Citation: Gerardo Abenes, Fenyong Liu. Pathogenetic Consequences of Cytomegalovirus-Host Co-evolution .VIROLOGICA SINICA, 2008, 23(6) : 438-448.  http://dx.doi.org/10.1007/s12250-008-3003-2

Pathogenetic Consequences of Cytomegalovirus-Host Co-evolution

  • Corresponding author: Fenyong Liu, liu_fy@uclink4.berkeley.edu
  • Received Date: 09 September 2008
    Accepted Date: 24 September 2008
    Available online: 01 December 2008

    Fund Project: This work was, in part, supported by US Public Health Service DE014145This work was, in part, supported by US Public Health Service AI041927This work was, in part, supported by US Public Health Service AI050468This work was, in part, supported by US Public Health Service DE016813

  • Co-evolution has been shown to result in an adaptive reciprocal modification in the respective behaviors of interacting populations over time. In the case of host-parasite co-evolution, the adaptive behavior is most evident from the reciprocal change in fitness of host and parasite-manifested in terms of pathogen survival versus host resistance. Cytomegaloviruses and their hosts represent a pairing of populations that has co-evolved over hundreds of years. This review explores the pathogenetic consequences emerging from the behavioral changes caused by co-evolutionary forces on the virus and its host.

  • 加载中
    1. Adler S P, Marshall B. 2007. Cytomegalovirus in-fections. Pediatr Rev, 28: 92-100.
        doi: 10.1542/pir.28-3-92

    2. Ahn K, Angulo A, Ghazal P, et al. 1996. Human cytomegalovirus inhibits antigen presentation by a sequential multistep process. Proc Natl Acad Sci USA, 93: 10990-10995.
        doi: 10.1073/pnas.93.20.10990

    3. Ahn K, Gruhler A, Galocha B, et al. 1997. The ER-luminal domain of the HCMV glycoprotein US6 inhibits peptide translocation by TAP. Immunity, 6: 613-621.
        doi: 10.1016/S1074-7613(00)80349-0

    4. Arase H, Mocarski E S, Campbell A E, et al. 2002. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science, 296: 1323-1326.
        doi: 10.1126/science.1070884

    5. Askin D F. 2004. Intrauterine infections. Neonatal Netw, 23: 23-30.
        doi: 10.1891/0730-0832.23.5.23

    6. Atreya I, Atreya R, Neurath M F. 2008. NF-kappaB in inflammatory bowel disease. J Intern Med, 263:591-596.
        doi: 10.1111/j.1365-2796.2008.01953.x

    7. Bale J F. 2002. Congenital infections. Neurol Clin, 20:1039-1060.
        doi: 10.1016/S0733-8619(02)00014-2

    8. Bason C, Corrocher R, Lunardi C, et al. 2003. Interaction of antibodies against cytomegalovirus with heat-shock protein 60 in pathogenesis of atherosclerosis. Lancet, 362: 1971-1977.
        doi: 10.1016/S0140-6736(03)15016-7

    9. Basta S, Bennink J R. 2003. A survival game of hide and seek: cytomegaloviruses and MHC class Ⅰ antigen presentation pathways. Viral Immunol, 16: 231-242.
        doi: 10.1089/088282403322396064

    10. Bauer D, Tampe R. 2002. Herpes viral proteins blocking the transporter associated with antigen processing TAP--from genes to function and structure. Curr Top Micro-biol Immunol, 269: 87-99.

    11. Bego M G, St Jeor S. 2006. Human cytomegalovirus infection of cells of hematopoietic origin: HCMV-induced immunosuppression, immune evasion, and latency. Exp Hematol, 34: 555-570.
        doi: 10.1016/j.exphem.2005.11.012

    12. Beisser P S, Goh C S, Cohen F E, et al. 2002. Viral chemokine receptors and chemokines in human cytomega-lovirus trafficking and interaction with the immune system. CMV chemokine receptors. Curr Top Microbiol Im-munol, 269: 203-34.

    13. Bentz G L, Yurochko A D. 2008. Human CMV infection of endothelial cells induces an angiogenic response through viral binding to EGF receptor and beta1 and beta3 integrins. Proc Natl Acad Sci USA, 105: 5531-5536.
        doi: 10.1073/pnas.0800037105

    14. Boomker J M, de Jong E K, de Leij L F, et al. 2006. Chemokine scavenging by the human cytomegalovirus chemokine decoy receptor US28 does not inhibit monocyte adherence to activated endothelium. Antiviral Res, 69: 124-127.
        doi: 10.1016/j.antiviral.2005.11.003

    15. Boomker J M, van Luyn M J, The T H, et al. 2005. US28 actions in HCMV infection: lessons from a versa-tile hijacker. Rev Med Virol, 15: 269-282.
        doi: 10.1002/(ISSN)1099-1654

    16. Braud V M, Tomasec P, Wilkinson G W. 2002. Viral evasion of natural killer cells during human cytomega-lovirus infection. Curr Top Microbiol Immunol, 269: 117-129.

    17. Bryant P, Morley C, Garland S, et al. 2002. Cytomega-lovirus transmission from breast milk in premature babies: does it matter? Arch Dis Child Fetal Neonatal Ed, 87: F75-77.
        doi: 10.1136/fn.87.2.F75

    18. Candore G, Balistreri C R, Colonna-Romano G, et al. 2008. Immunosenescence and anti-immunosenescence therapies: the case of probiotics. Rejuvenation Res, 11: 425-432.
        doi: 10.1089/rej.2008.0662

    19. Chalupny N J, Rein-Weston A, Dosch S, et al. 2006. Down-regulation of the NKG2D ligand MICA by the human cytomegalovirus glycoprotein UL142. Biochem Biophys Res Commun, 346: 175-181.
        doi: 10.1016/j.bbrc.2006.05.092

    20. Chang M, Pan M R, Chen D Y, et al. 2006. Human cytomegalovirus pp65 lower matrix protein: a humoral immunogen for systemic lupus erythematosus patients and autoantibody accelerator for NZB/W F1 mice. Clin Exp Immunol, 143: 167-179.
        doi: 10.1111/cei.2006.143.issue-1

    21. Chapman T L, Heikeman A P, Bjorkman P J. 1999. The inhibitory receptor LIR-1 uses a common binding interac-tion to recognize class Ⅰ MHC molecules and the viral homolog UL18. Immunity, 11: 603-613.
        doi: 10.1016/S1074-7613(00)80135-1

    22. Cosman D, Mullberg J, Sutherland C L, et al. 2001. ULBPs, novel MHC class Ⅰ-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity, 14: 123-133.
        doi: 10.1016/S1074-7613(01)00095-4

    23. Criscuoli V, Rizzuto M R, Cottone M. 2006. Cytomega-lovirus and inflammatory bowel disease: is there a link? World J Gastroenterol, 12: 4813-4818.

    24. Damato E G, Winnen C W. 2002. Cytomegalovirus infection: perinatal implications. J Obstet Gynecol Neo-natal Nurs, 31: 86-92.
        doi: 10.1111/j.1552-6909.2002.tb00026.x

    25. Davison A J, Dolan A, Akter P, et al. 2003. The human cytomegalovirus genome revisited: comparison with the chimpanzee cytomegalovirus genome. J Gen Virol, 84: 17-28.
        doi: 10.1099/vir.0.18606-0

    26. DeFilippis V R. 2007. Induction and evasion of the type Ⅰ interferon response by cytomegaloviruses. Adv Exp Med Biol, 598:309-24.
        doi: 10.1007/978-0-387-71767-8

    27. Diaz F, Urkijo J C, Mendoza F, et al. 2006. Systemic lupus erythematosus associated with acute cytomega-lovirus infection. J Clin Rheumatol, 12: 263-264.
        doi: 10.1097/01.rhu.0000239832.74804.17

    28. Dimitroulia E, Spanakis N, Konstantinidou A E, et al. 2006. Frequent detection of cytomegalovirus in the in-testine of patients with inflammatory bowel disease. Inflamm Bowel Dis, 12: 879-884.
        doi: 10.1097/01.mib.0000231576.11678.57

    29. Dugan G E, Hewitt E W. 2008. Structural and Func-tional Dissection of the Human Cytomegalovirus Immune Evasion Protein US6. J Virol, 82: 3271-3282.
        doi: 10.1128/JVI.01705-07

    30. Dunn C, Chalupny N J, Sutherland C L, et al. 2003. Human cytomegalovirus glycoprotein UL16 causes intracellular sequestration of NKG2D ligands, protecting against natural killer cell cytotoxicity. J Exp Med, 197: 1427-1439.
        doi: 10.1084/jem.20022059

    31. Dunn W, Chou C, Li H, et al. 2003. Functional profiling of a human cytomegalovirus genome. Proc Natl Acad Sci USA, 100: 14223-14228.
        doi: 10.1073/pnas.2334032100

    32. Editorial Comments. 2004. Cytomegalovirus. Am J Transplant, 4 Suppl 10:51-58.

    33. Emery V C. 2001. Cytomegalovirus and the aging popu-lation. Drugs Aging, 18: 927-933.
        doi: 10.2165/00002512-200118120-00004

    34. Emery V C. 2001. Investigation of CMV disease in immunocompromised patients. J Clin Pathol, 54: 84-88.
        doi: 10.1136/jcp.54.2.84

    35. Franceschi C. 2007. Inflammaging as a major charac-teristic of old people: can it be prevented or cured? Nutr Rev, 65: S173-176.

    36. Franceschi C, Bonafe M. 2003. Centenarians as a model for healthy aging. Biochem Soc Trans 31: 457-61.
        doi: 10.1042/bst0310457

    37. Froberg M K. 2004. Review: CMV escapes! Ann Clin Lab Sci, 34: 123-130.

    38. Gewurz B E, Gaudet R, Tortorella D, et al. 2001. Antigen presentation subverted: Structure of the human cytomegalovirus protein US2 bound to the class Ⅰ mo-lecule HLA-A2. Proc Natl Acad Sci USA, 98: 6794-6799.
        doi: 10.1073/pnas.121172898

    39. Giunta S. 2006. Is inflammaging an auto[innate] immunity subclinical syndrome? Immun Ageing, 3: 12.
        doi: 10.1186/1742-4933-3-12

    40. Gold E, Nankervis G A. 1976. Cytomegalovirus. In: Viral infections of humans: epidemiology and control (Evans A S ed.), Plenum Press: New York, p 143-161.

    41. Goldmacher V S. 2005. Cell death suppression by cytomegaloviruses. Apoptosis, 10: 251-265.
        doi: 10.1007/s10495-005-0800-z

    42. Goldmacher V S. 2002. vMIA, a viral inhibitor of apo-ptosis targeting mitochondria. Biochimie, 84: 177-185.
        doi: 10.1016/S0300-9084(02)01367-6

    43. Hengel H, Koopmann J O, Flohr T, et al. 1997. A viral ER-resident glycoprotein inactivates the MHC-encoded peptide transporter.Immunity, 6: 623-632.
        doi: 10.1016/S1074-7613(00)80350-7

    44. Hommes D W, Sterringa G, van Deventer S J, et al. 2004. The pathogenicity of cytomegalovirus in inflam-matory bowel disease: a systematic review and evidence-based recommendations for future research. Inflamm Bowel Dis, 10: 245-250.
        doi: 10.1097/00054725-200405000-00011

    45. Hooper M, Kallas E G, Coffin D, et al. 1999. Cytomega-lovirus seropositivity is associated with the expansion of CD4+CD28- and CD8+CD28- T cells in rheumatoid arthritis. J Rheumatol, 26: 1452-1457.

    46. Hrycek A, Kusmierz D, Mazurek U, et al. 2005. Human cytomegalovirus in patients with systemic lupus ery-thematosus. Autoimmunity, 38: 487-491.
        doi: 10.1080/08916930500285667

    47. Hussein K, Hayek T, Yassin K, et al. 2006. Acute cytomegalovirus infection associated with the onset of inflammatory bowel disease. Am J Med Sci, 331: 40-43.
        doi: 10.1097/00000441-200601000-00012

    48. Jarvis M A, Borton J A, Keech A M, et al. 2006. Human cytomegalovirus attenuates interleukin-1beta and tumor necrosis factor alpha proinflammatory signaling by inhibition of NF-kappaB activation. J Virol, 80: 5588-5598.
        doi: 10.1128/JVI.00060-06

    49. Jarvis M A, Nelson J A. 2002. Human cytomegalovirus persistence and latency in endothelial cells and macrop-hages.Curr Opin Microbiol, 5:403-407.
        doi: 10.1016/S1369-5274(02)00334-X

    50. Jarvis M A, Nelson J A. 2002. Mechanisms of human cytomegalovirus persistence and latency. Front Biosci, 7:d1575-1582.
        doi: 10.2741/A862

    51. Jenkins C, Abendroth A, Slobedman B. 2004. A novel viral transcript with homology to human interleukin-10 is expressed during latent human cytomegalovirus infection. J Virol, 78: 1440-1447.
        doi: 10.1128/JVI.78.3.1440-1447.2004

    52. Johnson D C, Hegde N R. 2002. Inhibition of the MHC class Ⅱ antigen presentation pathway by human cytomega-lovirus. Curr Top Microbiol Immunol, 269: 101-115.

    53. Jones T R, Wiertz E J, Sun L, et al. 1996. Human cytomegalovirus US3 impairs transport and maturation of major histocompatibility complex class Ⅰ heavy chains. Proc Natl Acad Sci USA, 93: 11327-11333.
        doi: 10.1073/pnas.93.21.11327

    54. Jurak I, Brune W. 2006. Induction of apoptosis limits cytomegalovirus cross-species infection. Embo J, 25: 2634-2642.
        doi: 10.1038/sj.emboj.7601133

    55. Kanapeckiene V, Kalibatas J, Redaitiene E, et al. 2007. The association between cytomegalovirus infection and aging process. Medicina (Kaunas), 43: 419-424.

    56. Kavanagh D G, Hill A B. 2001. Evasion of cytotoxic T lymphocytes by murine cytomegalovirus. Semin Im-munol, 13: 19-26.
        doi: 10.1006/smim.2001.0292

    57. Kerrey B T, Morrow A, Geraghty S, et al. 2006. Breast milk as a source for acquisition of cytomegalovirus (HCMV) in a premature infant with sepsis syndrome: detection by real-time PCR. J Clin Virol, 35: 313-316.
        doi: 10.1016/j.jcv.2005.09.013

    58. Khan N. 2007. The immunological burden of human cytomegalovirus infection. Arch Immunol Ther Exp (Warsz), 55: 299-308.
        doi: 10.1007/s00005-007-0037-3

    59. Khan N, Hislop A, Gudgeon N, et al. 2004. Herpes-virus-specific CD8 T cell immunity in old age: cytomega-lovirus impairs the response to a coresident EBV infection. J Immunol, 173: 7481-7489.
        doi: 10.4049/jimmunol.173.12.7481

    60. Khan N, Shariff N, Cobbold M, et al. 2002. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J Immunol, 169: 1984-1992.
        doi: 10.4049/jimmunol.169.4.1984

    61. Khoshnevis M, Tyring S K. 2002. Cytomegalovirus infections. Dermatol Clin, 20:291-299, vii.
        doi: 10.1016/S0733-8635(01)00007-9

    62. Kinney J S, Onorato I M, Stewart J A, et al. 1985. Cytomegaloviral infection and disease. J Infect Dis, 151: 772-774.
        doi: 10.1093/infdis/151.5.772

    63. Kojima T, Watanabe T, Hata K, et al. 2006. Cytomega-lovirus infection in ulcerative colitis. Scand J Gastroen-terol, 41: 706-711.
        doi: 10.1080/00365520500408584

    64. Koutouzov S, Jeronimo A L, Campos H, et al. 2004. Nucleosomes in the pathogenesis of systemic lupus ery-thematosus.Rheum Dis Clin North Am, 30: 529-558, ix.
        doi: 10.1016/j.rdc.2004.04.001

    65. Krmpotic A, Bubic I, Polic B, et al. 2003. Pathogenesis of murine cytomegalovirus infection. Microbes Infect, 5: 1263-1277.
        doi: 10.1016/j.micinf.2003.09.007

    66. Landolfo S, Gariglio M, Gribaudo G, et al. 2003. The human cytomegalovirus. Pharmacol Ther, 98: 269-297.
        doi: 10.1016/S0163-7258(03)00034-2

    67. Lehner P J, Karttunen J T, Wilkinson G W, et al. 1997. The human cytomegalovirus US6 glycoprotein inhibits transporter associated with antigen processing-dependent peptide translocation. Proc Natl Acad Sci USA, 94: 6904-6909.
        doi: 10.1073/pnas.94.13.6904

    68. Lenac T, Arapovic J, Traven L, et al. 2008. Murine cytomegalovirus regulation of NKG2D ligands. Med Microbiol Immunol, 197: 159-166.
        doi: 10.1007/s00430-008-0080-7

    69. Lin A, Xu H, Yan W. 2007. Modulation of HLA expres-sion in human cytomegalovirus immune evasion. Cell Mol Immunol, 4: 91-98.

    70. Lodoen M B, Abenes G, Umamoto S, et al. 2004. The cytomegalovirus m155 gene product subverts natural killer cell antiviral protection by disruption of H60-NKG2D interactions. J Exp Med, 200: 1075-1081.
        doi: 10.1084/jem.20040583

    71. Loenen W A, Bruggeman C A, Wiertz E J. 2001. Im-mune evasion by human cytomegalovirus: lessons in immunology and cell biology. Semin Immunol, 13: 41-49.
        doi: 10.1006/smim.2001.0294

    72. Lunardi C, Bason C, Corrocher R, et al. 2005. Induction of endothelial cell damage by hCMV molecular mimicry. Trends Immunol, 26: 19-24.
        doi: 10.1016/j.it.2004.10.009

    73. Lunardi C, Bason C, Navone R, et al. 2000. Systemic sclerosis immunoglobulin G autoantibodies bind the human cytomegalovirus late protein UL94 and induce apoptosis in human endothelial cells. Nat Med, 6: 1183-1186.
        doi: 10.1038/80533

    74. Lunardi C, Dolcino M, Peterlana D, et al. 2006. Antibodies against human cytomegalovirus in the patho-genesis of systemic sclerosis: a gene array approach. PLoS Med, 3: e2.

    75. Lunardi C, Dolcino M, Peterlana D, et al. 2007. Endothelial cells' activation and apoptosis induced by a subset of antibodies against human cytomegalovirus: relevance to the pathogenesis of atherosclerosis. PLoS ONE, 2: e473
        doi: 10.1371/journal.pone.0000473

    76. Mattey D L, Dawes P T, Nixon N B, et al. 2004. Increased levels of antibodies to cytokeratin 18 in patients with rheumatoid arthritis and ischaemic heart disease. Ann Rheum Dis, 63: 420-425.
        doi: 10.1136/ard.2003.008011

    77. McCormick A L. 2008. Control of apoptosis by human cytomegalovirus. Curr Top Microbiol Immunol, 325: 281-295.

    78. Mehraein Y, Lennerz C, Ehlhardt S, et al. 2004. Latent Epstein-Barr virus (EBV) infection and cytomegalovirus (CMV) infection in synovial tissue of autoimmune chronic arthritis determined by RNA-and DNA-in situ hybridization. Mod Pathol, 17: 781-789.
        doi: 10.1038/modpathol.3800119

    79. Mocarski E S. 2002. Immunomodulation by cytomega-loviruses: manipulative strategies beyond evasion. Trends Microbiol, 10:332-339.
        doi: 10.1016/S0966-842X(02)02393-4

    80. Mocarski E S. Courcelle, C. T. Cytomegalovirus and their replication, In: Fields' Virology (Knipe D M, Howley P M, ed. ), Lippincott : Philadelphia, p2629-2673.

    81. Murphy E, Rigoutsos I, Shibuya T, et al. 2003. Reevaluation of human cytomegalovirus coding potential. Proc Natl Acad Sci USA, 100: 13585-13590.
        doi: 10.1073/pnas.1735466100

    82. Murphy E, Yu D, Grimwood J, et al. 2003. Coding potential of laboratory and clinical strains of human cytomegalovirus. Proc Natl Acad Sci USA, 100: 14976-14981.
        doi: 10.1073/pnas.2136652100

    83. Namboodiri A M, Rocca K M, Kuwana M, et al. 2006. Antibodies to human cytomegalovirus protein UL83 in systemic sclerosis. Clin Exp Rheumatol, 24: 176-178.

    84. Noyola D E, Valdez-Lopez B H, Hernandez-Salinas A E, et al. 2005. Cytomegalovirus excretion in children attending day-care centers. Arch Med Res, 36: 590-593.
        doi: 10.1016/j.arcmed.2005.03.045

    85. Pandey J P. 2004. Immunoglobulin GM genes and IgG antibodies to cytomegalovirus in patients with systemic sclerosis. Clin Exp Rheumatol, 22: S35-7.

    86. Pass R F. 2001. Cytomegalovirus, In: Fields' Virology (Knipe D M, Howley P M ed. ), Lippincott: Philadelphia, p2675-2705.

    87. Pass R F. 2002. Cytomegalovirus infection. Pediatr Rev, 23: 163-170.
        doi: 10.1542/pir.23-5-163

    88. Pass R F. 1985. Epidemiology and transmission of cytomegalovirus. J Infect Dis, 152: 243-248.
        doi: 10.1093/infdis/152.2.243

    89. Pawelec G, Gouttefangeas C. 2006. T-cell dysregulation caused by chronic antigenic stress: the role of CMV in immunosenescence? Aging Clin Exp Res, 18: 171-173.
        doi: 10.1007/BF03327436

    90. Pawelec G, Larbi A. 2008. Immunity and ageing in man: Annual Review 2006/2007. Exp Gerontol, 43: 34-38.

    91. Peichl P, Scriba M, Haberhauer G, et al. 1988. Selective binding of rheumatoid factors to antigen structures of cytomegalovirus (CMV). Scand J Rheumatol, Suppl 75: 117-122.

    92. Pereira L, Maidji E, McDonagh S, et al. 2005. Insights into viral transmission at the uterine-placental interface. Trends Microbiol, 13: 164-174.
        doi: 10.1016/j.tim.2005.02.009

    93. Plachter B, Sinzger C, Jahn G. 1996. Cell types in-volved in replication and distribution of human cytomega-lovirus. Adv Virus Res, 46: 195-261.
        doi: 10.1016/S0065-3527(08)60073-1

    94. Rafailidis P I, Mourtzoukou E G, Varbobitis I C, et al. 2008. Severe cytomegalovirus infection in apparently im-munocompetent patients: a systematic review. Virol J, 5: 47.
        doi: 10.1186/1743-422X-5-47

    95. Rahbar A, Bostrom L, Soderberg-Naucler C. 2006. Detection of cytotoxic CD13-specific autoantibodies in sera from patients with ulcerative colitis and Crohn's disease. J Autoimmun, 26: 155-164.
        doi: 10.1016/j.jaut.2006.02.003

    96. Rajagopalan S, Long E O. 2005. Viral evasion of NK-cell activation. Trends Immunol, 26: 403-405.
        doi: 10.1016/j.it.2005.06.008

    97. Randolph-Habecker J R, Rahill B, Torok-Storb B, et al. 2002. The expression of the cytomegalovirus chemokine receptor homolog US28 sequesters biologically active CC chemokines and alters IL-8 production. Cytokine, 19: 37-46.
        doi: 10.1006/cyto.2002.0874

    98. Rawlinson W D, Farrell H E, Barrell B G. 1996. Analysis of the complete DNA sequence of murine cytomegalovirus. J Virol, 70: 8833-8849.

    99. Reddehase M J, Podlech J, Grzimek N K. 2002. Mouse models of cytomegalovirus latency: overview. J Clin Virol, 25 Suppl 2:S23-36.

    100. Reeves M B, Lehner P J, Sissons J G, et al. 2005. An in vitro model for the regulation of human cytomegalovirus latency and reactivation in dendritic cells by chromatin remodelling. J Gen Virol, 86: 2949-2954.
        doi: 10.1099/vir.0.81161-0

    101. Reeves M B, MacAry P A, Lehner P J, et al. 2005. Latency, chromatin remodeling, and reactivation of human cytomegalovirus in the dendritic cells of healthy carriers. Proc Natl Acad Sci USA, 102: 4140-4145.
        doi: 10.1073/pnas.0408994102

    102. Reynolds D W, Stagno S, Alford C A. 1981. Chronic congenital and perinatal infections, In: Neonatal pathop-hysiolaogy and management in the newborn (Avery G B, ed. ). Lippincott : Philadelphia, p748-789.

    103. Rezania D, Ouban A, Marcet J, et al. 2007. CMV colitis mimicking recurrent inflammatory bowel disease: report of three cases. Am Surg, 73: 58-61.

    104. Roback J D. 2002. CMV and blood transfusions. Rev Med Virol, 12: 211-219.
        doi: 10.1002/(ISSN)1099-1654

    105. Robinson J. 2001. Infectious diseases in schools and child care facilities. Pediatr Rev, 22: 39-46.
        doi: 10.1542/pir.22-2-39

    106. Ross S A, Boppana S B. 2005. Congenital cytomega-lovirus infection: outcome and diagnosis. Semin Pediatr Infect Dis, 16: 44-49.
        doi: 10.1053/j.spid.2004.09.011

    107. Sansoni P, Vescovini R, Fagnoni F, et al. 2008. The immune system in extreme longevity. Exp Gerontol, 43: 61-65.
        doi: 10.1016/j.exger.2007.06.008

    108. Scalzo A A, Corbett A J, Rawlinson W D, et al. 2007. The interplay between host and viral factors in shaping the outcome of cytomegalovirus infection. Immunol Cell Biol, 85: 46-54.
        doi: 10.1038/sj.icb.7100013

    109. Schleiss M R. 2003. Vertically transmitted herpesvirus infections. Herpes, 10:4-11.

    110. Sekigawa I, Nawata M, Seta N, et al. 2002. Cytomega-lovirus infection in patients with systemic lupus ery-thematosus.Clin Exp Rheumatol, 20: 559-564.

    111. Sinclair J. 2008. Human cytomegalovirus: Latency and reactivation in the myeloid lineage. J Clin Virol, 41: 180-185.
        doi: 10.1016/j.jcv.2007.11.014

    112. Sinclair J, Sissons P. 2006. Latency and reactivation of human cytomegalovirus. J Gen Virol, 87: 1763-1779.
        doi: 10.1099/vir.0.81891-0

    113. Sinzger C, Jahn G. 1996. Human cytomegalovirus cell tropism and pathogenesis. Intervirology, 39: 302-319.
        doi: 10.1159/000150502

    114. Sissons J G, Bain M, Wills M R. 2002. Latency and reactivation of human cytomegalovirus. J Infect, 44: 73-77.
        doi: 10.1053/jinf.2001.0948

    115. Skaletskaya A, Bartle L M, Chittenden T, et al. 2001. A cytomegalovirus-encoded inhibitor of apoptosis that suppresses caspase-8 activation. Proc Natl Acad Sci USA, 98: 7829-7834.
        doi: 10.1073/pnas.141108798

    116. Smith H R, Heusel J W, Mehta I K, et al. 2002. Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc Natl Acad Sci USA, 99: 8826-8831.
        doi: 10.1073/pnas.092258599

    117. Smith M G. 1956. Propagation in tissue cultures of a cytopathogenic virus from human salivary gland virus (SGV) disease. Proc Soc Exp Biol Med, 92: 424-440.
        doi: 10.3181/00379727-92-22498

    118. Smith M G. 1954. Propagation of salivary gland virus of the mouse in tissue cultures. Proc Soc Exp Biol Med, 86: 435-440.
        doi: 10.3181/00379727-86-21123

    119. Soderberg-Naucler C, Fish K N, Nelson J A. 1997. Reactivation of latent human cytomegalovirus by allogeneic stimulation of blood cells from healthy donors. Cell, 91: 119-126.
        doi: 10.1016/S0092-8674(01)80014-3

    120. Soderberg-Naucler C, Streblow D N, Fish K N, et al. 2001. Reactivation of latent human cytomegalovirus in CD14(+) monocytes is differentiation dependent. J Virol, 75: 7543-7554.
        doi: 10.1128/JVI.75.16.7543-7554.2001

    121. Stahl H D, Hubner B, Seidl B, et al. 2000. Detection of multiple viral DNA species in synovial tissue and fluid of patients with early arthritis. Ann Rheum Dis, 59: 342-346.
        doi: 10.1136/ard.59.5.342

    122. Staras S A, Dollard S C, Radford K W, et al. 2006. Seroprevalence of cytomegalovirus infection in the United States, 1988-1994. Clin Infect Dis, 43: 1143-1151.
        doi: 10.1086/508173

    123. Streblow D N, Dumortier J, Moses A V, et al. 2008. Mechanisms of cytomegalovirus-accelerated vascular disease: induction of paracrine factors that promote angiogenesis and wound healing. Curr Top Microbiol Immunol, 325: 397-415.

    124. Sutherland C L, Chalupny N J, Schooley K, et al. 2002. UL16-binding proteins, novel MHC class Ⅰ-related proteins, bind to NKG2D and activate multiple signaling pathways in primary NK cells. J Immunol, 168: 671-679.
        doi: 10.4049/jimmunol.168.2.671

    125. Tang Q, Maul G G. 2006. Mouse cytomegalovirus crosses the species barrier with help from a few human cytomegalovirus proteins. J Virol, 80: 7510-7521.
        doi: 10.1128/JVI.00684-06

    126. Terhune S, Torigoi E, Moorman N, et al. 2007. Human cytomegalovirus UL38 protein blocks apoptosis. J Virol, 81: 3109-3123.
        doi: 10.1128/JVI.02124-06

    127. Thompson J N. 1994. The Coevolutionary Process. University of Chicago Press: Chicago, USA.

    128. Tsuchiya N, Murayama T, Yoshinoya S, et al. 1993. Antibodies to human cytomegalovirus 65-kilodalton Fc binding protein in rheumatoid arthritis: idiotypic mimicry hypothesis of rheumatoid factor production. Autoim-munity, 15: 39-48.
        doi: 10.3109/08916939309004837

    129. Vancikova Z, Dvorak P. 2001. Cytomegalovirus in-fection in immunocompetent and immunocompromised individuals--a review. Curr Drug Targets Immune Endocr Metabol Disord, 1: 179-187.
        doi: 10.2174/1568005310101020179

    130. Vasto S, Colonna-Romano G, Larbi A et al. 2007. Role of persistent CMV infection in configuring T cell immunity in the elderly.Immun Ageing, 4: 2.
        doi: 10.1186/1742-4933-4-2

    131. Verdonk R C, Haagsma E B, Van Den Berg A P, et al. 2006. Inflammatory bowel disease after liver trans-plantation: a role for cytomegalovirus infection. Scand J Gastroenterol, 41: 205-211.
        doi: 10.1080/00365520500206293

    132. Vescovini R, Biasini C, Fagnoni F F, et al. 2007. Massive load of functional effector CD4+ and CD8+ T cells against cytomegalovirus in very old subjects. J Immunol, 179: 4283-4291.
        doi: 10.4049/jimmunol.179.6.4283

    133. Vink C, Beuken E, Bruggeman C A. 2000. Complete DNA sequence of the rat cytomegalovirus genome. J Virol, 74: 7656-7665.
        doi: 10.1128/JVI.74.16.7656-7665.2000

    134. Vitale M, Castriconi R, Parolini S, et al. 1999. The leukocyte Ig-like receptor (LIR)-1 for the cytomega-lovirus UL18 protein displays a broad specificity for different HLA class Ⅰ alleles: analysis of LIR-1 + NK cell clones. Int Immunol, 11: 29-35.
        doi: 10.1093/intimm/11.1.29

    135. Voigt S, Mesci A, Ettinger J, et al. 2007. Cytomega-lovirus evasion of innate immunity by subversion of the NKR-P1B:Clr-b missing-self axis. Immunity, 26: 617-627.
        doi: 10.1016/j.immuni.2007.03.013

    136. Wagner C S, Ljunggren H G, Achour A. 2008. Immune modulation by the human cytomegalovirus-encoded molecule UL18, a mystery yet to be solved. J Immunol, 180: 19-24.
        doi: 10.4049/jimmunol.180.1.19

    137. Waller E C, Day E, Sissons J G, et al. 2008. Dynamics of T cell memory in human cytomegalovirus infection. Med Microbiol Immunol, 197: 83-96.
        doi: 10.1007/s00430-008-0082-5

    138. Weller T H, Hanshaw J B, Scott D E. 1960. Serological differentiation of viruses responsible for cytomegalic inclusion disease. Virology, 12: 130-132.
        doi: 10.1016/0042-6822(60)90156-2

    139. Wiertz E J, Jones T R, Sun L, et al. 1996. The human cytomegalovirus US11 gene product dislocates MHC class Ⅰ heavy chains from the endoplasmic reticulum to the cytosol. Cell, 84: 769-779.
        doi: 10.1016/S0092-8674(00)81054-5

    140. Woolhouse M E, Webster J P, Domingo E, et al. 2002. Biological and biomedical implications of the co-evolution of pathogens and their hosts. Nat Genet, 32: 569-577.
        doi: 10.1038/ng1202-569

    141. Yi L, Lin J Y, Gao Y, et al. 2008. Detection of human cytomegalovirus in the atherosclerotic cerebral arteries in han population in china. Acta Virol, 52: 99-106.

    142. Ziemann M, Krueger S, Maier A B, et al. 2007. High prevalence of cytomegalovirus DNA in plasma samples of blood donors in connection with seroconversion. Transfusion, 47: 1972-1983.
        doi: 10.1111/trf.2007.47.issue-11

  • 加载中

Article Metrics

Article views(3845) PDF downloads(15) Cited by()

Related
Proportional views

    Pathogenetic Consequences of Cytomegalovirus-Host Co-evolution

      Corresponding author: Fenyong Liu, liu_fy@uclink4.berkeley.edu
    • Division of Infectious Diseases, School of Public Health, University of California, Berkeley, CA 94720, USA
    Fund Project:  This work was, in part, supported by US Public Health Service DE014145This work was, in part, supported by US Public Health Service AI041927This work was, in part, supported by US Public Health Service AI050468This work was, in part, supported by US Public Health Service DE016813

    Abstract: Co-evolution has been shown to result in an adaptive reciprocal modification in the respective behaviors of interacting populations over time. In the case of host-parasite co-evolution, the adaptive behavior is most evident from the reciprocal change in fitness of host and parasite-manifested in terms of pathogen survival versus host resistance. Cytomegaloviruses and their hosts represent a pairing of populations that has co-evolved over hundreds of years. This review explores the pathogenetic consequences emerging from the behavioral changes caused by co-evolutionary forces on the virus and its host.

    • Co-evolution is the process of reciprocal, adaptive genetic change in two or more species (127, 140). It can occur between any interacting populations, but it is expected to be particularly important in host-pathogen systems because of the intimate nature of the association and the strong selective pressures that each can exert on the other (140). Cytomegaloviruses (CMV) and their hosts are thought to have co-evolved over hundreds of years (40). Recent data brought to light by studies on CMV pathogenesis provide an interesting glimpse on how the lengthy association between virus and host has shaped many of the viral pathogenetic features.

    • Human CMV (HCMV) is a ubiquitous human herpesvirus that infects a great majority of the world's population. The virus is a member of the β-group of herpesviruses characterized by their strict host speci-ficity, comparatively large genome, and slow growth in culture (66, 80). Cells infected with the virus present a characteristic ballooned-cytopathology from which the name of the etiologic agent-"cytomega-lovirus"-was derived (117, 118, 138). The linear double stranded viral DNA genome of 200-240 kbp encodes at least 150 proteins, many of which contri-bute to making the virus one of the most successful intracellular parasites in nature (25, 31, 81, 82, 98, 133).

      The worldwide incidence of HCMV infection varies widely from 60%-100% (40, 122). Disparity in the incidence of the disease among population groups is influenced by prevailing health, hygiene, and socio-economic status (40, 86, 102, 122). Although the infection is mostly well tolerated by people with intact immune systems, it can be life threatening or severely debilitating for immunocompromised individuals such as AIDS patients, cancer patients, organ or tissue transplant recipients undergoing immunosuppressive therapy, infants, fetuses, and the elderly (1, 33, 34, 86, 87, 102, 129).

      Like other herpesviruses, the CMV infectious cycle starts with an acute episode which progresses to a persistent stage that paves the way to the latent state (1, 49, 62, 66). Acute infection is usually precipitated by contact of virus with epithelial body surfaces but may also occur after organ transplant, blood trans-fusion, or as a result of congenital or perinatal infection (17, 24, 88, 104-106). It is characterized by robust viral replication at the site of primary infection and may become disseminated systemically via the lymphatic system then through cell-associated viremia. Terminal cases of acute CMV infection typically reveal widespread lesions of cellular cytomegaly in almost all organs of the body (94). The most common cell types found to be infected at autopsy are ductal epithelial cells, but other cell types also known to be infected are endothelial cells, smooth muscle cells, hepatocytes, granulocytes, monocytes, neurons, and glial cells (61, 65, 66, 93, 113, 129).

      The persistent phase of CMV disease is characteri-zed by low-level viral replication in infected organs. There is intermittent viral shedding in body fluids due to persistent infection of ductal epithelia. This stage usually presages latent infection wherein the infected host is negative for virus excretion but harbor the viral genome in a non-replicative form in some cells (49, 50, 99, 111, 112, 114). Latent virus may reactivate when the host is immunosupressed or presumably, when the latently infected progenitor cell undergoes differentiation into the mature phenotype (112, 114, 119, 120). The molecular mechanisms underlying viral reactivation from latency are not well understood. In the blood mononuclear cell lineage where data are most abundant, it has been shown that latently infected monocyte progenitor cells reactivate the virus in the course of terminal differentiation. Interestingly, reactivation appears to be associated with chromatin remodeling that accompanies cellular differentiation. Indeed, in one model, it has been shown that euchromatin formation resulting from acetylation of histone tails also loosens important regions in the latent viral DNA, notably the IE region, thereby allowing access of factors involved in initiating viral transcription that culminate in reactivation (101).

      CMV disease is transmitted vertically from mother to offspring, or horizontally between individuals (1, 62, 87, 88). Vertical transmission can occur trans-placentally, during birth, or through breast milk (5, 7, 17, 24, 57, 92, 109). Horizontal transmission can occur by direct exchange of body fluids, organ/tissue transplantation, or through contaminated materials as occurs in day care centers (32, 84, 142).

    • Reciprocal traits involved in pathogen-host co-evolution usually operate in opposite directions: what enhances fitness of the host diminishes the fitness of the pathogen, and vice-versa (140). In the context of CMV-host co-evolution, enhanced features of the virus include focused resources towards infecting a specific host species, striking evasive adaptability to the host's repertoire of antiviral strategies, and ability to perpetuate itself in the host. As such, CMVs, do not cross the species barrier (54, 108, 125), they have developed cunning ways of evading the host immune response (9-12, 14-16, 19, 26, 29, 37, 48, 52, 56, 68, 69, 71, 79, 96, 135, 136), they code for functions that temper host perturbation (31) or delay apoptosis of infected cells (41, 42, 77, 115, 126), and most impor-tantly-they can establish latency and subsequently reactivate when the conditions are right (100, 101, 111, 112, 114).

      On the part of the virus' co-evolutionary partner-the host, the reciprocal loss of fitness is reflected in terms of pathogenetic manifestations that include prolonged persistence or life-long latent infection with recurrent disease episodes or periodic reactivation, autoimmune syndromes, inflammaging and immuno-senescence.

    • The hallmark of all herpesvirus infections is prolonged persistence and latent infection. After an initial acute episode in which the virus goes through a burst of robust replication in various organ systems, the virus transitions into a low-level replication stage with intermittent shedding by the infected host. This is the persistent stage of CMV infection and it probably mirrors the shifting events between the host immune system and the viral replication machinery. At the host end, NK cells and other components of the cellular immune system are actively trying to root out in-fection; on the virus end, gene products are actively trying to subvert the host immune system in order to escape it.

      Among the viral gene products reputed to modulate host immune function by interfering with natural killer (NK) cell activity are the gene products of UL40, pUL18, and UL16 of the human virus, and those of m155 and M157 of the mouse virus (MCMV). UL40 gene product upregulates HLA-E expression for recognition by inhibitory NK cell receptors, pUL18 (MHC Ⅰ heavy chain homologue) replaces host MHC Ⅰ in infected cell surface and bind natural killer inhibi-tory receptors (KIRs) (21, 134), UL16 upregulates expression of UL-binding proteins (ULBPs) that sequester NKG2D ligands (22, 30, 124), m155 of MCMV downregulates expression of H60 which is a high affinity ligand for activating receptor NKG2D (70), M157 of MCMV binds to an inhibitory NK receptor in susceptible mice and to an activating receptor (Ly-49H) in resistant mice (4, 116).

      The virus also tampers with the host immune function by interfering with MHC Ⅰ gene expression and antigen processing. For example, US2 and US11 displace the heavy chain from the ER to the cytosol where it is degraded by proteosome (38, 139), US6 binds to TAP and prevents transport of peptides produced in the proteasome to the ER (3, 43, 67), and US3 promotes retention of assembled MHC molecules in the ER (2, 53).

      The virus' repertoire for immune modulation also includes chemokine perturbation, exemplified as follows. HCMV encodes chemokine receptor homolo-gues (e.g. US28) that can act as a chemokine sink-sequestering chemokines and preventing them from activating target effectors (97). The UL146 gene product also causes chemotaxis of cells harboring the homologous receptor, thus acting as a bait to propagate infection to other cells (97). UL111.5A, a homologue of IL-10 that functions to dampen host inflammatory response, has been found to be ex-pressed during viral latency in granulocyte-macro-phage progenitor cells (51); presumably, protecting infected cells from the influx of inflammatory cells.

      One might question whether viral latency actually represents as much a triumph for the virus as it is for the host. It is clear that the host immune system is a factor in maintaining the viral latent state since immunosuppression elicits reactivation. Nevertheless, from the viral perspective, the latent state also allows for a relatively safe refuge in the host and freedom from immunologic threat and elimination. Moreover, the latent state provides the virus the opportunity to wait out a hostile environment and the chance to reactivate as soon as conditions become more favo-rable.

    • Because part of the viral armamentarium for survival consists of molecular mimics of host proteins, and because persistent CMV infection and its intermittent reactivation put constant immunological pressure on the host, it has been suggested that CMV infection predisposes the host to develop autoimmune disorders. Indeed, antibodies directed against some viral proteins have been found to cross-react with the host proteins that they resemble. Alternatively, sera from indivi-duals suffering from autoimmune diseases have been shown to react with CMV viral antigens. Examples of autoimmune disorders with known or suspected links to CMV are rheumatoid arthritis (RA) (45, 76, 78, 121), Systemic Lupus Erythematosus (SLE) (27, 46, 110), Inflammatory Bowel Disease (IBD) (23, 28, 44, 47, 63, 103), Systemic Sclerosis (Ssc) (72-74, 83, 85), and vascular diseases such as atherosclerosis, restenosis, and transplant vascular stenosis (13, 72, 75, 123, 141).

      CMV involvement in RA is thought to be related to production of rheumatoid factors (RFs) in the form of antiidiotypic antibodies to anti-viral Fc gamma-binding proteins (FcBPs) of CMV and other herpes-viruses (91, 128). In SLE, a large array of autoanti-bodies is produced that primarily target the whole chromatin (antinucleosome) and its individual com-ponents, dsDNA and histones (64). Apoptotic defects and impaired removal of apoptotic cells could contri-bute to an overload of autoantigens (and in particular of nucleosomes) in circulation or in target tissues that could become available to initiate an autoimmune response (20, 64, 74). Clinical evidence of CMV involvement in SLE is mounting (20, 46, 110).

      The chronic mucosal inflammation in IBD is caused by hyperactivation of effector immune cells, which produce high levels of pro-inflammatory cytokines like tumor necrosis factor-alpha, interleukin-6 and interferon-gamma, resulting in colonic tissue damage (6). The nuclear transcription factor NF-kB was identified as one of the key regulators in this immuno-logical setting. NF-kB activation is markedly induced in IBD patients and it strongly influences the course of mucosal inflammation through its ability to promote the expression of various pro-inflammatory genes. CMV genome is frequently found in IBD tissues, and epidemiologic studies strongly suggest association between the virus and the disease, although a definite causal relationship is yet to be established (23, 28, 95, 131).

      Lunardi et al showed that in Ssc, endothelial cells undergo apoptosis following tetraspan novel antigen-2 (NAG-2) receptor binding by antibodies directed against the HCMV UL94 protein (73, 83). Moreover, they also showed that anti-HCMV antibodies not only caused endothelial cell activation and apoptosis but also activation of fibroblasts, which is a hallmark of the disease (74). The vascular endothelial injury, with intimal proliferation and luminal narrowing in arteries and arterioles, excessive extracellular matrix accumulation, and fibroblast activation presumably leads to fibrosis of the skin and internal organs. Namboordiri et al. also showed significantly higher antibodies to HCMV UL83 among SSc patients, but could not detect any major Ssc autoantibody related to this protein (83).

      In atherosclerosis, apoptosis and cytotoxic damage to endothelial cells have been attributed to binding of receptors and HSP 60 by antibodies directed to US28 and UL122 proteins resulting in plaque-formation and narrowing of involved blood vessel (8, 72, 75). Recently, HCMV infection has also been shown to result in increased endothelial cell proliferation, moti-lity, and capillary tube formation (13). The observed HCMV-induced angiogenic response depended on viral binding to and signaling through the β1 and β3 integrins and the epidermal growth factor receptor, via their ability to activate the phosphatidylinositol 3-kinase and the mitogen-activated protein kinase signaling pathways.

    • These terms have found their way into scientific literatures fairly recently as a result of growing consciousness for the health and welfare of the rapidly graying segment of the population. Inflammaging has been proposed to describe the low-grade, chronic, systemic inflammatory state that characterizes the aging process (39), while immunosenescence describes the decline in immunologic responsiveness due to adaptive remodeling of parts of the immune system that occurs with age (18, 36, 58). Both conditions are related to chronic antigenic exposure to a variety of antigens, especially to some viruses, and contribute significantly to age-associated morbidity and mortality (35, 90). Presumably, the extended and sustained immunologic burden that occurs with chronic in-fections causes deterioration of clonotypic immunity, while innate immunity is largely preserved. There is accumulation of memory effector T-cells and exhaus-tion of naïve T-cells that is further exacerbated by the age-related involution of the thymus (18, 36). Emer-ging data suggest that CMV is a major contributor in inflammaging and immunosenescence because of its chronicity, widespread incidence, and immunogeni-city (55, 107, 132, 137). Lifelong infection with CMV causes chronic antigenic stress resulting in the accumu-lation of anergic, apoptosis-resistant CD8 T-cells that burden the immune system (60, 89). Indeed, CMV seropositivity among the elderly is associated with oligoclonal expansion of T cells, especially CD8 cells of which as much as 10-14% carry receptors for just a single HCMV epitope (59, 130). Such a specific oligoclonal CD8 expansion, especially in the elderly, squanders limited immunologic space and is regarded to be a poor prognostic factor for survival in the elderly.

    • The co-evolution of CMV with its host has remark-ably increased CMV's success in perpetuating itself in nature. As a result of its long-term intimate interaction with the host, CMV has acquired a complex array of functions to counterbalance the defensive repertoire of its host. In keeping with the principle that reciprocal traits involved in pathogen-host co-evolution operate in opposite directions, the enhanced viral fitness is mirrored by the diminished fitness of the host. Consequently, the host is overrun by a wide spectrum of diseases, many of which reflect the virus-host co-evolutionary struggle.

    Reference (142) Relative (20)

    目录

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return