dependent manner (105). The fact that HSV-1 caused a higher mortality in wild type mice than in TLR2-/- mice revealed a detrimental effect of TLR2-mediated cytokine overpro-duction (50). Intriguingly, a recent study, in which a cell line expressing four HSV-1 entry glycoproteins (gB, gD, and the heterodimer gHgL) was used to stimulate monocyte-derived DCs, demonstrated that the collective induction of type Ⅰ IFNs in DCs by HSV-1 glycoprotein complex was TLR2-independent (86). This thus supports a model of two parallel pathways in DCs, one mediated by TLR2 recognition of an unidentified agonist (s) from HSV-1 and the other one mediated by undefined receptor (s) on DC surface recognizing the HSV-1 glycoprotein complex. HSV genomic DNA is normally unmethylated and very GC-rich, thereby containing abundant CpG motifs (88, 118) which is the ligand for TLR9(30). Genomic HSV-2 DNA has been shown to trigger TLR9 and mediate activation through a MyD88-dependent endocytic pathway leading to type Ⅰ IFN production in plasmacytoid dendritic cells (pDCs) (54). These are a restricted subset of DCs that are equipped with high levels of TLR7/9 and specialize in secreting copious type Ⅰ IFNs, particularly IFN-α, after stimulation with viral nucleic acids (42). The strict requirement for TLR9 in IFN-α secretion in-duced by HSV-2 was further confirmed in vivo (54). In line with this, a study using KOS HSV-1 recom-binant virus showed that HSV induction of type Ⅰ IFNs in pDCs was indeed mediated by the TLR9/ MyD88 pathway in vitro (46). In support of the aforementioned studies using laboratory strains of HSV, clinically isolated TLR2-activating HSV-1 and HSV-2 subspecies have been shown to induce the production of inflammatory cytokines and type Ⅰ IFNs by a sequential recognition mechanism of TLR2 → TLR9 in conventional DCs (cDCs) (90). However, the requirement of TLR9 for HSV induction of type Ⅰ IFNs seems cell type-specific. Mice lacking either MyD88 or TLR9 are still capable of controlling HSV-1 replication in vivo after local infection, indicating that TLR9-and MyD88-independent path-ways in cells can compensate for this host defect (46). Indeed, TLR9-independent type Ⅰ IFN response to HSV infection has been reported to exist in pDCs, cDCs, macrophages and fibroblasts (32, 46, 56, 85), suggesting that TLR9 only partially contributes to type Ⅰ IFN response to HSV in cell types other than pDCs.

      Recent studies suggest that the double-stranded structure of DNA possesses immunomodulatory effects by TLR-independent pathways when introduced into the cytosol (34). Introduction of B-from dsDNA into the cytosol induces TLR/MyD88/TRIF-independent, TBK/IKKε-and IRF3-mediated production of type Ⅰ IFNs, especially IFN-β, and their inducible genes in both immune and non-immune cells (15, 35, 36, 74, 97, 100, 104). These studies suggest that the double-stranded HSV genomic DNA is recognized by cyto-plasmic receptors, resulting in type Ⅰ IFN production. However, the mechanisms of DNA virus recognition are not well understood to date. Thus far only one cytoplasmic DNA sensor, named DAI, has been discovered and shown to contribute to HSV-1-mediated type Ⅰ IFN gene induction (104). Interes-tingly, mouse embryonic fibroblasts (MEFs) deficient in IPS-1, are unable to produce type Ⅰ IFNs in response to HSV (85), indicating the possible involve-ment of dsRNA sensors RIG-Ⅰ/MDA5. In line with this, Cheng et al. (15) showed that HSV has the potential to trigger dsDNA signaling pathways in a RIG-Ⅰ/IPS-1-dependent manner leading to IFN-β production in human hepatoma cells. Nevertheless, the notion that IPS-1 and RIG-Ⅰ are required for cytosolic dsDNA-triggered type Ⅰ IFN production is contradictory to previous findings (48, 77, 102). Current knowledge suggests that type Ⅰ IFN expression activated by either cytosolic dsDNA or HSV-1 particle (containing genomic DNA) entry share the same pathway downstream of DAI and RIG-Ⅰ/IPS-1 that is mediated by TBK1/IKKε and IRF3. The role of RIG-Ⅰ/IPS-1 in HSV induction of type Ⅰ IFNs still awaits further investigation.

      Recently, a genetic etiology for herpes simplex virus-1 encephalitis (HSE) in two children with auto-somal recessive deficiency in the intracellular protein UNC-93B was identified (7), shedding light on the possible link between TLR3 and HSV-1 pathogenesis. Human UNC-93B, which has orthologues in several distantly related species, is an endoplasmic reticulum protein with 12 membrane-spanning domains control-ling TLR3, TLR7, TLR8 (TLR8 is absent in mice) and TLR9 response by physically interacting with these TLRs (117). When challenged with either TLR3 agonist or HSV-1, skin-derived fibroblasts from the UNC-93B-deficient HSE patients cannot produce IFN-β and IFN-λ, nor are they able to mount immune responses to TLR7/8/9 agonists (7). It has been also reported that patients deficient in IRAK4, an adaptor involved in all TLR-mediated signaling except for TLR3 and TLR4, a LPS sensor (81), are not suscep-tible to most viral infections, indicating that the type Ⅰ IFN response through TLR7, 8, and 9 is dispensable for protective immunity to most microbes (47, 112). Particularly, IRAK-4 deficient patients whose TLR3 signaling is still intact are not susceptible to HSE. Given that UNC-93B deficient patients whose TLR3-mediated pathway is impaired are susceptible to HSE (7), these data implicate that TLR3 may play an important role in HSE resistance. In addition, two unrelated patients with HSE have been identified carrying the same heterozygous dominant negative mutation in a TLR3 allele (20). Fibroblasts derived from the patients displayed a significantly diminished responsiveness to TLR3 agonist and HSV-1 challenge in terms of abnormally weak type Ⅰ IFN production and markedly increased susceptibility to HSV-1 infection. Furthermore, stimulated with several viruses, these TLR3 heterozygous fibroblasts produced type Ⅰ IFNs in response to most infections, except HSV-1 and vesicular stomatitis virus (VSV), consistent with the natural resistance of these patients to most viruses other than HSV-1. Thus, this establishes the essential role of TLR3 and the importance of TLR3-UNC93B-signaling pathway for primary immunity to HSV-1 in CNS, although human TLR3 appears to be redundant in host defenses to most other microbes. Nonetheless, the question remains how HSV-1, a DNA virus, triggers dsRNA sensor TLR3. This may be answered by the fact that HSV-1 generates dsRNA intermediates via symmetrical transcription (37, 108).

    • Facing the intricate yet delicate host immune system consisting of innate and adaptive immunity, many viruses have developed multiple cunning strategies to evade host immune attacks at different stages. HSV is one of the most successful human pathogens, interfering with the antiviral responses using versatile strategies. For instance, γ134.5, one critical virulence factor of HSV-1, is able to counteract the antiviral response initiated by double-stranded RNA-activated protein kinase (PKR) (13, 28), a type Ⅰ IFN inducible cellular enzyme which dictates translational arrest and apoptosis by phosphorylating the α-subunit of the eukaryotic translation initiation factor 2 (eIF2α) upon dsRNA recognition (23). HSV-1 infection also causes the disappearance of Jak1 and Stat2 molecules partially by virion host shutoff (vhs) protein (11), an HSV encoded mRNA-specific RNase (98), leading to inhibition of Jak/Stat-mediated IFN signaling and subsequent robust antiviral response.

      It has been shown that virus entry of HSV triggers the rapid activation of IRF3 leading to innate immune responses including production of IFN-β and a subset of ISGs (16, 66, 70, 72, 84). However, restriction or inhibition of these immediate early responses after HSV viral replication has also been observed (16, 66, 70), suggesting that HSV-1 encodes viral products that inhibit the early IFN response mediated by IRF3. Infected cell protein 0 (ICP0), an immediate-early protein of HSV-1, is the first viral component whose antagonistic function has been described. HSV-1 bearing deletion in the ICP0 gene was found hypersensitive to IFN while IFN only marginally reduced the replication of wild-type (wt) HSV-1 (65). Expression of ICP0 was then shown to suppress the induction of ISGs during early HSV infection (19). Further investigation revealed that the ICP0 RING finger domain inhibits IRF3-and IRF7-mediated activation of ISGs and this inhibition process involves the proteasome, although no proteasomal degradation of known IRF3 signaling components can be observed (53). A study with immunofluorescence staining tech-niques revealed that ICP0 sequesters activated IRF-3 and CBP/p300, critical transcriptional coactivators for efficient IRF3-mediated transcription of IFN-β and certain ISGs (107, 111, 113), to nuclear structures away from the host chromatin at early times post HSV-1 infection, thus resulting in reduced transcri-ption of IFN-β/ISGs and an inhibition of the host response (60). However, the presence of ICP0 promoted proteasome-dependent degradation of activated IRF3 (60), which is paradoxical to Mossman's report (53). The discrepancy between these studies might be due not only to the sensitivity of the techniques used but also to different focuses-one studied the nuclear portion of IRF3 (activated form) (60) while the other evaluated the entire endogenous level of IRF3 (53).

      Although ICP0 has been shown to play a pivotal role in antagonizing innate immune responses, studies using an ICP0 mutant virus suggest that multiple viral products/mechanisms may exist to block HSV-induced IRF3-mediated IFN production(59, 60). In fact, the critical virulence factor γ134.5 has been linked to IFN resistance of HSV several years ago. One study using mice lacking IFN signaling due to the knockout of IFN-α/β receptor (IFN-α/β/R-/-) showed that the replication of γ134.5-null virus in normal mice was strongly inhibited while its replication in IFN-α/βR-/- mice was restored to almost wild type level, and a γ134.5 rescue marker can largely suppress the phenotype of the γ234.5-null virus (52). Dissection of γ134.5 protein also supports the notion that it renders HSV resistant to IFN response (10, 12), which seems to be correlated with γ134.5's anti-PKR function, a well characterized feature of the γ134.5 protein (13, 28). In contrast, a γ134.5 second-site suppressor mutant, which harbors a null γ134.5 gene and a second-site deletion causing early expression of the late gene US11 (27, 64), suppresses the phenotype of the γ134.5-null mutant by inhibiting PKR activity in vitro (8, 9) but remains as severely attenuated as the γ134.5-null mutant in vivo (63). This implicates additional functions of the γ134.5 protein other than anti-PKR to antagonize host responses. In accordance with this, counteracting PKR by γ134.5 was found insufficient for overcoming host responses and for efficient viral replication (14). Microarray analysis in mouse embryonic fibroblasts (MEFs) showed that antiviral genes, including IFN-β and a subset of ISGs, in γ134.5-null virus-infected cells were differentially regulated compared to those in wt HSV-1-infected cells during the early phase of infection (78). This result implies that γ134.5 may modulate type Ⅰ IFN induction either directly or indirectly.

      Other HSV viral components such as immediate-early protein ICP27 and mRNA-specific RNase vhs also appear to be able to interfere with virus induction of type Ⅰ IFNs/ISGs. It has been reported that ICP27-deficient virus elicits a much higher production of IFN-α/β and inflammatory cytokines in monocyte-derived macrophages and DCs, and activates both NF-κB and IRF3 more potently than wt HSV-1 (58). Other studies reveal that ICP27 also functions in both the suppression of NF-κB activity by stabilizing ⅠκB, the inhibitor of NF-κB (43), and the interruption of IFN signaling pathways downstream of IFN induction by down-regulating the phosphorylation of STAT1, a component of the Jak-Stat pathway (39). However, whether ICP27 exerts a direct or indirect impact on type Ⅰ IFN production and what the precise mechanism (s) is are still open questions awaiting further investigation. Although vhs protein renders HSV resistant to IFN response (69, 76, 101), the inhibitory effect of vhs on IFN response is contro-versial (53, 103). A study using IFN-α/β receptor knockout mice suggests that HSV-2 vhs down-regulates type Ⅰ IFN-mediated responses in vivo (68). But whether vhs interferes with the induction of type Ⅰ IFNs directly is not clarified. A clue was provided by the finding that MEFs infected with vhs-null HSV-2 produced > 50-fold more IFN-α/β than cells infected with wt HSV-2 and vhs rescue virus (18). The precise mechanism by which the presence of vhs suppresses type Ⅰ IFN production remains to be defined. None-theless, with potent RNase activity, vhs may limit the amount of mRNAs encoding type Ⅰ IFNs and ISGs, thereby inhibiting their synthesis. A study using an HSV-2 point mutant that synthesizes full-length vhs protein lacking RNase activity lends support to this idea (45). It is also reported that cells infected with vhs defective HSV display higher levels of viral RNA (79), which leads to another hypothesis that the loss of vhs allows accumulation of double-stranded viral RNA, a potent PAMP which can trigger cellular dsRNA sensor TLR3/RIG-Ⅰ/MDA5 and finally induce a high production of type Ⅰ IFNs and ISGs.

    • Innate immunity constitutes the first line of host defenses against invading pathogens. The arsenal of type Ⅰ IFN responses represents the critical part of innate immunity. Several TLRs and cytosolic sensors recognizing virus-associated molecular patterns have been discovered and their molecular signaling path-ways leading to the production of type Ⅰ IFNs and ISGs have been illustrated, while the list of PRRs, their agonists as well as cellular mediators is still growing. HSV infections can induce the production of type Ⅰ IFNs, ISGs and inflammatory cytokines, especially during early phases of acute infection, via multiple mechanisms in a cell type-specific manner. Several components of HSV such as envelope gly-coproteins, genomic DNAs and replication dsRNA intermediates are all indicated to be inducers of type Ⅰ IFNs. Concordantly, cellular receptors TLR2, TLR9/ DAI and TLR3/RIG-Ⅰ/MAD5 that sense lipoproteins, DNAs and dsRNAs have all been reported to be involved in HSV-induced innate responses, although molecular mechanisms remain elusive. Since these PRRs are expressed, albeit differentially, by almost all cell types, the induction of type Ⅰ IFNs by HSV infection predictably involves multiple pathways simultaneously. However, in order to establish in-fection, HSV has developed multiple strategies to counteract these host responses, making it one of the most successful human pathogens. HSV encodes several proteins such as ICP0, ICP27 and vhs, which may function cooperatively against the induction of type Ⅰ IFNs. In addition, HSV interferes with IFN signaling, IFN-inducible product (including ISGs, PKR etc.) -mediated responses and adaptive immunity (6, 67), which is not discussed in this review. Therefore, the battle between HSV and the host immune system is a complex, dynamic process, the outcome of which will determine pathogenesis. Further understanding of this process will aid us to find effective solutions for prevention and cure of diseases caused by HSV.

    Reference (118) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return