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...

Roles for Endothelial Cells in Dengue Virus Infection

DOI: 10.1155/2012/840654

Full-Text   Cite this paper   Add to My Lib

Abstract:

Dengue viruses cause two severe diseases that alter vascular fluid barrier functions, dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). The endothelium is the primary fluid barrier of the vasculature and ultimately the effects of dengue virus infection that cause capillary leakage impact endothelial cell (EC) barrier functions. The ability of dengue virus to infect the endothelium provides a direct means for dengue to alter capillary permeability, permit virus replication, and induce responses that recruit immune cells to the endothelium. Recent studies focused on dengue virus infection of primary ECs have demonstrated that ECs are efficiently infected, rapidly produce viral progeny, and elicit immune enhancing cytokine responses that may contribute to pathogenesis. Furthermore, infected ECs have also been implicated in enhancing viremia and immunopathogenesis within murine dengue disease models. Thus dengue-infected ECs have the potential to directly contribute to immune enhancement, capillary permeability, viremia, and immune targeting of the endothelium. These effects implicate responses of the infected endothelium in dengue pathogenesis and rationalize therapeutic targeting of the endothelium and EC responses as a means of reducing the severity of dengue virus disease. 1. Introduction Dengue viruses are transmitted by mosquitoes and infect ~ 50 million people annually with an additional 2.5 billion people at risk living in tropical areas [1–3]. Expanding mosquito habitats are increasing the range of dengue virus outbreaks and the occurrence of severe diseases with 5–30% mortality rates: dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) [1–3]. The majority of patients are asymptomatic or display mild symptoms of dengue fever (DF) which include rapid onset of fever, viremia, headache, pain, and rash [4]. Patients with DHF and DSS display symptoms of DF in addition to increased edema, hemorrhage, thrombocytopenia, and shock [1–3]. Although patient progression to DHF and DSS is not fully understood [3, 5], antibody-dependent enhancement (ADE) of dengue infection increases the potential for DSS and DHF [3, 6, 7]. There are four dengue virus serotypes (types 1–4) and infection by one serotype predisposes individuals to more severe disease following a subsequent infection by a different dengue serotype. The circulation of serotype-specific cross-reactive antibodies or preexisting maternal antibodies may contribute to progression to DHF/DSS by facilitating viral infection of immune cells and eliciting cytokine and chemotactic

References

[1]  D. J. Gubler, “Dengue/dengue haemorrhagic fever: history and current status,” Novartis Foundation Symposium, vol. 277, pp. 3–16, 2006.
[2]  S. B. Halstead, “Dengue,” The Lancet, vol. 370, no. 9599, pp. 1644–1652, 2007.
[3]  S. B. Halstead, “Pathophysiology,” in Dengue, S. B. Halstead, Ed., vol. 5, pp. 265–326, Imperial College Press, London, UK, 2008.
[4]  B. E. E. Martina, P. Koraka, and A. D. M. E. Osterhaus, “Dengue virus pathogenesis: an integrated view,” Clinical Microbiology Reviews, vol. 22, no. 4, pp. 564–581, 2009.
[5]  A. Basu and U. C. Chaturvedi, “Vascular endothelium: the battlefield of dengue viruses,” FEMS Immunology and Medical Microbiology, vol. 53, no. 3, pp. 287–299, 2008.
[6]  A. Takada and Y. Kawaoka, “Antibody-dependent enhancement of viral infection: molecular mechanisms and in vivo implications,” Reviews in Medical Virology, vol. 13, no. 6, pp. 387–398, 2003.
[7]  S. B. Halstead, “Antibodies determine virulence in Dengue,” Annals of the New York Academy of Sciences, vol. 1171, no. 1, pp. E48–E56, 2009.
[8]  R. M. Zellweger, T. R. Prestwood, and S. Shresta, “Enhanced infection of liver sinusoidal endothelial cells in a mouse model of antibody-induced severe Dengue disease,” Cell Host and Microbe, vol. 7, no. 2, pp. 128–139, 2010.
[9]  K. Jessie, M. Y. Fong, S. Devi, S. K. Lam, and K. T. Wong, “Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization,” Journal of Infectious Diseases, vol. 189, no. 8, pp. 1411–1418, 2004.
[10]  A. Nisalak, S. B. Halstead, P. Singharaj, S. Udomsakdi, S. W. Nye, and K. Vinijchaikul, “Observations related to pathogenesis of dengue hemorrhagic fever. 3. Virologic studies of fatal disease.,” Yale Journal of Biology and Medicine, vol. 42, no. 5, pp. 293–310, 1970.
[11]  M. R. Huerre, N. Trong Lan, P. Marianneau et al., “Liver histopathology and biological correlates in five cases of fatal dengue fever in Vietnamese children,” Virchows Archiv, vol. 438, no. 2, pp. 107–115, 2001.
[12]  L. Rosen, M. T. Drouet, and V. Deubel, “Detection of dengue virus RNA by reverse transcription-polymerase chain reaction in the liver and lymphoid organs but not in the brain in fatal human infection,” American Journal of Tropical Medicine and Hygiene, vol. 61, no. 5, pp. 720–724, 1999.
[13]  L. Rosen, M. M. Khin, and T. U, “Recovery of virus from the liver of children with fatal dengue: reflections on the pathogenesis of the disease and its possible analogy with that of yellow fever,” Research in Virology, vol. 140, no. 4, pp. 351–360, 1989.
[14]  D. M. Salgado, J. M. Eltit, K. Mansfield et al., “Heart and skeletal muscle are targets of dengue virus infection,” Pediatric Infectious Disease Journal, vol. 29, no. 3, pp. 238–242, 2010.
[15]  J. Fink, F. Gu, L. Ling, et al., “Host gene expression profiling of dengue virus infection in cell lines and patients,” PLoS Neglected Tropical Diseases, vol. 1, no. 2, p. e86, 2007.
[16]  P. Avirutnan, N. Punyadee, S. Noisakran et al., “Vascular leakage in severe dengue virus infections: a potential role for the nonstructural viral protein NS1 and complement,” Journal of Infectious Diseases, vol. 193, no. 8, pp. 1078–1088, 2006.
[17]  K. A. Stoermer and T. E. Morrison, “Complement and viral pathogenesis,” Virology, vol. 411, no. 2, pp. 362–373, 2011.
[18]  T. H. Nguyen, H. Y. Lei, T. L. Nguyen et al., “Dengue hemorrhagic fever in infants: a study of clinical and cytokine profiles,” Journal of Infectious Diseases, vol. 189, no. 2, pp. 221–232, 2004.
[19]  A. R. Johnson, T. E. Hugli, and H. J. Mueller Eberhard, “Release of histamine from rat mast cells by the complement peptides C3a and C5a,” Immunology, vol. 28, no. 6, pp. 1067–1080, 1975.
[20]  G. Nilsson, M. Johnell, C. H. Hammer et al., “C3a and C5a are chemotaxins for human mast cells and act through distinct receptors via a pertussis toxin-sensitive signal transduction pathway,” Journal of Immunology, vol. 157, no. 4, pp. 1693–1698, 1996.
[21]  C. Kemper and D. E. Hourcade, “Properdin: new roles in pattern recognition and target clearance,” Molecular Immunology, vol. 45, no. 16, pp. 4048–4056, 2009.
[22]  C. Kemper, L. M. Mitchell, L. Zhang, and D. E. Hourcade, “The complement protein properdin binds apoptotic T cells and promotes complement activation and phagocytosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 26, pp. 9023–9028, 2008.
[23]  M. Tuchinda, B. Dhorranintra, and P. Tuchinda, “Histamine content in 24 hour urine in patients with dengue haemorrhagic fever,” Southeast Asian Journal of Tropical Medicine and Public Health, vol. 8, no. 1, pp. 80–83, 1977.
[24]  A. L. S. John, A. P. S. Rathore, H. Yap et al., “Immune surveillance by mast cells during dengue infection promotes natural killer (NK) and NKT-cell recruitment and viral clearance,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 22, pp. 9190–9195, 2011.
[25]  A. Srikiatkhachorn, “Plasma leakage in dengue haemorrhagic fever,” Thrombosis and Haemostasis, vol. 102, no. 6, pp. 1042–1049, 2009.
[26]  P. Avirutnan, P. Malasit, B. Seliger, S. Bhakdi, and M. Husmann, “Dengue virus infection of human endothelial cells leads to chemokine production, complement activation, and apoptosis,” Journal of Immunology, vol. 161, no. 11, pp. 6338–6346, 1998.
[27]  A. Azizan, J. Sweat, C. Espino, J. Gemmer, L. Stark, and D. Kazanis, “Differential proinflammatory and angiogenesis-specific cytokine production in human pulmonary endothelial cells, HPMEC-ST1.6R infected with dengue-2 and dengue-3 virus,” Journal of Virological Methods, vol. 138, no. 1-2, pp. 211–217, 2006.
[28]  I. Kurane, B. L. Innis, S. Nimmannitya et al., “Human immune responses to dengue viruses.,” The Southeast Asian Journal of Tropical Medicine and Public Health, vol. 21, no. 4, pp. 658–662, 1990.
[29]  S. Green, D. W. Vaughn, S. Kalayanarooj et al., “Early immune activation in acute dengue illness is related to development of plasma leakage and disease severity,” Journal of Infectious Diseases, vol. 179, no. 4, pp. 755–762, 1999.
[30]  S. J. Balsitis, J. Coloma, G. Castro et al., “Tropism of dengue virus in mice and humans defined by viral nonstructural protein 3-specific immunostaining,” American Journal of Tropical Medicine and Hygiene, vol. 80, no. 3, pp. 416–424, 2009.
[31]  S. J. Balsitis and E. Harris, “Animal models of dengue virus infection and disease: applications, insights and frontiers,” in Frontiers in Dengue Virus Research, K. A. Hanley and S. C. Weaver, Eds., pp. 103–115, Caister Academic Press, 2010.
[32]  H. F. Dvorak, “Vascular permeability to plasma, plasma proteins, and cells: an update,” Current Opinion in Hematology, vol. 17, no. 3, pp. 225–229, 2010.
[33]  D. B. Cines, E. S. Pollak, C. A. Buck et al., “Endothelial cells in physiology and in the pathophysiology of vascular disorders,” Blood, vol. 91, no. 10, pp. 3527–3561, 1998.
[34]  W. C. Aird, “Endothelium as an organ system,” Critical Care Medicine, vol. 32, no. 5, supplement, pp. S271–279, 2004.
[35]  G. Valbuena and D. H. Walker, “The endothelium as a target for infections,” Annual Review of Pathology, vol. 1, pp. 171–198, 2006.
[36]  M. G. Lampugnani and E. Dejana, “Adherens junctions in endothelial cells regulate vessel maintenance and angiogenesis,” Thrombosis Research, vol. 120, supplement 2, pp. S1–S6, 2007.
[37]  R. V. Warke, K. Xhaja, K. J. Martin et al., “Dengue virus induces novel changes in gene expression of human umbilical vein endothelial cells,” Journal of Virology, vol. 77, no. 21, pp. 11822–11832, 2004.
[38]  S. Kalayanarooj, D. W. Vaughn, S. Nimmannitya et al., “Early clinical and laboratory indicators of acute dengue illness,” Journal of Infectious Diseases, vol. 176, no. 2, pp. 313–321, 1997.
[39]  J. E. Cardier, B. Rivas, E. Romano et al., “Evidence of vascular damage in dengue disease: demonstration of high levels of soluble cell adhesion molecules and circulating endothelial cells,” Endothelium, vol. 13, no. 5, pp. 335–340, 2006.
[40]  D. Sosothikul, P. Seksarn, S. Pongsewalak, U. Thisyakorn, and J. Lusher, “Activation of endothelial cells, coagulation and fibrinolysis in children with Dengue virus infection,” Thrombosis and Haemostasis, vol. 97, no. 4, pp. 627–634, 2007.
[41]  M. G. Lampugnani and E. Dejana, “The control of endothelial cell functions by adherens junctions,” Novartis Foundation Symposium, vol. 283, pp. 4–13, 2007.
[42]  P. Malasit, “Complement and dengue haemorrhagic fever/shock syndrome,” The Southeast Asian Journal of Tropical Medicine and Public Health, vol. 18, no. 3, pp. 316–320, 1987.
[43]  V. Churdboonchart, N. Bhamarapravati, and P. Futrakul, “Crossed immunoelectrophoresis for the detection of split products of the third complement in dengue hemorrhagic fever. I. Observations in patients' plasma,” American Journal of Tropical Medicine and Hygiene, vol. 32, no. 3, pp. 569–576, 1983.
[44]  K. Nishioka, “Serum complement level in dengue hemorrhagic fever,” Allergie und Immunologie, vol. 20-21, no. 4, pp. 385–392, 1974.
[45]  M. F. Shaio, F. Y. Chang, and S. C. Hou, “Complement pathway activity in serum from patients with classical dengue fever,” Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 86, no. 6, pp. 672–675, 1992.
[46]  N. A. Dalrymple and E. R. Mackow, “Endothelial cells elicit immune-enhancing responses to dengue virus infection,” Journal of Virology, vol. 86, no. 12, pp. 6408–6415, 2012.
[47]  S. Alcon-LePoder, M. T. Drouet, P. Roux et al., “The secreted form of dengue virus nonstructural protein NS1 is endocytosed by hepatocytes and accumulates in late endosomes: implications for viral infectivity,” Journal of Virology, vol. 79, no. 17, pp. 11403–11411, 2005.
[48]  M. G. Jacobs, P. J. Robinson, C. Bletchly, J. M. Mackenzie, and P. R. Young, “Dengue virus nonstructural protein 1 is expressed in a glycosyl-phosphatidylinositol-linked form that is capable of signal transduction,” FASEB Journal, vol. 14, no. 11, pp. 1603–1610, 2000.
[49]  S. Noisakran, T. Dechtawewat, P. Avirutnan et al., “Association of dengue virus NS1 protein with lipid rafts,” Journal of General Virology, vol. 89, no. 10, pp. 2492–2500, 2008.
[50]  D. H. Libraty, P. R. Young, D. Pickering et al., “High circulating levels of the dengue virus nonstructural protein NS1 early in dengue illness correlate with the development of dengue hemorrhagic fever,” Journal of Infectious Diseases, vol. 186, no. 8, pp. 1165–1168, 2002.
[51]  C. F. Lin, H. Y. Lei, A. L. Shiau et al., “Antibodies from dengue patient sera cross-react with endothelial cells and induce damage,” Journal of Medical Virology, vol. 69, no. 1, pp. 82–90, 2003.
[52]  C. F. Lin, H. Y. Lei, A. L. Shiau et al., “Endothelial cell apoptosis induced by antibodies against dengue virus nonstructural protein 1 via production of nitric oxide,” Journal of Immunology, vol. 169, no. 2, pp. 657–664, 2002.
[53]  Y. S. Lin, C. F. Lin, H. Y. Lei et al., “Antibody-mediated endothelial cell damage via nitric oxide,” Current Pharmaceutical Design, vol. 10, no. 2, pp. 213–221, 2004.
[54]  P. Avirutnan, L. Zhang, N. Punyadee et al., “Secreted NS1 of dengue virus attaches to the surface of cells via interactions with heparan sulfate and chondroitin sulfate E,” PLoS Pathogens, vol. 3, no. 11, article e183, 2007.
[55]  K. M. Chung, B. S. Thompson, D. H. Fremont, and M. S. Diamond, “Antibody recognition of cell surface-associated NS1 triggers Fc-γ receptor-mediated phagocytosis and clearance of West Nile virus-infected cells,” Journal of Virology, vol. 81, no. 17, pp. 9551–9555, 2007.
[56]  K. M. Chung, G. E. Nybakken, B. S. Thompson et al., “Antibodies against West Nile virus nonstructural protein NS1 prevent lethal infection through Fc γ receptor-dependent and -independent mechanisms,” Journal of Virology, vol. 80, no. 3, pp. 1340–1351, 2006.
[57]  P. Avirutnan, R. E. Hauhart, P. Somnuke, A. M. Blom, M. S. Diamond, and J. P. Atkinson, “Binding of flavivirus nonstructural protein NS1 to C4b binding protein modulates complement activation,” Journal of Immunology, vol. 187, no. 1, pp. 424–433, 2011.
[58]  P. Avirutnan, A. Fuchs, R. E. Hauhart et al., “Antagonism of the complement component C4 by flavivirus nonstructural protein NS1,” Journal of Experimental Medicine, vol. 207, no. 4, pp. 793–806, 2010.
[59]  C. F. Lin, S. C. Chiu, Y. L. Hsiao et al., “Expression of cytokine, chemokine, and adhesion molecules during endothelial cell activation induced by antibodies against dengue virus nonstructural protein 1,” Journal of Immunology, vol. 174, no. 1, pp. 395–403, 2005.
[60]  T. E. Mollnes and M. Kirschfink, “Strategies of therapeutic complement inhibition,” Molecular Immunology, vol. 43, no. 1-2, pp. 107–121, 2006.
[61]  J. M. Thurman, D. M. Kraus, G. Girardi et al., “A novel inhibitor of the alternative complement pathway prevents antiphospholipid antibody-induced pregnancy loss in mice,” Molecular Immunology, vol. 42, no. 1, pp. 87–97, 2005.
[62]  B. P. Morgan and C. L. Harris, “Complement therapeutics; history and current progress,” Molecular Immunology, vol. 40, no. 2–4, pp. 159–170, 2003.
[63]  A. ündar, H. C. Eichstaedt, F. J. Clubb et al., “Novel anti-factor D monoclonal antibody inhibits complement and leukocyte activation in a baboon model of cardiopulmonary bypass,” Annals of Thoracic Surgery, vol. 74, no. 2, pp. 355–362, 2002.
[64]  J. R. Teijaro, K. B. Walsh, S. Cahalan, et al., “Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection,” Cell, vol. 146, no. 6, pp. 980–991, 2011.
[65]  J. A. Pawitan, “Potential agents against plasma leakage,” ISRN Pharmacol, vol. 2011, Article ID 975048, 2011.
[66]  S. J. Balsitis, K. L. Williams, R. Lachica et al., “Lethal antibody enhancement of dengue disease in mice is prevented by Fc modification,” PLoS Pathogens, vol. 6, no. 2, Article ID e1000790, 2010.
[67]  G. K. Tan, J. K. W. Ng, S. L. Trasti, W. Schul, G. Yip, and S. Alonso, “A non mouse-adapted dengue virus strain as a new model of severe dengue infection in AG129 mice,” PLoS Neglected Tropical Diseases, vol. 4, no. 4, article e672, 2010.
[68]  S. Shresta, K. L. Sharar, D. M. Prigozhin, P. R. Beatty, and E. Harris, “Murine model for dengue virus-induced lethal disease with increased vascular permeability,” Journal of Virology, vol. 80, no. 20, pp. 10208–10217, 2006.
[69]  H. C. Chen, F. M. Hofman, J. T. Kung, Y. D. Lin, and B. A. Wu-Hsieh, “Both virus and tumor necrosis factor alpha are critical for endothelium damage in a mouse model of dengue virus-induced hemorrhage,” Journal of Virology, vol. 81, no. 11, pp. 5518–5526, 2007.
[70]  M. T. Arévalo, P. J. Simpson-Haidaris, Z. Kou, J. J. Schlesinger, and X. Jin, “Primary human endothelial cells support direct but not antibody-dependent enhancement of dengue viral infection,” Journal of Medical Virology, vol. 81, no. 3, pp. 519–528, 2009.
[71]  K. Elvevold, B. Smedsr?d, and I. Martinez, “The liver sinusoidal endothelial cell: a cell type of controversial and confusing identity,” American Journal of Physiology, vol. 294, no. 2, pp. G391–G400, 2008.
[72]  E. Navarro-Sánchez, P. Desprès, and L. Cedillo-Barrón, “Innate immune responses to dengue virus,” Archives of Medical Research, vol. 36, no. 5, pp. 425–435, 2005.
[73]  M. S. Diamond and E. Harris, “Interferon inhibits dengue virus infection by preventing translation of viral RNA through a PKR-independent mechanism,” Virology, vol. 289, no. 2, pp. 297–311, 2001.
[74]  M. S. Diamond, T. G. Roberts, D. Edgil, B. Lu, J. Ernst, and E. Harris, “Modulation of dengue virus infection in human cells by alpha, beta, and gamma interferons,” Journal of Virology, vol. 74, no. 11, pp. 4957–4966, 2000.
[75]  J. L. Mu?oz-Jordán, G. G. Sánchez-Burgos, M. Laurent-Rolle, and A. García-Sastre, “Inhibition of interferon signaling by dengue virus,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 2, pp. 14333–14338, 2003.
[76]  D. Gomez and N. C. Reich, “Stimulation of primary human endothelial cell proliferation by IFN1,” Journal of Immunology, vol. 170, no. 11, pp. 5373–5381, 2003.
[77]  A. J. Johnson and J. T. Roehrig, “New mouse model for dengue virus vaccine testing,” Journal of Virology, vol. 73, no. 1, pp. 783–786, 1999.
[78]  J. Ashour, M. Laurent-Rolle, P. Y. Shi, and A. García-Sastre, “NS5 of dengue virus mediates STAT2 binding and degradation,” Journal of Virology, vol. 83, no. 11, pp. 5408–5418, 2009.
[79]  J. Ashour, J. Morrison, M. Laurent-Rolle et al., “Mouse STAT2 restricts early dengue virus replication,” Cell Host and Microbe, vol. 8, no. 5, pp. 410–421, 2010.
[80]  M. C. Cassetti, A. Durbin, E. Harris et al., “Report of an NIAID workshop on dengue animal models,” Vaccine, vol. 28, no. 26, pp. 4229–4234, 2010.
[81]  C. A. Sariol, J. L. Mu?oz-Jordan, K. Abel et al., “Transcriptional activation of interferon-stimulated genes but not of cytokine genes after primary infection of rhesus macaques with dengue virus type 1,” Clinical and Vaccine Immunology, vol. 14, no. 6, pp. 756–766, 2007.
[82]  R. E. Unger, V. Krump-Konvalinkova, K. Peters, and C. James Kirkpatrick, “In vitro expression of the endothelial phenotype: comparative study of primary isolated cells and cell lines, including the novel cell line HPMEC-ST1.6R,” Microvascular Research, vol. 64, no. 3, pp. 384–397, 2002.
[83]  F. Kiessling, C. Haller, and J. Kartenbeck, “Cell-cell contacts in the human cell line ECV304 exhibit both endothelial and epithelial characteristics,” Cell and Tissue Research, vol. 297, no. 1, pp. 131–140, 1999.
[84]  W. G. Dirks, R. A. F. Macleod, and H. G. Drexler, “ECV304 (endothelial) is really T24 (bladder carcinoma): cell line cross- contamination at source,” In Vitro Cellular and Developmental Biology, vol. 35, no. 10, pp. 558–559, 1999.
[85]  I. Bosch, K. Xhaja, L. Estevez et al., “Increased production of interleukin-8 in primary human monocytes and in human epithelial and endothelial cell lines after dengue virus challenge,” Journal of Virology, vol. 76, no. 11, pp. 5588–5597, 2002.
[86]  A. Bunyaratvej, P. Butthep, S. Yoksan, and N. Bhamarapravati, “Dengue viruses induce cell proliferation and morphological changes of endothelial cells,” Southeast Asian Journal of Tropical Medicine and Public Health, vol. 28, supplement 3, pp. 32–37, 1997.
[87]  Y. H. Huang, H. Y. Lei, H. S. Liu, Y. S. Lin, C. C. Liu, and T. M. Yeh, “Dengue virus infects human endothelial cells and induces IL-6 and IL-8 production,” American Journal of Tropical Medicine and Hygiene, vol. 63, no. 1-2, pp. 71–75, 2000.
[88]  N. Dalrymple and E. R. Mackow, “Productive dengue virus infection of human endothelial cells is directed by heparan sulfate-containing proteoglycan receptors,” Journal of Virology, vol. 85, pp. 9478–9485, 2011.
[89]  D. Talavera, A. M. Castillo, M. C. Dominguez, A. Escobar Gutierrez, and I. Meza, “IL8 release, tight junction and cytoskeleton dynamic reorganization conducive to permeability increase are induced by dengue virus infection of microvascular endothelial monolayers,” Journal of General Virology, vol. 85, no. 7, pp. 1801–1813, 2004.
[90]  K. Okamoto, H. Kinoshita, C. Parquet Mdel, et al., “Dengue virus strain DEN2 16681 utilizes a specific glycochain of syndecan-2 proteoglycan as a receptor,” Journal of General Virology, vol. 93, no. 4, pp. 761–770, 2012.
[91]  R. Anderson, S. Wang, C. Osiowy, and A. C. Issekutz, “Activation of endothelial cells via antibody-enhanced dengue virus infection of peripheral blood monocytes,” Journal of Virology, vol. 71, no. 6, pp. 4226–4232, 1997.
[92]  C. N. Peyrefitte, B. Pastorino, G. E. Grau, J. Lou, H. Tolou, and P. Couissinier-Paris, “Dengue virus infection of human microvascular endothelial cells from different vascular beds promotes both common and specific functional changes,” Journal of Medical Virology, vol. 78, no. 2, pp. 229–242, 2006.
[93]  B. E. Dewi, T. Takasaki, and I. Kurane, “Peripheral blood mononuclear cells increase the permeability of dengue virus-infected endothelial cells in association with downregulation of vascular endothelial cadherin,” Journal of General Virology, vol. 89, no. 3, pp. 642–652, 2008.
[94]  M. Corada, M. Mariotti, G. Thurston et al., “Vascular endothelial-cadherin is an important determinant of microvascular integrity in vivo,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 17, pp. 9815–9820, 1999.
[95]  A. Azizan, K. Fitzpatrick, A. Signorovitz et al., “Profile of time-dependent VEGF upregulation in human pulmonary endothelial cells, HPMEC-ST1.6R infected with DENV-1, -2, -3, and -4 viruses,” Virology Journal, vol. 6, article 49, 2009.
[96]  M. Rampart, J. Van Damme, L. Zonnekeyn, and A. G. Herman, “Granulocyte chemotactic protein/interleukin-8 induces plasma leakage and neutrophil accumulation in rabbit skin,” American Journal of Pathology, vol. 135, no. 1, pp. 21–25, 1989.
[97]  M. E. W. Hammond, G. R. Lapointe, P. H. Feucht et al., “IL-8 induces neutrophil chemotaxis predominantly via type I IL-8 receptors,” Journal of Immunology, vol. 155, no. 3, pp. 1428–1433, 1995.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133