全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Hantavirus Regulation of Type I Interferon Responses

DOI: 10.1155/2012/524024

Full-Text   Cite this paper   Add to My Lib

Abstract:

Hantaviruses primarily infect human endothelial cells (ECs) and cause two highly lethal human diseases. Early addition of Type I interferon (IFN) to ECs blocks hantavirus replication and thus for hantaviruses to be pathogenic they need to prevent early interferon induction. PHV replication is blocked in human ECs, but not inhibited in IFN deficient VeroE6 cells and consistent with this, infecting ECs with PHV results in the early induction of IFNβ and an array of interferon stimulated genes (ISGs). In contrast, ANDV, HTNV, NY-1V and TULV hantaviruses, inhibit early ISG induction and successfully replicate within human ECs. Hantavirus inhibition of IFN responses has been attributed to several viral proteins including regulation by the Gn proteins cytoplasmic tail (Gn-T). The Gn-T interferes with the formation of STING-TBK1-TRAF3 complexes required for IRF3 activation and IFN induction, while the PHV Gn-T fails to alter this complex or regulate IFN induction. These findings indicate that interfering with early IFN induction is necessary for hantaviruses to replicate in human ECs, and suggest that additional determinants are required for hantaviruses to be pathogenic. The mechanism by which Gn-Ts disrupt IFN signaling is likely to reveal potential therapeutic interventions and suggest protein targets for attenuating hantaviruses. 1. Introduction 1.1. Disease Hantaviruses are present worldwide and responsible for two diseases: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). HFRS is primarily present in Eurasia and caused by several hantaviruses including Hantaan virus (HTNV), Seoul virus (SEOV), Puumala virus (PUUV) and Dobrava virus (DOBV) [1–3]. HFRS has a mortality rate ranging from 0.1–5% with causes of death including shock (75%), uremia (50%), pulmonary edema (15%), and central nervous system hemorrhage or encephalopathy (5%) [1–7]. In 1993, a discrete North American hantavirus (Sin Nombre virus, SNV) was found in the southwestern United States as the cause of a new highly lethal respiratory syndrome termed HPS [1, 2, 8–14]. HPS causing hantaviruses have since been found throughout the Americas [15–20]. Andes virus (ANDV) is a prototypic South American HPS causing hantavirus and the only hantavirus that is reportedly spread from person to person [21–24]. Although hantaviruses are predominantly pathogenic, Prospect Hill virus (PHV) and Tula virus (TULV) are hantaviruses which are not associated with human disease, and are referred to here as nonpathogenic, although it is unclear whether these viruses cause

References

[1]  K. M. Johnson, “Hantaviruses: history and overview,” in Hantaviruses, C. A. N. Schmaljohn, Ed., vol. 256, pp. 1–14, Springer, 2001.
[2]  C. Schmaljohn and B. Hjelle, “Hantaviruses: a global disease problem,” Emerging Infectious Diseases, vol. 3, no. 2, pp. 95–104, 1997.
[3]  H. W. Lee, “Hemorrhagic fever with renal syndrome in Korea,” Reviews of Infectious Diseases, vol. 11, supplement 1, pp. S864–S876, 1989.
[4]  J. P. Chen and T. M. Cosgriff, “Hemorrhagic fever virus-induced changes in hemostasis and vascular biology,” Blood Coagulation and Fibrinolysis, vol. 11, no. 5, pp. 461–483, 2000.
[5]  J. Lahdevirta, “Clinical features of HFRS in Scandinavia as compared with East Asia,” Scandinavian Journal of Infectious Diseases, vol. 14, no. 36, pp. 93–95, 1982.
[6]  J. L?hdevirta, E. Enger, O. H. Hunderi, T. Traavik, and H. W. Lee, “Hantaan virus is related to hemorrhagic fever with renal syndrome in Norway,” The Lancet, vol. 2, no. 8298, p. 606, 1982.
[7]  K. Penttinen, J. Lahdevirta, and R. Kekomaki, “Circulating immune complexes, immunoconglutinins, and rheumatoid factors in nephropathia epidemica,” Journal of Infectious Diseases, vol. 143, no. 1, pp. 15–21, 1981.
[8]  S. R. Zaki, P. W. Greer, L. M. Coffield et al., “Hantavirus pulmonary syndrome: pathogenesis of an emerging infectious disease,” American Journal of Pathology, vol. 146, no. 3, pp. 552–579, 1995.
[9]  A. S. Khan and J. C. Young, “Hantavirus pulmonary syndrome: at the crossroads,” Current Opinion in Infectious Diseases, vol. 14, no. 2, pp. 205–209, 2001.
[10]  E. A. Bustamante, H. Levy, and S. Q. Simpson, “Pleural fluid characteristics in hantavirus pulmonary syndrome,” Chest, vol. 112, no. 4, pp. 1133–1136, 1997.
[11]  J. S. Duchin, F. T. Koster, C. J. Peters et al., “Hantavirus pulmonary syndrome: a clinical description of 17 patients with a newly recognized disease,” New England Journal of Medicine, vol. 330, no. 14, pp. 949–955, 1994.
[12]  L. H. Elliott, T. G. Ksiazek, P. E. Rollin et al., “Isolation of the causative agent of hantavirus pulmonary syndrome,” American Journal of Tropical Medicine and Hygiene, vol. 51, no. 1, pp. 102–108, 1994.
[13]  K. B. Nolte, R. M. Feddersen, K. Foucar et al., “Hantavirus pulmonary syndrome in the United States: a pathological description of a disease caused by a new agent,” Human Pathology, vol. 26, no. 1, pp. 110–120, 1995.
[14]  C. Schmaljohn, “Bunyaviridae and their replication,” in Fields Virology, vol. 1, pp. 1581–1602, Lippincott Williams & Wilkins, 4th edition, 2001.
[15]  D. Enria, P. Padula, E. L. Segura et al., “Hantavirus pulmonary syndrome in Argentina possibility of person to person transmission,” Medicina, vol. 56, no. 6, pp. 709–711, 1996.
[16]  I. Gavrilovskaya, R. LaMonica, M. E. Fay et al., “New York 1 and sin nombre viruses are serotypically distinct viruses associated with hantavirus pulmonary syndrome,” Journal of Clinical Microbiology, vol. 37, no. 1, pp. 122–126, 1999.
[17]  B. Hjelle, S. W. Lee, W. Song et al., “Molecular linkage of hantavirus pulmonary syndrome to the white-fooled mouse, Peromyscus leucopus: genetic characterization of the M genome of New York virus,” Journal of Virology, vol. 69, no. 12, pp. 8137–8141, 1995.
[18]  S. T. Nichol, C. F. Spiropoulou, S. Morzunov et al., “Genetic identification of a hantavirus associated with an outbreak of acute respiratory illness,” Science, vol. 262, no. 5135, pp. 914–917, 1993.
[19]  A. L. Schmaljohn, D. Li, D. L. Negley et al., “Isolation and initial characterization of a newfound hantavirus from California,” Virology, vol. 206, no. 2, pp. 963–972, 1995.
[20]  J. W. Song, L. J. Baek, D. Carleton Gajdusek et al., “Isolation of pathogenic hantavirus from white-footed mouse (Peromyscus leucopus),” The Lancet, vol. 344, no. 8937, p. 1637, 1994.
[21]  H. Galeno, J. Mora, E. Villagra et al., “First human isolate of hantavirus (Andes virus) in the Americas,” Emerging Infectious Diseases, vol. 8, no. 7, pp. 657–661, 2002.
[22]  N. López, P. Padula, C. Rossi et al., “Genetic characterization and phylogeny of andes virus and variants from Argentina and Chile,” Virus Research, vol. 50, no. 1, pp. 77–84, 1997.
[23]  V. P. Martinez, C. Bellomo, J. San Juan et al., “Person-to-person transmission of Andes virus,” Emerging Infectious Diseases, vol. 11, no. 12, pp. 1848–1853, 2005.
[24]  P. J. Padula, A. Edelstein, S. D. L. Miguel, N. M. López, C. M. Rossi, and R. D. Rabinovich, “Hantavirus pulmonary syndrome outbreak in Argentina: molecular evidence for person-to-person transmission of Andes virus,” Virology, vol. 241, no. 2, pp. 323–330, 1998.
[25]  A. Plyusnin, O. Vapalahti, H. Lankinen et al., “Tula virus: a newly detected hantavirus carried by European common voles,” Journal of Virology, vol. 68, no. 12, pp. 7833–7839, 1994.
[26]  O. Vapalahti, S. K. Lundkvist, S. K. J. Kukkonen et al., “Isolation and characterization of Tula virus, a distinct serotype in the genus Hantavirus, family Bunyaviridae,” Journal of General Virology, vol. 77, no. 12, pp. 3063–3067, 1996.
[27]  A. A. Kraus, M. J. Raftery, T. Giese et al., “Differential antiviral response of endothelial cells after infection with pathogenic and nonpathogenic hantaviruses,” Journal of Virology, vol. 78, no. 12, pp. 6143–6150, 2004.
[28]  R. Yanagihara, C. A. Daum, P. W. Lee et al., “Serological survey of Prospect Hill virus infection in indigenous wild rodents in the USA,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 81, no. 1, pp. 42–45, 1987.
[29]  R. Yanagihara and D. J. Silverman, “Experimental infection of human vascular endothelial cells by pathogenic and nonpathogenic hantaviruses,” Archives of Virology, vol. 111, no. 3-4, pp. 281–286, 1990.
[30]  M. N. Pensiero, J. B. Sharefkin, C. W. Dieffenbach, and J. Hay, “Hantaan virus infection of human endothelial cells,” Journal of Virology, vol. 66, no. 10, pp. 5929–5936, 1992.
[31]  J. P. Clement, “Hantavirus,” Antiviral Research, vol. 57, no. 1-2, pp. 121–127, 2003.
[32]  A. Plyusnin and S. P. Morzunov, “Virus evolution and genetic diversity of hantaviruses and their rodent hosts,” Current Topics in Microbiology and Immunology, vol. 256, pp. 47–75, 2000.
[33]  N. D. Tischler, H. Galeno, M. Rosemblatt, and P. D. T. Valenzuela, “Human and rodent humoral immune responses to Andes virus structural proteins,” Virology, vol. 334, no. 2, pp. 319–326, 2005.
[34]  J. Hepojoki, T. Strandin, H. Wang, O. Vapalahti, A. Vaheri, and H. Lankinen, “Cytoplasmic tails of hantavirus glycoproteins interact with the nucleocapsid protein,” Journal of General Virology, vol. 91, no. 9, pp. 2341–2350, 2010.
[35]  P. Kaukinen, A. Vaheri, and A. Plyusnin, “Hantavirus nucleocapsid protein: a multifunctional molecule with both housekeeping and ambassadorial duties,” Archives of Virology, vol. 150, no. 9, pp. 1693–1713, 2005.
[36]  J. O. Virtanen, K. M. Jaaskelainen, J. Djupsj?backa, A. Vaheri, and A. Plyusnin, “Tula hantavirus NSs protein accumulates in the perinuclear area in infected and transfected cells,” Archives of Virology, vol. 155, no. 1, pp. 117–121, 2010.
[37]  K. M. Jaaskelainen, A. Plyusnina, S. K. Lundkvist, A. Vaheri, and A. Plyusnin, “Tula hantavirus isolate with the full-length ORF for nonstructural protein NSs survives for more consequent passages in interferon-competent cells than the isolate having truncated NSs ORF,” Virology Journal, vol. 5, article 3, 2008.
[38]  C. S. Schmaljohn, A. L. Schmaljohn, and J. M. Dalrymple, “Hantaan virus M RNA: coding strategy, nucleotide sequence, and gene order,” Virology, vol. 157, no. 1, pp. 31–39, 1987.
[39]  M. N. Pensiero and J. Hay, “The Hantaan virus M-segment glycoproteins G1 and G2 can be expressed independently,” Journal of Virology, vol. 66, no. 4, pp. 1907–1914, 1992.
[40]  M. N. Pensiero, G. B. Jennings, C. S. Schmaljohn, and J. Hay, “Expression of the Hantaan virus M genome segment by using a vaccinia virus recombinant,” Journal of Virology, vol. 62, no. 3, pp. 696–702, 1988.
[41]  C. L?ber, B. Anheier, S. Lindow, H. D. Klenk, and H. Feldmann, “The Hantaan virus glycoprotein precursor is cleaved at the conserved pentapeptide WAASA,” Virology, vol. 289, no. 2, pp. 224–229, 2001.
[42]  K. I. Kamrud and C. S. Schmaljohn, “Expression strategy of the M genome segment of Hantaan virus,” Virus Research, vol. 31, no. 1, pp. 109–121, 1994.
[43]  V. M. Deyde, A. A. Rizvanov, J. Chase, E. W. Otteson, and S. C. S. Jeor, “Interactions and trafficking of Andes and Sin Nombre Hantavirus glycoproteins G1 and G2,” Virology, vol. 331, no. 2, pp. 307–315, 2005.
[44]  D. Antic, K. E. Wright, and C. Y. Kang, “Maturation of Hantaan virus glycoproteins G1 and G2,” Virology, vol. 189, no. 1, pp. 324–328, 1992.
[45]  N. D. Tischler, A. Gonzales, T. Perez-Acle, M. Rosemblatt, and P. D. T. Valenzuela, “Hantavirus Gc glycoprotein: evidence for a class ll fusion protein,” Journal of General Virology, vol. 86, no. 11, pp. 2937–2947, 2005.
[46]  C. F. Spiropoulou, C. S. Goldsmith, T. R. Shoemaker, C. J. Peters, and R. W. Compans, “Sin Nombre virus glycoprotein trafficking,” Virology, vol. 308, no. 1, pp. 48–63, 2003.
[47]  E. Geimonen, I. Fernandez, I. N. Gavrilovskaya, and E. R. Mackow, “Tyrosine residues direct the ubiquitination and degradation of the NY-1 hantavirus G1 cytoplasmic tail,” Journal of Virology, vol. 77, no. 20, pp. 10760–10768, 2003.
[48]  E. Geimonen, R. LaMonica, K. Springer, Y. Farooqui, I. N. Gavrilovskaya, and E. R. Mackowl, “Hantavirus pulmonary syndrome-associated hantaviruses contain conserved and functional ITAM signaling elements,” Journal of Virology, vol. 77, no. 2, pp. 1638–1643, 2003.
[49]  P. J. Alff, I. N. Gavrilovskaya, E. Gorbunova et al., “The pathogenic NY-1 hantavirus G1 cytoplasmic tail inhibits RIG-I-and TBK-1-directed interferon responses,” Journal of Virology, vol. 80, no. 19, pp. 9676–9686, 2006.
[50]  P. J. Alff, N. Sen, E. Gorbunova, I. N. Gavrilovskaya, and E. R. Mackow, “The NY-1 hantavirus Gn cytoplasmic tail coprecipitates TRAF3 and inhibits cellular interferon responses by disrupting TBK1-TRAF3 complex formation,” Journal of Virology, vol. 82, no. 18, pp. 9115–9122, 2008.
[51]  V. Matthys, E. E. Gorbunova, I. N. Gavrilovskaya, T. Pepini, and E. R. Mackow, “The C-terminal 42 residues of the tula virus Gn protein regulate interferon induction,” Journal of Virology, vol. 85, no. 10, pp. 4752–4760, 2011.
[52]  N. Sen, A. Sen, and E. R. Mackow, “Degrons at the C terminus of the pathogenic but not the nonpathogenic hantavirus G1 tail direct proteasomal degradation,” Journal of Virology, vol. 81, no. 8, pp. 4323–4330, 2007.
[53]  H. Wang, T. Strandin, J. Hepojoki, H. Lankinen, and A. Vaheri, “Degradation and aggresome formation of the Gn tail of the apathogenic Tula hantavirus,” Journal of General Virology, vol. 90, no. 12, pp. 2995–3001, 2009.
[54]  R. B. Seth, L. Sun, and Z. J. Chen, “Antiviral innate immunity pathways,” Cell Research, vol. 16, no. 2, pp. 141–147, 2006.
[55]  G. R. Stark, I. M. Kerr, B. R. G. Williams, R. H. Silverman, and R. D. Schreiber, “How cells respond to interferons,” Annual Review of Biochemistry, vol. 67, pp. 227–264, 1998.
[56]  E. Meylan and J. Tschopp, “Toll-like receptors and RNA helicases: two parallel ways to trigger antiviral responses,” Molecular Cell, vol. 22, no. 5, pp. 561–569, 2006.
[57]  M. Yoneyama and T. Fujita, “RIG-I family RNA helicases: cytoplasmic sensor for antiviral innate immunity,” Cytokine and Growth Factor Reviews, vol. 18, no. 5-6, pp. 545–551, 2007.
[58]  M. Yoneyama, M. Kikuchi, T. Natsukawa et al., “The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses,” Nature Immunology, vol. 5, no. 7, pp. 730–737, 2004.
[59]  G. N. Barber, “Innate immune DNA sensing pathways: STING, AIMII and the regulation of interferon production and inflammatory responses,” Current Opinion in Immunology, vol. 23, no. 1, pp. 10–20, 2011.
[60]  A. Bowie, “The STING in the tail for cytosolic DNA-dependent activation of IRF3,” Science Signaling, vol. 5, no. 214, Article ID pe9, 2012.
[61]  H. Ishikawa and G. N. Barber, “STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling,” Nature, vol. 455, no. 7213, pp. 674–678, 2008.
[62]  H. Ishikawa and G. N. Barber, “The STING pathway and regulation of innate immune signaling in response to DNA pathogens,” Cellular and Molecular Life Sciences, vol. 68, no. 7, pp. 1157–1165, 2011.
[63]  H. Ishikawa, Z. Ma, and G. N. Barber, “STING regulates intracellular DNA-mediated, type i interferon-dependent innate immunity,” Nature, vol. 461, no. 7265, pp. 788–792, 2009.
[64]  S. Ouyang, X. Song, Y. Wang et al., “Structural analysis of the STING adaptor protein reveals a hydrophobic dimer interface and mode of cyclic di-GMP binding,” Immunity, vol. 36, no. 6, pp. 1073–1086, 2012.
[65]  Y. Tanaka and Z. J. Chen, “STING specifies IRF3 phosphorylation by TBK1 in the cytosolic DNA signaling pathway,” Science Signaling, vol. 5, no. 214, Article ID ra20, 2012.
[66]  T. Kawai, K. Takahashi, S. Sato et al., “IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction,” Nature Immunology, vol. 6, no. 10, pp. 981–988, 2005.
[67]  S. K. Saha, E. M. Pietras, J. Q. He et al., “Regulation of antiviral responses by a direct and specific interaction between TRAF3 and Cardif,” EMBO Journal, vol. 25, no. 14, pp. 3257–3263, 2006.
[68]  M. Yoneyama and T. Fujita, “Function of RIG-I-like receptors in antiviral innate immunity,” Journal of Biological Chemistry, vol. 282, no. 21, pp. 15315–15318, 2007.
[69]  L. He, A. C. Grammer, X. Wu, and P. E. Lipsky, “TRAF3 forms heterotrimers with TRAF2 and modulates its ability to mediate NF-κB activation,” Journal of Biological Chemistry, vol. 279, no. 53, pp. 55855–55865, 2004.
[70]  K. Hoebe and B. Beutler, “TRAF3: a new component of the TLR-signaling apparatus,” Trends in Molecular Medicine, vol. 12, no. 5, pp. 187–189, 2006.
[71]  G. Oganesyan, S. K. Saha, B. Guo et al., “Critical role of TRAF3 in the Toll-like receptor-dependent and -independent antiviral response,” Nature, vol. 439, no. 7073, pp. 208–211, 2006.
[72]  S. K. Saha and G. Cheng, “TRAF3: a new regulator of type I interferons,” Cell Cycle, vol. 5, no. 8, pp. 804–807, 2006.
[73]  K. A. Fitzgerald, S. M. McWhirter, K. L. Faia et al., “IKKE and TBKI are essential components of the IRF3 signalling pathway,” Nature Immunology, vol. 4, no. 5, pp. 491–496, 2003.
[74]  T. L. Chau, R. Gioia, J. S. Gatot et al., “Are the IKKs and IKK-related kinases TBK1 and IKK-ε similarly activated?” Trends in Biochemical Sciences, vol. 33, no. 4, pp. 171–180, 2008.
[75]  K. Parvatiyar, G. N. Barber, and E. W. Harhaj, “TAX1BP1 and A20 inhibit antiviral signaling by targeting TBK1-IKKi kinases,” Journal of Biological Chemistry, vol. 285, no. 20, pp. 14999–15009, 2010.
[76]  J. L. Pomerantz and D. Baltimore, “NF-κB activation by a signaling complex containing TRAF2, TANK and TBK1, a novel IKK-related kinase,” EMBO Journal, vol. 18, no. 23, pp. 6694–6704, 1999.
[77]  M. R. S. Rani and R. M. Ransohoff, “Alternative and accessory pathways in the regulation of IFN-β-mediated gene expression,” Journal of Interferon and Cytokine Research, vol. 25, no. 12, pp. 788–798, 2005.
[78]  J. E. Darnell, I. M. Kerr, and G. R. Stark, “Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins,” Science, vol. 264, no. 5164, pp. 1415–1421, 1994.
[79]  C. Schindler and J. E. Darnell, “Transcriptional responses to polypeptide ligands: the JAK-STAT pathway,” Annual Review of Biochemistry, vol. 64, pp. 621–651, 1995.
[80]  G. Shang, D. Zhu, N. Li et al., “Crystal structures of STING protein reveal basis for recognition of cyclic di-GMP,” Nature Structural and Molecular Biology, vol. 19, pp. 725–727, 2012.
[81]  S. F. Khaiboullina, A. A. Rizvanov, V. M. Deyde, and S. C. S. Jeor, “Andes virus stimulates interferon-inducible MxA protein expression in endothelial cells,” Journal of Medical Virology, vol. 75, no. 2, pp. 267–275, 2005.
[82]  M. Kanerva, J. Mustonen, and A. Vaheri, “Pathogenesis of Puumala and other hantavirus infections,” Reviews in Medical Virology, vol. 8, no. 2, pp. 67–86, 1998.
[83]  M. Frese, G. Kochs, H. Feldmann, C. Hertkorn, and O. Haller, “Inhibition of Bunyaviruses, Phleboviruses, and Hantaviruses by human MxA protein,” Journal of Virology, vol. 70, no. 2, pp. 915–923, 1996.
[84]  C. B. Jonsson, J. Hooper, and G. Mertz, “Treatment of hantavirus pulmonary syndrome,” Antiviral Research, vol. 78, no. 1, pp. 162–169, 2008.
[85]  M. Tamura, H. Asada, K. Kondo, M. Takahashi, and K. Yamanishi, “Effects of human and murine interferons against hemorrhagic fever with renal syndrome (HFRS) virus (Hantaan virus),” Antiviral Research, vol. 8, no. 4, pp. 171–178, 1987.
[86]  C. B. Jonsson, L. T. M. Figueiredo, and O. Vapalahti, “A global perspective on hantavirus ecology, epidemiology, and disease,” Clinical Microbiology Reviews, vol. 23, no. 2, pp. 412–441, 2010.
[87]  E. Geimonen, S. Neff, T. Raymond, S. S. Kocer, I. N. Gavrilovskaya, and E. R. Mackow, “Pathogenic and nonpathogenic hantaviruses differentially regulate endothelial cell responses,” Proceedings of the National Academy of Sciences of the United States of America, vol. 99, no. 21, pp. 13837–13842, 2002.
[88]  J. Prescott, C. Ye, G. Sen, and B. Hjelle, “Induction of innate immune response genes by sin nombre hantavirus does not require viral replication,” Journal of Virology, vol. 79, no. 24, pp. 15007–15015, 2005.
[89]  C. F. Spiropoulou, C. G. Albari?o, T. G. Ksiazek, and P. E. Rollin, “Andes and Prospect Hill hantaviruses differ in early induction of interferon although both can downregulate interferon signaling,” Journal of Virology, vol. 81, no. 6, pp. 2769–2776, 2007.
[90]  S. H. Shim, M.-S. Park, S. Moon et al., “Comparison of innate immune responses to pathogenic and putative non-pathogenic hantaviruses in vitro,” Virus Research, vol. 160, no. 1-2, pp. 367–373, 2011.
[91]  J. M. Emeny and M. J. Morgan, “Regulation of the interferon system: evidence that vero cells have a genetic defect in interferon production,” Journal of General Virology, vol. 43, no. 1, pp. 247–252, 1979.
[92]  M. G. Wathelet, P. M. Berr, and G. A. Huez, “Regulation of gene expression by cytokines and virus in human cells lacking the type-I interferon locus,” European Journal of Biochemistry, vol. 206, no. 3, pp. 901–910, 1992.
[93]  K. M. Jaaskelainen, P. Kaukinen, E. S. Minskaya et al., “Tula and Puumala hantavirus NSs ORFs are functional and the products inhibit activation of the interferon-beta promoter,” Journal of Medical Virology, vol. 79, no. 10, pp. 1527–1536, 2007.
[94]  J. R. Levine, J. Prescott, K. S. Brown, S. M. Best, H. Ebihara, and H. Feldmann, “Antagonism of type I interferon responses by new world hantaviruses,” Journal of Virology, vol. 84, no. 22, pp. 11790–11801, 2010.
[95]  S. L. Taylor, N. Frias-Staheli, A. García-Sastre, and C. S. Schmaljohn, “Hantaan virus nucleocapsid protein binds to importin α proteins and inhibits tumor necrosis factor alpha-induced activation of nuclear factor kappa B,” Journal of Virology, vol. 83, no. 3, pp. 1271–1279, 2009.
[96]  S. L. Taylor, R. L. Krempel, and C. S. Schmaljohn, “Inhibition of TNF-α-induced Activation of NF-κB by hantavirus nucleocapsid proteins,” Annals of the New York Academy of Sciences, vol. 1171, supplement 1, pp. E86–E93, 2009.
[97]  A. Billecocq, M. Spiegel, P. Vialat et al., “NSs protein of Rift Valley fever virus blocks interferon production by inhibiting host gene transcription,” Journal of Virology, vol. 78, no. 18, pp. 9798–9806, 2004.
[98]  G. Blakqori, S. Delhaye, M. Habjan et al., “La Crosse bunyavirus nonstructural protein NSs serves to suppress the type I interferon system of mammalian hosts,” Journal of Virology, vol. 81, no. 10, pp. 4991–4999, 2007.
[99]  M. Bouloy, C. Janzen, P. Vialat et al., “Genetic evidence for an interferon-antagonistic function of Rift Valley fever virus nonstructural protein NSs,” Journal of Virology, vol. 75, no. 3, pp. 1371–1377, 2001.
[100]  F. Weber, A. Bridgen, J. K. Fazakerley et al., “Bunyamwera Bunyavirus nonstructural protein NSs counteracts the induction of alpha/beta interferon,” Journal of Virology, vol. 76, no. 16, pp. 7949–7955, 2002.
[101]  H. H?cker, V. Redecke, B. Blagoev et al., “Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6,” Nature, vol. 439, no. 7073, pp. 204–207, 2006.
[102]  K. R. Ely and C. Li, “Structurally adaptive hot spots at a protein interaction interface on TRAF3,” Journal of Molecular Recognition, vol. 15, no. 5, pp. 286–290, 2002.
[103]  J. Q. He, S. K. Saha, J. R. Kang, B. Zarnegar, and G. Cheng, “Specificity of TRAF3 in its negative regulation of the noncanonical NF-κB pathway,” Journal of Biological Chemistry, vol. 282, no. 6, pp. 3688–3694, 2007.
[104]  C. Li, C. Z. Ni, M. L. Havert et al., “Downstream regulator TANK binds to the CD40 recognition site on TRAF3,” Structure, vol. 10, no. 3, pp. 403–411, 2002.
[105]  I. N. Gavrilovskaya, E. E. Gorbunova, N. A. Mackow, and E. R. Mackow, “Hantaviruses direct endothelial cell permeability by sensitizing cells to the vascular permeability factor VEGF, while angiopoietin 1 and sphingosine 1-phosphate inhibit hantavirus-directed permeability,” Journal of Virology, vol. 82, no. 12, pp. 5797–5806, 2008.
[106]  I. N. Gavrilovskaya, E. E. Gorbunova, and E. R. Mackow, “Pathogenic hantaviruses direct the adherence of quiescent platelets to infected endothelial cells,” Journal of Virology, vol. 84, no. 9, pp. 4832–4839, 2010.
[107]  E. Gorbunova, I. N. Gavrilovskaya, and E. R. Mackow, “Pathogenic hantaviruses Andes virus and Hantaan virus induce adherens junction disassembly by directing vascular endothelial cadherin internalization in human endothelial cell,” Journal of Virology, vol. 84, no. 14, pp. 7405–7411, 2010.
[108]  N. Kayagaki, Q. Phung, S. Chan et al., “DUBA: a deubiquitinase that regulates type I interferon production,” Science, vol. 318, no. 5856, pp. 1628–1632, 2007.
[109]  I. N. Gavrilovskaya, T. Peresleni, E. Geimonen, and E. R. Mackow, “Pathogenic hantaviruses selectively inhibit β3 integrin directed endothelial cell migration,” Archives of Virology, vol. 147, no. 10, pp. 1913–1931, 2002.
[110]  I. N. Gavrilovskaya, M. Shepley, R. Shaw, M. H. Ginsberg, and E. R. Mackow, “β3 integrins mediate the cellular entry of hantaviruses that cause respiratory failure,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 12, pp. 7074–7079, 1998.
[111]  I. N. Gavrilovskaya, E. J. Brown, M. H. Ginsberg, and E. R. Mackow, “Cellular entry of hantaviruses which cause hemorrhagic fever with renal syndrome is mediated by β3 integrins,” Journal of Virology, vol. 73, no. 5, pp. 3951–3959, 1999.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413