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...

Skewed Helper T-Cell Responses to IL-12 Family Cytokines Produced by Antigen-Presenting Cells and the Genetic Background in Behcet’s Disease

DOI: 10.1155/2013/363859

Full-Text   Cite this paper   Add to My Lib

Abstract:

Behcet’s disease (BD) is a multisystemic inflammatory disease and is characterized by recurrent attacks on eyes, brain, skin, and gut. There is evidence that skewed T-cell responses contributed to its pathophysiology in patients with BD. Recently, we found that Th17 cells, a new helper T (Th) cell subset, were increased in patients with BD, and both Th type 1 (Th1) and Th17 cell differentiation signaling pathways were overactivated. Several researches revealed that genetic polymorphisms in Th1/Th17 cell differentiation signaling pathways were associated with the onset of BD. Here, we summarize current findings on the Th cell subsets, their contribution to the pathogenesis of BD and the genetic backgrounds, especially in view of IL-12 family cytokine production and pattern recognition receptors of macrophages/monocytes. 1. Introduction Behcet’s disease (BD) is a systemic inflammatory disease, characterized by recurrent signs and symptoms of oral aphthosis, genital ulcers, skin lesions, and uveitis. BD is not chronic inflammatory disease, but patients with BD suffer from recurrent attacks of acute and self-limiting inflammation. Repeated attacks of uveitis can lead to blindness. The etiology of BD is largely unknown and skewed T-cell responses are associated with the development and maintenance of BD [1]. Excessive cytokine production by Th1 cells was reported using immunohistochemistry [2, 3] and intracellular cytokine staining [4, 5]. Th1 dominance was observed in BD uveitis [6] and stomatitis as well [7]. We reported excessive Th1 cell infiltration in BD skin and intestinal lesions but interleukin- (IL-) producing T cells were rarely detected [8–10]. T cells and peripheral blood mononuclear cells (PBMC) from patients with BD responded to KTH1 antigens of Streptococcus sanguinis in oral cavity of patients with BD and produced interferon γ (IFNγ) and IL-12 [11]. Recently, Th1/Th2 paradigm was challenged by the discovery of various subsets of Th cells, such as Th17 cells and regulatory T (Treg) cells [12] (Figure 1). Researchers showed that Th cell differentiation in each subset was closely related and sometimes converted into another subset in response to environmental signals both in peripheral blood and in organs [13]. Recent studies on innate immune system suggested that antigen-presenting cells (APC) stimulated with pattern-recognition receptors (PRR) and corresponding ligands regulated Th cell differentiation by cytokine production [14]. Figure 1: Current view of helper T (Th) cell subsets in humans [ 12]. Na?ve Th cells differentiate into several

References

[1]  T. Sakane, M. Takeno, N. Suzuki, and G. Inaba, “Behcet's disease,” The New England Journal of Medicine, vol. 341, no. 17, pp. 1284–1291, 1999.
[2]  M. Melikoglu, S. Uysal, J. G. Krueger et al., “Characterization of the divergent wound-healing responses occurring in the pathergy reaction and normal healthy volunteers,” The Journal of Immunology, vol. 177, no. 9, pp. 6415–6421, 2006.
[3]  M. Ben Ahmed, H. Houman, M. Miled, K. Dellagi, and H. Louzir, “Involvement of chemokines and Th1 cytokines in the pathogenesis of mucocutaneous lesions of Beh?et's disease,” Arthritis and Rheumatism, vol. 50, no. 7, pp. 2291–2295, 2004.
[4]  S. Koarada, Y. Haruta, Y. Tada et al., “Increased entry of CD4+ T cells into the Th1 cytokine effector pathway during T-cell division following stimulation in Beh?et's disease,” Rheumatology, vol. 43, no. 7, pp. 843–851, 2004.
[5]  H. Houman, A. Hamzaoui, I. Ben Ghorbal, M. Khanfir, M. Feki, and K. Hamzaoui, “Abnormal expression of chemokine receptors in Beh?et's disease: relationship to intracellular Th1/Th2 cytokines and to clinical manifestations,” Journal of Autoimmunity, vol. 23, no. 3, pp. 267–273, 2004.
[6]  F. Ilhan, T. Demir, P. Türk?üo?lu, B. Turgut, N. Demir, and A. G?dekmerdan, “Th1 polarization of the immune response in uveitis in Beh?et's disease,” Canadian Journal of Ophthalmology, vol. 43, no. 1, pp. 105–108, 2008.
[7]  A. M. Dalghous, J. Freysdottir, and F. Fortune, “Expression of cytokines, chemokines, and chemokine receptors in oral ulcers of patients with Behcet's disease (BD) and recurrent aphthous stomatitis is Th1-associated, although Th2-association is also observed in patients with BD,” Scandinavian Journal of Rheumatology, vol. 35, no. 6, pp. 472–475, 2006.
[8]  K. Nara, M. S. Kurokawa, S. Chiba et al., “Involvement of innate immunity in the pathogenesis of intestinal Beh?et's disease,” Clinical and Experimental Immunology, vol. 152, no. 2, pp. 245–251, 2008.
[9]  Y. Imamura, M. S. Kurokawa, H. Yoshikawa et al., “Involvement of Th1 cells and heat shock protein 60 in the pathogenesis of intestinal Beh?et's disease,” Clinical and Experimental Immunology, vol. 139, no. 2, pp. 371–378, 2005.
[10]  H. Nagafuchi, M. Takeno, H. Yoshikawa et al., “Excessive expression of Txk, a member of the Tec family of tyrosine kinases, contributes to excessive Th1 cytokine production by T lymphocytes in patients with Behcet's disease,” Clinical and Experimental Immunology, vol. 139, no. 2, pp. 363–370, 2005.
[11]  H. Yanagihori, N. Oyama, K. Nakamura, N. Mizuki, K. Oguma, and F. Kaneko, “Role of IL-12B promoter polymorphism in Adamantiades-Behcet's disease susceptibility: an involvement of Th1 immunoreactivity against Streptococcus sanguinis antigen,” Journal of Investigative Dermatology, vol. 126, no. 7, pp. 1534–1540, 2006.
[12]  M. T. Palmer and C. T. Weaver, “Autoimmunity: increasing suspects in the CD4+ T cell lineup,” Nature Immunology, vol. 11, no. 1, pp. 36–40, 2010.
[13]  R. Basu, R. D. Hatton, and C. T. Weaver, “The Th17 family: flexibility follows function,” Immunological Reviews, vol. 252, no. 1, pp. 89–103, 2013.
[14]  J. M. Reynolds and C. Dong, “Toll-like receptor regulation of effector T lymphocyte function,” Trends in Immunology, vol. 34, no. 10, pp. 511–519, 2013.
[15]  L. Lu, J. Wang, F. Zhang et al., “Role of SMAD and non-SMAD signals in the development of Th17 and regulatory T cells,” The Journal of Immunology, vol. 184, no. 8, pp. 4295–4306, 2010.
[16]  E. Bettelli, Y. Carrier, W. Gao et al., “Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells,” Nature, vol. 441, no. 7090, pp. 235–238, 2006.
[17]  D. A. Vignali and V. K. Kuchroo, “IL-12 family cytokines: immunological playmakers,” Nature Immunology, vol. 13, no. 8, pp. 722–728, 2012.
[18]  R. M. Pope and S. Shahrara, “Possible roles of IL-12-family cytokines in rheumatoid arthritis,” Nature Reviews, Rheumatology, vol. 9, no. 4, pp. 252–256, 2013.
[19]  C. Parham, M. Chirica, J. Timans et al., “A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rβ1 and a novel cytokine receptor subunit, IL-23R,” The Journal of Immunology, vol. 168, no. 11, pp. 5699–5708, 2002.
[20]  J. S. Stumhofer, J. S. Silver, A. Laurence et al., “Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10,” Nature Immunology, vol. 8, no. 12, pp. 1363–1371, 2007.
[21]  L. W. Collison, C. J. Workman, T. T. Kuo et al., “The inhibitory cytokine IL-35 contributes to regulatory T-cell function,” Nature, vol. 450, no. 7169, pp. 566–569, 2007.
[22]  E. Bardel, F. Larousserie, P. Charlot-Rabiega, A. Coulomb-L'Herminé, and O. Devergne, “Human CD4+CD25+Foxp3+ regulatory T cells do not constitutively express IL-35,” The Journal of Immunology, vol. 181, no. 10, pp. 6898–6905, 2008.
[23]  C. Garbers, H. M. Hermanns, F. Schaper, et al., “Plasticity and cross-talk of interleukin 6-type cytokines,” Cytokine & Growth Factor Reviews, vol. 23, no. 3, pp. 85–97, 2012.
[24]  F. Annunziato, L. Cosmi, F. Liotta, E. Maggi, and S. Romagnani, “Defining the human T helper 17 cell phenotype,” Trends in Immunology, vol. 33, no. 10, pp. 505–512, 2012.
[25]  P. Muranski and N. P. Restifo, “Essentials of Th17 cell commitment and plasticity,” Blood, vol. 121, no. 13, pp. 2402–2414, 2013.
[26]  N. J. Wilson, K. Boniface, J. R. Chan et al., “Development, cytokine profile and function of human interleukin 17-producing helper T cells,” Nature Immunology, vol. 8, no. 9, pp. 950–957, 2007.
[27]  B. W. Kirkham, M. N. Lassere, J. P. Edmonds et al., “Synovial membrane cytokine expression is predictive of joint damage progression in rheumatoid arthritis: a two-year prospective study (the DAMAGE study cohort),” Arthritis and Rheumatism, vol. 54, no. 4, pp. 1122–1131, 2006.
[28]  L. J. Edwards, R. A. Robins, and C. S. Constantinescu, “Th17/Th1 phenotype in demyelinating disease,” Cytokine, vol. 50, no. 1, pp. 19–23, 2010.
[29]  Z. Hovhannisyan, J. Treatman, D. R. Littman, and L. Mayer, “Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases,” Gastroenterology, vol. 140, no. 3, pp. 957–965, 2011.
[30]  F. Annunziato, L. Cosmi, V. Santarlasci et al., “Phenotypic and functional features of human Th17 cells,” The Journal of Experimental Medicine, vol. 204, no. 8, pp. 1849–1861, 2007.
[31]  A. H?nsel, C. Günther, J. Ingwersen et al., “Human slan (6-sulfo LacNAc) dendritic cells are inflammatory dermal dendritic cells in psoriasis and drive strong Th17/Th1 T-cell responses,” Journal of Allergy and Clinical Immunology, vol. 127, no. 3, pp. 787–794, 2011.
[32]  H. Kebir, I. Ifergan, J. I. Alvarez et al., “Preferential recruitment of interferon-γ-expressing TH17 cells in multiple sclerosis,” Annals of Neurology, vol. 66, no. 3, pp. 390–402, 2009.
[33]  K. Nistala, S. Adams, H. Cambrook et al., “Th17 plasticity in human autoimmune arthritis is driven by the inflammatory environment,” Proceedings of the National Academy of Sciences of the United States of America, vol. 107, no. 33, pp. 14751–14756, 2010.
[34]  L. Cosmi, R. Cimaz, L. Maggi et al., “Evidence of the transient nature of the Th17 phenotype of CD4+CD161+ T cells in the synovial fluid of patients with juvenile idiopathic arthritis,” Arthritis and Rheumatism, vol. 63, no. 8, pp. 2504–2515, 2011.
[35]  M. Miyara, G. Gorochov, M. Ehrenstein, L. Musset, S. Sakaguchi, and Z. Amoura, “Human FoxP3+ regulatory T cells in systemic autoimmune diseases,” Autoimmunity Reviews, vol. 10, no. 12, pp. 744–755, 2011.
[36]  S. Sakaguchi, D. A. Vignali, A. Y. Rudensky, R. E. Niec, and H. Waldmann, “The plasticity and stability of regulatory T cells,” Nature Reviews Immunology, vol. 13, no. 6, pp. 461–467, 2013.
[37]  X. O. Yang, R. Nurieva, G. J. Martinez et al., “Molecular antagonism and plasticity of regulatory and inflammatory T cell programs,” Immunity, vol. 29, no. 1, pp. 44–56, 2008.
[38]  N. Ohkura, Y. Kitagawa, and S. Sakaguchi, “Development and maintenance of regulatory T cells,” Immunity, vol. 38, no. 3, pp. 414–423, 2013.
[39]  K. Hamzaoui, E. Bouali, I. Ghorbel, M. Khanfir, H. Houman, and A. Hamzaoui, “Expression of Th-17 and RORγt mRNA in Beh?et's disease,” Medical Science Monitor, vol. 17, no. 4, pp. CR227–CR234, 2011.
[40]  J. Shimizu, K. Takai, N. Fujiwara et al., “Excessive CD4+ T cells co-expressing interleukin-17 and interferon-γ in patients with Beh?et's disease,” Clinical and Experimental Immunology, vol. 168, no. 1, pp. 68–74, 2012.
[41]  G. Geri, B. Terrier, M. Rosenzwajg et al., “Critical role of IL-21 in modulating TH17 and regulatory T cells in Behet disease,” Journal of Allergy and Clinical Immunology, vol. 128, no. 3, pp. 655–664, 2011.
[42]  Y. Nanke, S. Kotake, M. Goto, H. Ujihara, M. Matsubara, and N. Kamatani, “Decreased percentages of regulatory T cells in peripheral blood of patients with Behcet's disease before ocular attack: a possible predictive marker of ocular attack,” Modern Rheumatology, vol. 18, no. 4, pp. 354–358, 2008.
[43]  W. Chi, X. Zhu, P. Yang et al., “Upregulated IL-23 and IL-17 in Beh?et patients with active uveitis,” Investigative Ophthalmology and Visual Science, vol. 49, no. 7, pp. 3058–3064, 2008.
[44]  J. Shimizu, T. Izumi, N. Arimitsu, et al., “Skewed TGFβ/Smad signaling pathway of T cells in patients with Behcet’s disease,” Clinical and Experimental Rheumatology, vol. 30, supplement 72, pp. S35–S39, 2012.
[45]  A. Osterloh and M. Breloer, “Heat shock proteins: linking danger and pathogen recognition,” Medical Microbiology and Immunology, vol. 197, no. 1, pp. 1–8, 2008.
[46]  S. Akira, S. Uematsu, and O. Takeuchi, “Pathogen recognition and innate immunity,” Cell, vol. 124, no. 4, pp. 783–801, 2006.
[47]  C. A. Janeway Jr., U. Dianzani, P. Portoles et al., “Cross-linking and conformational change in T-cell receptors: role in activation and in repertoire selection,” Cold Spring Harbor Symposia on Quantitative Biology, vol. 54, no. 2, pp. 657–666, 1989.
[48]  T. Kawai and S. Akira, “Toll-like receptors and their crosstalk with other innate receptors in infection and immunity,” Immunity, vol. 34, no. 5, pp. 637–650, 2011.
[49]  A. L. Blasius and B. Beutler, “Intracellular toll-like receptors,” Immunity, vol. 32, no. 3, pp. 305–315, 2010.
[50]  A. M. Piccinini and K. S. Midwood, “DAMPening inflammation by modulating TLR signalling,” Mediators of Inflammation, vol. 2010, Article ID 672395, 21 pages, 2010.
[51]  M. Schnare, G. M. Barton, A. C. Holt, K. Takeda, S. Akira, and R. Medzhitov, “Toll-like receptors control activation of adaptive immune responses,” Nature Immunology, vol. 2, no. 10, pp. 947–950, 2001.
[52]  A. F. Heiseke, A. C. Faul, H. Lehr et al., “CCL17 promotes intestinal inflammation in mice and counteracts regulatory T cellmediated protection from colitis,” Gastroenterology, vol. 142, no. 2, pp. 335–345, 2012.
[53]  M. E. Remoli, V. Gafa, E. Giacomini, M. Severa, R. Lande, and E. M. Coccia, “IFN-β modulates the response to TLR stimulation in human DC: involvement of IFN regulatory factor-1 (IRF-1) in IL-27 gene expression,” European Journal of Immunology, vol. 37, no. 12, pp. 3499–3508, 2007.
[54]  J. Pirhonen, J. Sirén, I. Julkunen, and S. Matikainen, “IFN-α regulates toll-like receptor-mediated IL-27 gene expression in human macrophages,” Journal of Leukocyte Biology, vol. 82, no. 5, pp. 1185–1192, 2007.
[55]  N. Schuetze, S. Schoeneberger, U. Mueller, M. A. Freudenberg, G. Alber, and R. K. Straubinger, “IL-12 family members: differential kinetics of their TLR4-mediated induction by Salmonella enteritidis and the impact of IL-10 in bone marrow-derived macrophages,” International Immunology, vol. 17, no. 5, pp. 649–659, 2005.
[56]  N. M. Chapman, M. Y. Bilal, N. Cruz-Orcutt, et al., “Distinct signaling pathways regulate TLR2 co-stimulatory function in human T cells,” Cellular Signalling, vol. 25, no. 3, pp. 639–650, 2013.
[57]  H. Liu, M. Komai-Koma, D. Xu, and F. Y. Liew, “Toll-like receptor 2 signaling modulates the functions of CD4+CD25+ regulatory T cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 18, pp. 7048–7053, 2006.
[58]  T. Imanishi, H. Hara, S. Suzuki, N. Suzuki, S. Akira, and T. Saito, “Cutting edge: TLR2 directly triggers Th1 effector functions,” The Journal of Immunology, vol. 178, no. 11, pp. 6715–6719, 2007.
[59]  A. Biswas, P. Banerjee, and T. Biswas, “Porin of Shigella dysenteriae directly promotes toll-like receptor 2-mediated CD4+ T cell survival and effector function,” Molecular Immunology, vol. 46, no. 15, pp. 3076–3085, 2009.
[60]  J. M. Reynolds, B. P. Pappu, J. Peng et al., “Toll-like receptor 2 signaling in CD4+ T lymphocytes promotes T helper 17 responses and regulates the pathogenesis of autoimmune disease,” Immunity, vol. 32, no. 5, pp. 692–702, 2010.
[61]  M. H. Nyirenda, L. Sanvito, P. J. Darlington et al., “TLR2 stimulation drives human naive and effector regulatory T cells into a Th17-like phenotype with reduced suppressive function,” The Journal of Immunology, vol. 187, no. 5, pp. 2278–2290, 2011.
[62]  X. Chen, M. Zhang, X. Zhu et al., “Engagement of toll-like receptor 2 on CD4+ T cells facilitates local immune responses in patients with tuberculous pleurisy,” Journal of Infectious Diseases, vol. 200, no. 3, pp. 399–408, 2009.
[63]  S. Babu, C. P. Blauvelt, V. Kumaraswami, and T. B. Nutman, “Cutting edge: diminished T cell TLR expression and function modulates the immune response in human filarial infection,” The Journal of Immunology, vol. 176, no. 7, pp. 3885–3889, 2006.
[64]  J. M. González-Navajas, S. Fine, J. Law et al., “TLR4 signaling in effector CD4+ T cells regulates TCR activation and experimental colitis in mice,” The Journal of Clinical Investigation, vol. 120, no. 2, pp. 570–581, 2010.
[65]  J. M. Reynolds, G. J. Martinez, Y. Chung, and C. Dong, “Toll-like receptor 4 signaling in T cells promotes autoimmune inflammation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 32, pp. 13064–13069, 2012.
[66]  J. C. Young, V. R. Agashe, K. Siegers, and F. U. Hartl, “Pathways of chaperone-mediated protein folding in the cytosol,” Nature Reviews Molecular Cell Biology, vol. 5, no. 10, pp. 781–791, 2004.
[67]  A. K. Dudani and R. S. Gupta, “Immunological characterization of a human homolog of the 65-kilodalton mycobacterial antigen,” Infection and Immunity, vol. 57, no. 9, pp. 2786–2793, 1989.
[68]  G. Y. Chen and G. Nu?ez, “Sterile inflammation: sensing and reacting to damage,” Nature Reviews Immunology, vol. 10, no. 12, pp. 826–837, 2010.
[69]  Q.-Q. Huang, R. Sobkoviak, A. R. Jockheck-Clark et al., “Heat shock protein 96 is elevated in rheumatoid arthritis and activates macrophages primarily via TLR2 signaling,” The Journal of Immunology, vol. 182, no. 8, pp. 4965–4973, 2009.
[70]  C. J. P. Boog, E. R. de Graeff-Meeder, M. A. Lucassen et al., “Two monoclonal antibodies generated against human hsp60 show reactivity with synovial membranes of patients with juvenile chronic arthritis,” The Journal of Experimental Medicine, vol. 175, no. 6, pp. 1805–1810, 1992.
[71]  E. F. Elst, M. Klein, W. De Jager et al., “Hsp60 in inflamed muscle tissue is the target of regulatory autoreactive T cells in patients with juvenile dermatomyositis,” Arthritis and Rheumatism, vol. 58, no. 2, pp. 547–555, 2008.
[72]  G. L. Puga Yung, M. Fidler, E. Albani et al., “Heat shock protein-derived T-cell epitopes contribute to autoimmune inflammation in pediatric Crohn's disease,” PLoS ONE, vol. 4, no. 11, Article ID e7714, 2009.
[73]  D. O. Sobel and K. Creswell, “Characterization of anti-islet cytotoxic human T-cell clones from patients with type 1 diabetes mellitus,” Autoimmunity, vol. 39, no. 4, pp. 323–332, 2006.
[74]  H. Jiang, S. M. Canfield, M. P. Gallagher et al., “HLA-E-restricted regulatory CD8+ T cells are involved in development and control of human autoimmune type 1 diabetes,” The Journal of Clinical Investigation, vol. 120, no. 10, pp. 3641–3650, 2010.
[75]  B. J. Prakken, R. Samodal, T. D. Le et al., “Epitope-specific immunotherapy induces immune deviation of proinflammatory T cells in rheumatoid arthritis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 12, pp. 4228–4233, 2004.
[76]  C.-T. C. Wu, L.-S. Ou, K.-W. Yeh, W.-I. Lee, and J.-L. Huang, “Serum heat shock protein 60 can predict remission of flare-up in juvenile idiopathic arthritis,” Clinical Rheumatology, vol. 30, no. 7, pp. 959–965, 2011.
[77]  E. Zonneveld-Huijssoon, S. Albani, B. J. Prakken, and F. van Wijk, “Heat shock protein bystander antigens for peptide immunotherapy in autoimmune disease,” Clinical and Experimental Immunology, vol. 171, no. 1, pp. 20–29, 2013.
[78]  E. C. Koffeman, M. Genovese, D. Amox et al., “Epitope-specific immunotherapy of rheumatoid arthritis: clinical responsiveness occurs with immune deviation and relies on the expression of a cluster of molecules associated with T cell tolerance in a double-blind, placebo-controlled, pilot phase II trial,” Arthritis and Rheumatism, vol. 60, no. 11, pp. 3207–3216, 2009.
[79]  J. E. Do, S. Y. Kwon, S. Park, and E.-S. Lee, “Effects of vitamin D on expression of toll-like receptors of monocytes from patients with Beh?et's disease,” Rheumatology, vol. 47, no. 6, pp. 840–848, 2008.
[80]  Y. Kirino, M. Takeno, R. Watanabe et al., “Association of reduced heme oxygenase-1 with excessive toll-like receptor 4 expression in peripheral blood mononuclear cells in Beh?et's disease,” Arthritis Research and Therapy, vol. 10, no. 1, article R16, 2008.
[81]  K. Hamzaoui, H. Abid, A. Berraies, J. Ammar, and A. Hamzaoui, “NOD2 is highly expressed in Beh?et disease with pulmonary manifestations,” Journal of Inflammation, vol. 9, no. 1, article 3, 2012.
[82]  N. Seoudi, L. A. Bergmeier, E. Hagi-Pavli, D. Bibby, M. A. Curtis, and F. Fortune, “The role of TLR2 and 4 in Behcet's disease pathogenesis,” Innate Immunity, 2013.
[83]  O. Durrani, K. Banahan, F. J. Sheedy et al., “TIRAP Ser180Leu polymorphism is associated with Beh?et's disease,” Rheumatology, vol. 50, no. 10, pp. 1760–1765, 2011.
[84]  S. Yavuz, Y. Elbir, A. Tulunay, E. Eksioglu-Demiralp, and H. Direskeneli, “Differential expression of toll-like receptor 6 on granulocytes and monocytes implicates the role of microorganisms in Behcet's disease etiopathogenesis,” Rheumatology International, vol. 28, no. 5, pp. 401–406, 2008.
[85]  T. Ergun, U. Ince, E. Ek?io?lu-Demiralp, et al., “HSP 60 expression in mucocutaneous lesions of Beh?et's disease,” Journal of the American Academy of Dermatology, vol. 45, no. 6, pp. 904–909, 2001.
[86]  E. Deniz, U. Guc, N. Buyukbabani, and A. Gul, “HSP 60 expression in recurrent oral ulcerations of Behet's disease,” Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology and Endodontology, vol. 110, no. 2, pp. 196–200, 2010.
[87]  T. E. Bramanti, N. P. Dekker, F. Lozada-Nur, J. J. Sauk, and J. A. Regezi, “Heat shock (stress) proteins and γδ T lymphocytes in oral lichen planus,” Oral Surgery, Oral Medicine, Oral Pathology, Oral Radiology and, vol. 80, no. 6, pp. 698–704, 1995.
[88]  K. Pervin, A. Childerstone, T. Shinnick et al., “T cell epitope expression of mycobacterial and homologous human 65- kilodalton heat shock protein peptides in short term cell lines from patients with Behcet's disease,” The Journal of Immunology, vol. 151, no. 4, pp. 2273–2282, 1993.
[89]  H. Direskeneli, E. Ek?io?lu-Demiralp, ?. Yavuz et al., “T cell responses to 60/65 kDa heat shock protein derived peptides in Turkish patients with Behcet's disease,” Journal of Rheumatology, vol. 27, no. 3, pp. 708–713, 2000.
[90]  S. Kaneko, N. Suzuki, N. Yamashita et al., “Characterization of T cells specific for an epitope of human 60-kD heat shock protein (hsp) in patients with Behcet's disease (BD) in Japan,” Clinical and Experimental Immunology, vol. 108, no. 2, pp. 204–212, 1997.
[91]  J. Shimizu, T. Izumi, and N. Suzuki, “Aberrant activation of heat shock protein 60/65 reactive T cells in patients with Behcet's disease,” Autoimmune Diseases, vol. 2012, Article ID 105205, 7 pages, 2012.
[92]  W. W. Eaton, N. R. Rose, A. Kalaydjian, M. G. Pedersen, and P. B. Mortensen, “Epidemiology of autoimmune diseases in Denmark,” Journal of Autoimmunity, vol. 29, no. 1, pp. 1–9, 2007.
[93]  R. L. van Wanrooij, A. Zwiers, G. Kraal, and G. Bouma, “Genetic variations in interleukin-12 related genes in immune-mediated diseases,” Journal of Autoimmunity, vol. 39, no. 4, pp. 359–368, 2012.
[94]  R. H. Duerr, K. D. Taylor, S. R. Brant et al., “A genome-wide association study identifies IL23R as an inflammatory bowel disease gene,” Science, vol. 314, no. 5804, pp. 1461–1463, 2006.
[95]  R. P. Nair, K. C. Duffin, C. Helms et al., “Genome-wide scan reveals association of psoriasis with IL-23 and NF-κB pathways,” Nature Genetics, vol. 41, no. 2, pp. 199–204, 2009.
[96]  J. D. Reveille, A.-M. Sims, P. Danoy et al., “Genome-wide association study of ankylosing spondylitis identifies non-MHC susceptibility loci,” Nature Genetics, vol. 42, no. 2, pp. 123–127, 2010.
[97]  M. Chen-Xu, R. Topless, C. McKinney et al., “Replication of association of the interleukin 23 receptor rs1343151 variant with rheumatoid arthritis in Caucasian sample sets,” Annals of the Rheumatic Diseases, vol. 71, no. 1, pp. 155–157, 2012.
[98]  G. F. Mells, J. A. B. Floyd, K. I. Morley et al., “Genome-wide association study identifies 12 new susceptibility loci for primary biliary cirrhosis,” Nature Genetics, vol. 43, no. 4, pp. 332–333, 2011.
[99]  T. Guo, S. Yang, N. Liu, S. Wang, B. Cui, and G. Ning, “Association study of interleukin-12A gene polymorphisms with Graves' disease in two Chinese populations,” Clinical Endocrinology, vol. 74, no. 1, pp. 125–129, 2011.
[100]  C. Nú?ez, B. Dema, M. C. Cénit, et al., “IL23R: a susceptibility locus for celiac disease and multiple sclerosis?” Genes and Immunity, vol. 9, no. 4, pp. 289–293, 2008.
[101]  S. Sawcer, G. Hellenthal, M. Pirinen, et al., “Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis,” Nature, vol. 476, no. 7359, pp. 214–219, 2011.
[102]  Y. Kirino, G. Bertsias, Y. Ishigatsubo, et al., “Genome-wide association analysis identifies new susceptibility loci for Beh?et's disease and epistasis between HLA-B*51 and ERAP1,” Nature Genetics, vol. 45, no. 2, pp. 202–207, 2013.
[103]  E. F. Remmers, F. Cosan, Y. Kirino et al., “Genome-wide association study identifies variants in the MHC class I, IL10, and IL23R-IL12RB2 regions associated with Beh?et's disease,” Nature Genetics, vol. 42, no. 8, pp. 698–702, 2010.
[104]  Y. Fei, R. Webb, B. L. Cobb, H. Direskeneli, G. Saruhan-Direskeneli, and A. H. Sawalha, “Identification of novel genetic susceptibility loci for Beh?et's disease using a genome-wide association study,” Arthritis Research and Therapy, vol. 11, no. 3, article R66, 2009.
[105]  N. Mizuki, A. Meguro, M. Ota et al., “Genome-wide association studies identify IL23R-IL12RB2 and IL10 as Beh?et's disease susceptibility loci,” Nature Genetics, vol. 42, no. 8, pp. 703–706, 2010.
[106]  A. Meguro, H. Inoko, M. Ota et al., “Genetics of Beh?et disease inside and outside the MHC,” Annals of the Rheumatic Diseases, vol. 69, no. 4, pp. 747–754, 2010.
[107]  S. Hou, Z. Yang, L. Du, et al., “Identification of a susceptibility locus in STAT4 for Beh?et's disease in Han Chinese in a genome-wide association study,” Arthritis and Rheumatism, vol. 64, no. 12, pp. 4104–4113, 2012.
[108]  J. M. Xavier, F. Shahram, F. Davatchi, et al., “Association study of IL10 and IL23R-IL12RB2 in Iranian patients with Beh?et's disease,” Arthritis and Rheumatism, vol. 64, no. 8, pp. 2761–2772, 2012.
[109]  E. S. Kim, S. W. Kim, C. M. Moon et al., “Interactions between IL17A, IL23R, and STAT4 polymorphisms confer susceptibility to intestinal Behcet's disease in Korean population,” Life Sciences, vol. 90, no. 19-20, pp. 740–746, 2012.
[110]  M. de Menthon, M. P. Lavalley, C. Maldini, L. Guillevin, and A. Mahr, “HLA-B51/B5 and the risk of Beh?et's disease: a systematic review and meta-analysis of case-control genetic association studies,” Arthritis Care and Research, vol. 61, no. 10, pp. 1287–1296, 2009.
[111]  S. Pestka, C. D. Krause, D. Sarkar, M. R. Walter, Y. Shi, and P. B. Fisher, “Interleukin-10 and related cytokines and receptors,” Annual Review of Immunology, vol. 22, pp. 929–979, 2004.
[112]  M. Fager?s B?ttcher, M. Hmani-Aifa, A. Lindstr?m et al., “A TLR4 polymorphism is associated with asthma and reduced lipopolysaccharide-induced interleukin-12(p70) responses in Swedish children,” Journal of Allergy and Clinical Immunology, vol. 114, no. 3, pp. 561–567, 2004.
[113]  C. Salpietro, L. Rigoli, M. Miraglia del Giudice et al., “TLR2 and TLR4 gene polymorphisms and atopic dermatitis in Italian children: a multicenter study,” International Journal of Immunopathology and Pharmacology, vol. 24, no. 4, pp. 33–40, 2011.
[114]  P. Ahmad-Nejad, S. Mrabet-Dahbi, K. Breuer et al., “The toll-like receptor 2 R753Q polymorphism defines a subgroup of patients with atopic dermatitis having severe phenotype,” Journal of Allergy and Clinical Immunology, vol. 113, no. 3, pp. 565–567, 2004.
[115]  M. Kerkhof, D. S. Postma, B. Brunekreef et al., “Toll-like receptor 2 and 4 genes influence susceptibility to adverse effects of traffic-related air pollution on childhood asthma,” Thorax, vol. 65, no. 8, pp. 690–697, 2010.
[116]  K. G. E. Miedema, W. J. E. Tissing, E. M. te Poele et al., “Polymorphisms in the TLR6 gene associated with the inverse association between childhood acute lymphoblastic leukemia and atopic disease,” Leukemia, vol. 26, no. 6, pp. 1203–1210, 2012.
[117]  T. Sada, M. Ota, Y. Katsuyama et al., “Association analysis of toll-like receptor 7 gene polymorphisms and Beh?et's disease in Japanese patients,” Human Immunology, vol. 72, no. 3, pp. 269–272, 2011.
[118]  I. Ben Dhifallah, J. Lachheb, H. Houman, and K. Hamzaoui, “Toll-like-receptor gene polymorphisms in a Tunisian population with Beh?et's disease,” Clinical and Experimental Rheumatology, vol. 27, no. 2, supplement 53, pp. S58–S62, 2009.
[119]  L. Boiardi, F. Atzeni, B. Casali et al., “Toll-like receptor 4 (TLR4) gene polymorphisms in Italian patients with Beh?et's disease,” Clinical and Experimental Rheumatology, vol. 27, no. 2, supplement 53, pp. S43–S47, 2009.
[120]  F. Co?an, B. Oku, A. ?akiris et al., “No association of the TLR2 gene Arg753Gln polymorphism with rheumatic heart disease and Beh?et's disease,” Clinical Rheumatology, vol. 28, no. 12, pp. 1385–1388, 2009.
[121]  R. Tomiyama, A. Meguro, M. Ota et al., “Investigation of the association between toll-like receptor 2 gene polymorphisms and Beh?et's disease in Japanese patients,” Human Immunology, vol. 70, no. 1, pp. 41–44, 2009.
[122]  A. Meguro, M. Ota, Y. Katsuyama et al., “Association of the toll-like receptor 4 gene polymorphisms with Beh?et's disease,” Annals of the Rheumatic Diseases, vol. 67, no. 5, pp. 725–727, 2008.
[123]  A. Ito, M. Ota, Y. Katsuyama, H. Inoko, S. Ohno, and N. Mizuki, “Lack of association of toll-like receptor 9 gene polymorphism with Beh?et's disease in Japanese patients,” Tissue Antigens, vol. 70, no. 5, pp. 423–426, 2007.
[124]  A. Bacanli, N. Sallakci, U. Yavuzer, E. Alpsoy, and O. Yegin, “Toll-like receptor 2 Arg753Gln gene polymorphism in Turkish patients with Beh?et's disease,” Clinical and Experimental Dermatology, vol. 31, no. 5, pp. 699–701, 2006.
[125]  Y. Kirino, Q. Zhou, Y. Ishigatsubo, et al., “Targeted resequencing implicates the familial Mediterranean fever gene MEFV and the toll-like receptor 4 gene TLR4 in Beh?et disease,” Proceedings of the National Academy of Sciences of the United States of America, vol. 110, no. 20, pp. 8134–8139, 2013.
[126]  P. P. Sfikakis, N. Markomichelakis, E. Alpsoy et al., “Anti-TNF therapy in the management of Beh?et's disease—review and basis for recommendations,” Rheumatology, vol. 46, no. 5, pp. 736–741, 2007.
[127]  H. Ideguchi, A. Suda, M. Takeno, et al., “Gastrointestinal manifestations of Beh?et's disease in Japan: a study of 43 patients,” Rheumatology International, 2013.
[128]  J. Chen, J. Ren, G. Gu, et al., “Crohn's disease and polymorphism of heat shock protein gene HSP70-2 in the Chinese population,” Journal of Gastroenterology and Hepatology, vol. 28, no. 5, pp. 814–818, 2013.
[129]  S. M. Davis, E. A. S. Clark, L. T. Nelson, and R. M. Silver, “The association of innate immune response gene polymorphisms and puerperal group A streptococcal sepsis,” American Journal of Obstetrics and Gynecology, vol. 202, no. 3, pp. 308.e1–308.e8, 2010.
[130]  Y. Zhao, L. Tao, D. Jiang, et al., “The -144C/A polymorphism in the promoter of HSP90beta is associated with multiple organ dysfunction scores,” PLoS ONE, vol. 8, no. 3, Article ID e58646, 2013.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133