| 研究生: |
邢中熹 Hsing, Chung-Hsi |
|---|---|
| 論文名稱: |
介白素十九在全身性發炎性疾病的研究 Study of Interleukin-19 in Systemic Inflammatory Diseases |
| 指導教授: |
張明熙
Chang, Ming-Shi |
| 學位類別: |
博士 Doctor |
| 系所名稱: |
醫學院 - 藥物生物科技研究所 Institute of Biopharmaceutical Sciences |
| 論文出版年: | 2008 |
| 畢業學年度: | 96 |
| 語文別: | 英文 |
| 論文頁數: | 122 |
| 中文關鍵詞: | 介白素十九 、細胞素 、發炎反應 、體外循環 、敗血症 、慢性腎衰竭 |
| 外文關鍵詞: | cytokines, interleukin-19, systemic inflammation, uremia, sepsis, cardiopulmonary bypass |
| 相關次數: | 點閱:118 下載:1 |
| 分享至: |
| 查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報 |
細胞素在發炎性疾病佔有重要的角色,研究細胞素在發炎性疾病的作用對疾病的控制有很大的幫助,新型細胞素的發現也可增加對發炎性疾病機制的了解,甚至成為治療標的,是目前醫藥界積極進行的研究。介白素(IL)-19是最近新發現的細胞素,屬於IL-10家族之一,其生物功能所知仍有限。目前已知IL-19能引發免疫細胞產生IL-6、IL-8、TNF-α、ROS及單核球凋亡。IL-19的另一特徵是會影響Th1/Th2的分布,因此IL-19被認為與免疫調節相關。IL-19在臨床疾病上的探討很少,僅知與慢性局部發炎性疾病如氣喘與乾癬有關,而IL-19在全身性發炎性疾病的影響尚無所知。我們初步觀察發現IL-19在心臟手術體外循環後所引起的急性全身性發炎反應中,會明顯上升,但作用不明,於是我們首先假設:在體外循環後IL-19的上升與其他發炎性細胞素的變化相關;而在敗血症等急性全身性發炎反應中,IL-19亦可能上升且與組織傷害的機制有關。另在長期血液透析病患常合併慢性全身性發炎反應,且會發生Th1/Th2調節異常與免疫抑制作用,於是我們再假設:接受長期血液透析的腎衰竭病患其產生之慢性全身發炎會引起IL-19的變化且與Th1/Th2調節異常相關。研究結果發現,體外循環後血清中IL-19的上升與IL-6、IL-10、及TNF-α的變化相似,單核球活化是IL-19的重要來源之一,體外細胞實驗也發現IL-6及TNF-α可促使單核球製造IL-19。我們也證實敗血症病患血液中IL-19濃度較正常人明顯增加。以免疫組織化學染色法分析IL-19在正常人身體內的組織及細胞存在情形,發現IL-19主要出現在皮膚、肺臟、肝臟、腎臟等器官的上皮細胞,巨噬細胞,及內皮細胞。因我們也發現這些細胞具有IL-19接受器,進一步推測IL-19對組織細胞有重要作用。在體外細胞實驗中,IL-19可引起肺上皮細胞凋亡及肝上皮細胞產生ROS等發炎物質;IL-19也促使肺與肝上皮細胞產生趨化因子IL-1β、IL-6、IL-8、CCL5、及CXCL9;IL-19並增加嗜中性球的移動,且減少其凋亡,此現象在敗血症病患更為明顯。在動物實驗中,內毒素休克小鼠的主要器官包括肺臟、肝臟、腎臟、及心臟組織中IL-19及其接受器 mRNA明顯增加;以electroporation方法將可溶性IL-19接受器DNA質體轉移入小鼠體內做為拮抗IL-19之用,可發現小鼠在拮抗IL-19處理後可減少內毒素休克造成的肺臟及肝臟嗜中性球浸潤與血清中ALT及AST的上升,表示IL-19在急性全身性發炎反應中是傷害性的角色,IL-19會加重內毒素休克所引起組織的傷害。在洗腎患者,血清中CRP上升,IL-19含量也有偏高的現象且與發炎性細胞素(IL-6與TNF-α)及Th2細胞素(IL-4、IL-5、IL-6、IL-10與IL-13)間有很高的統計相關性;正常人的單核球在體外給予洗腎病患的血清刺激,可增加IL-19產生;IL-19也可刺激T細胞產生更多的Th2細胞素。我們的研究顯示,洗腎病患之全身性慢性發炎反應可引起IL-19上升,其可能參予腎衰竭病患的Th1/Th2調節異常作用。我們的研究提出了IL-19在全身性發炎性疾病中可能的作用,相信本研究對於進一步釐清IL-19的功能及其與臨床病症的關連性有很重要的貢獻。
Cytokines are important mediators in systemic inflammatory diseases. It is valuable while to discovery novel cytokines and to identify their functions and effects on the inflammatory diseases. IL-19 is a newly discovered proinflammatory cytokine belonging to IL-10 family. At the present time, knowledge about IL-19 on inflammatory diseases is still limited. Previous reports showed that IL-19 induced immune cells to produce IL-6, IL-8, TNF-α and ROS and undergo apoptosis. IL-19 was also involved in Th1/Th2 polarization which is related to immune regulation. Clinically, IL-19 was reported to be involved in the mechanism of chronic local inflammatory diseases such as psoriasis and asthma. In our preliminary exploration, we found that IL-19 was obviously increased in systemic inflammatory response syndrome (SIRS) after cardiac surgery with cardiopulmonary bypass (CPB). We, therefore, were interesting about the novel effects of IL-19 on acute and chronic systemic inflammatory diseases. We hypothesize that, first, the levels of IL-19 after CPB is associated with the changes of other proinflammatory cytokines which were associated with organ dysfunction after cardiac surgery. Second, IL-19 is induced in sepsis/SIRS and related to the pathogenesis of tissue injury. Third, according to the status of chronic systemic inflammation in uremic patients with hemodialysis, we hypothesize that IL-19 is altered and associated with Th1/Th2 dysregulation in uremia which may affect immunosupression in these patients. Our results showed that IL-19 was induced and associated with the production of IL-6, IL-10, and TNF-α after CPB. IL-19 transcripts in monocytes from patients were increased after CPB, which indicated that monocyte is one of the important sources of IL-19. In vitro experiments showed that IL-6 and TNF-α upregulated IL-19 protein expression in monocytes. We then survey the distribution of IL-19 in tissues and cells using immunohistochemistry study and found that skin, lung, liver and kidney were major organs expressing IL-19. Specific epithelial cells, macrophages, and endothelial cells were the major cell types stained positive for IL-19. In addition, these cells also expressed IL-19 receptors indicated that IL-19 plays an important role in these cells. In patients of severe sepsis, serum levels of IL-19 were higher in patients than in healthy volunteers. IL-19 induced apoptosis in lung epithelial cells and ROS production in liver cells in vitro. IL-19 also promoted neutrophil chemotaxis, reduced neutrophil apoptosis, and induced the production of proinflammatory cytokines and chemokines (IL-1β, IL-6, IL-8, CCL5, and CXCL9) in lung epithelial cells. In lipopolysaccharide (LPS)-challenged mice, transcripts of IL-19 and its receptors were upregulated in heart, lung, liver, and kidney tissue. Neutrophil infiltration in lung and liver tissue, and serum levels of ALT and AST, were lower in mice electroporated with IL-19 soluble receptor plasmid DNA before LPS treatment compared to control mice. These findings suggested that IL-19 plays as a detrimental role in SIRS. In uremic patients, a status of chronic systemic inflammatory disease, serum levels of IL-19 showed wide distributed amount individuals. However, IL-19 levels in the uremic patients but not in healthy controls correlated positively with both the proinflammatory cytokines (IL-6 and TNF-α) and the Th2 cytokines (IL-4, IL-5, IL-6, IL-10, and IL-13). Cultured monocytes from uremic patients with high IL-19 serum levels produced more IL-19 in vitro. In addition, uremic serum upregulated the production of IL-19 in resting monocytes. Compared with T cells from healthy controls, uremic T cells expressed more endogenous Th2 cytokine transcripts and further responded to IL-19 stimulation in Th2 cytokine production in vitro.
In conclusion, serum IL-19 was induced and correlated with IL-6 and TNF-α in acute and chronic systemic inflammation. The upregulated IL-19 in endotoxic shock/sepsis was involved in lung and liver tissue injury. IL-19 also correlated with Th2 immune responses and induced Th1/Th2 cytokine dysregulation in uremia. Our findings provide a new concept that IL-19, the recent discovered cytokine, is an important molecule and might to be a potential therapeutic target in systemic inflammatory diseases.
1. Pestka, S., C.D. Krause, D. Sarkar, M.R. Walter, Y. Shi, and P.B. Fisher. Interleukin-10 and related cytokines and receptors. Annu Rev Immunol 2004; 22: 929-79.
2. Fickenscher, H., S. Hor, H. Kupers, A. Knappe, S. Wittmann, and H. Sticht. The interleukin-10 family of cytokines. Trends Immunol 2002; 23: 89-96.
3. Langer, J.A., E.C. Cutrone, and S. Kotenko. The Class II cytokine receptor (CRF2) family: overview and patterns of receptor-ligand interactions. Cytokine Growth Factor Rev 2004; 15: 33-48.
4. Conti, P., D. Kempuraj, S. Frydas, et al. IL-10 subfamily members: IL-19, IL-20, IL-22, IL-24 and IL-26. Immunol Lett 2003; 88: 171-4.
5. Zdanov, A. Structural features of the interleukin-10 family of cytokines. Curr Pharm Des 2004; 10: 3873-84.
6. Liao, Y.C., W.G. Liang, F.W. Chen, J.H. Hsu, J.J. Yang, and M.S. Chang. IL-19 induces production of IL-6 and TNF-alpha and results in cell apoptosis through TNF-alpha. J Immunol 2002; 169: 4288-97.
7. Liao, S.C., Y.C. Cheng, Y.C. Wang, et al. IL-19 induced Th2 cytokines and was up-regulated in asthma patients. J Immunol 2004; 173: 6712-8.
8. Rich, B.E. IL-20: a new target for the treatment of inflammatory skin disease. Expert Opin Ther Targets 2003; 7: 165-74.
9. Dumoutier, L., C. Leemans, D. Lejeune, S.V. Kotenko, and J.C. Renauld. Cutting edge: STAT activation by IL-19, IL-20 and mda-7 through IL-20 receptor complexes of two types. J Immunol 2001; 167: 3545-9.
10. Gallagher, G., H. Dickensheets, J. Eskdale, et al. Cloning, expression and initial characterization of interleukin-19 (IL-19), a novel homologue of human interleukin-10 (IL-10). Genes Immun 2000; 1: 442-50.
11. Chang, C., E. Magracheva, S. Kozlov, et al. Crystal structure of interleukin-19 defines a new subfamily of helical cytokines. J Biol Chem 2003; 278: 3308-13.
12. Parrish-Novak, J., W. Xu, T. Brender, et al. Interleukins 19, 20, and 24 signal through two distinct receptor complexes. Differences in receptor-ligand interactions mediate unique biological functions. J Biol Chem 2002; 277: 47517-23.
13. Wolk, K., S. Kunz, K. Asadullah, and R. Sabat. Cutting edge: immune cells as sources and targets of the IL-10 family members? J Immunol 2002; 168: 5397-402.
14. Jordan, W.J., J. Eskdale, M. Boniotto, et al. Human IL-19 regulates immunity through auto-induction of IL-19 and production of IL-10. Eur J Immunol 2005; 35: 1576-82.
15. Li, H.H., Y.C. Lin, P.J. Chen, et al. Interleukin-19 upregulates keratinocyte growth factor and is associated with psoriasis. Br J Dermatol 2005; 153: 591-5.
16. Blumberg, H., D. Conklin, W.F. Xu, et al. Interleukin 20: discovery, receptor identification, and role in epidermal function. Cell 2001; 104: 9-19.
17. Sauane, M., R.V. Gopalkrishnan, I. Lebedeva, et al. Mda-7/IL-24 induces apoptosis of diverse cancer cell lines through JAK/STAT-independent pathways. J Cell Physiol 2003; 196: 334-45.
18. Gallagher, G., J. Eskdale, W. Jordan, et al. Human interleukin-19 and its receptor: a potential role in the induction of Th2 responses. Int Immunopharmacol 2004; 4: 615-26.
19. Lederer, J.A., M.L. Rodrick, and J.A. Mannick. The effects of injury on the adaptive immune response. Shock 1999; 11: 153-9.
20. O'Sullivan, S.T., J.A. Lederer, A.F. Horgan, D.H. Chin, J.A. Mannick, and M.L. Rodrick. Major injury leads to predominance of the T helper-2 lymphocyte phenotype and diminished interleukin-12 production associated with decreased resistance to infection. Ann Surg 1995; 222: 482-90; discussion 490-2.
21. Ikeuchi, H., T. Kuroiwa, N. Hiramatsu, et al. Expression of interleukin-22 in rheumatoid arthritis: potential role as a proinflammatory cytokine. Arthritis Rheum 2005; 52: 1037-46.
22. Sigmundsdottir, H., J.E. Gudjonsson, I. Jonsdottir, B.R. Ludviksson, and H. Valdimarsson. The frequency of CLA+ CD8+ T cells in the blood of psoriasis patients correlates closely with the severity of their disease. Clin Exp Immunol 2001; 126: 365-9.
23. Finch, P.W., F. Murphy, I. Cardinale, and J.G. Krueger. Altered expression of keratinocyte growth factor and its receptor in psoriasis. Am J Pathol 1997; 151: 1619-28.
24. Strange, P., K.D. Cooper, E.R. Hansen, et al. T-lymphocyte clones initiated from lesional psoriatic skin release growth factors that induce keratinocyte proliferation. J Invest Dermatol 1993; 101: 695-700.
25. Romer, J., E. Hasselager, P.L. Norby, T. Steiniche, J. Thorn Clausen, and K. Kragballe. Epidermal overexpression of interleukin-19 and -20 mRNA in psoriatic skin disappears after short-term treatment with cyclosporine a or calcipotriol. J Invest Dermatol 2003; 121: 1306-11.
26. Swain, S.L., A.D. Weinberg, M. English, and G. Huston. IL-4 directs the development of Th2-like helper effectors. J Immunol 1990; 145: 3796-806.
27. Reents, W., J. Babin-Ebell, M.R. Misoph, A. Schwarzkopf, and O. Elert. Influence of different autotransfusion devices on the quality of salvaged blood. Ann Thorac Surg 1999; 68: 58-62.
28. Clutterbuck, E.J., E.M. Hirst, and C.J. Sanderson. Human interleukin-5 (IL-5) regulates the production of eosinophils in human bone marrow cultures: comparison and interaction with IL-1, IL-3, IL-6, and GMCSF. Blood 1989; 73: 1504-12.
29. Grunig, G., M. Warnock, A.E. Wakil, et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science 1998; 282: 2261-3.
30. Bochud, P.Y. and T. Calandra. Pathogenesis of sepsis: new concepts and implications for future treatment. Bmj 2003; 326: 262-6.
31. Sessler, C.N. and W. Shepherd. New concepts in sepsis. Curr Opin Crit Care 2002; 8: 465-72.
32. Kilger, E., F. Weis, J. Briegel, et al. Stress doses of hydrocortisone reduce severe systemic inflammatory response syndrome and improve early outcome in a risk group of patients after cardiac surgery. Crit Care Med 2003; 31: 1068-74.
33. Netea, M.G., J.W. van der Meer, M. van Deuren, and B.J. Kullberg. Proinflammatory cytokines and sepsis syndrome: not enough, or too much of a good thing? Trends Immunol 2003; 24: 254-8.
34. Crouser, E.D. Therapeutic benefits of antioxidants during sepsis: is protection against oxidant-mediated tissue damage only half the story? Crit Care Med 2004; 32: 589-90.
35. Ritter, C., M. Andrades, M.L. Frota Junior, et al. Oxidative parameters and mortality in sepsis induced by cecal ligation and perforation. Intensive Care Med 2003; 29: 1782-9.
36. Taylor, D.E., A.J. Ghio, and C.A. Piantadosi. Reactive oxygen species produced by liver mitochondria of rats in sepsis. Arch Biochem Biophys 1995; 316: 70-6.
37. Power, C., N. Fanning, and H.P. Redmond. Cellular apoptosis and organ injury in sepsis: a review. Shock 2002; 18: 197-211.
38. Perl, M., C.S. Chung, J. Lomas-Neira, et al. Silencing of Fas, but not caspase-8, in lung epithelial cells ameliorates pulmonary apoptosis, inflammation, and neutrophil influx after hemorrhagic shock and sepsis. Am J Pathol 2005; 167: 1545-59.
39. Coopersmith, C.M., P.E. Stromberg, W.M. Dunne, et al. Inhibition of intestinal epithelial apoptosis and survival in a murine model of pneumonia-induced sepsis. Jama 2002; 287: 1716-21.
40. Wan, S., J.L. LeClerc, and J.L. Vincent. Cytokine responses to cardiopulmonary bypass: lessons learned from cardiac transplantation. Ann Thorac Surg 1997; 63: 269-76.
41. Giomarelli, P., S. Scolletta, E. Borrelli, and B. Biagioli. Myocardial and lung injury after cardiopulmonary bypass: role of interleukin (IL)-10. Ann Thorac Surg 2003; 76: 117-23.
42. Gesser, B., H. Leffers, T. Jinquan, et al. Identification of functional domains on human interleukin 10. Proc Natl Acad Sci U S A 1997; 94: 14620-5.
43. Ding, Y., L. Qin, S.V. Kotenko, S. Pestka, and J.S. Bromberg. A single amino acid determines the immunostimulatory activity of interleukin 10. J Exp Med 2000; 191: 213-24.
44. Jansen, N.J., W. van Oeveren, L. van den Broek, et al. Inhibition by dexamethasone of the reperfusion phenomena in cardiopulmonary bypass. J Thorac Cardiovasc Surg 1991; 102: 515-25.
45. Menasche, P., S. Haydar, J. Peynet, et al. A potential mechanism of vasodilation after warm heart surgery. The temperature-dependent release of cytokines. J Thorac Cardiovasc Surg 1994; 107: 293-9.
46. Finn, A., S. Naik, N. Klein, R.J. Levinsky, S. Strobel, and M. Elliott. Interleukin-8 release and neutrophil degranulation after pediatric cardiopulmonary bypass. J Thorac Cardiovasc Surg 1993; 105: 234-41.
47. Kawamura, T., R. Wakusawa, K. Okada, and S. Inada. Elevation of cytokines during open heart surgery with cardiopulmonary bypass: participation of interleukin 8 and 6 in reperfusion injury. Can J Anaesth 1993; 40: 1016-21.
48. Cremer, J., M. Martin, H. Redl, et al. Systemic inflammatory response syndrome after cardiac operations. Ann Thorac Surg 1996; 61: 1714-20.
49. Edmunds, L.H., Jr. Inflammatory response to cardiopulmonary bypass. Ann Thorac Surg 1998; 66: S12-6; discussion S25-8.
50. Kaysen, G.A. The microinflammatory state in uremia: causes and potential consequences. J Am Soc Nephrol 2001; 12: 1549-57.
51. Girndt, M., H. Kohler, E. Schiedhelm-Weick, J.F. Schlaak, K.H. Meyer zum Buschenfelde, and B. Fleischer. Production of interleukin-6, tumor necrosis factor alpha and interleukin-10 in vitro correlates with the clinical immune defect in chronic hemodialysis patients. Kidney Int 1995; 47: 559-65.
52. Dinarello, C.A. Cytokines: agents provocateurs in hemodialysis? Kidney Int 1992; 41: 683-94.
53. Schindler, R., G. Lonnemann, S. Shaldon, K.M. Koch, and C.A. Dinarello. Transcription, not synthesis, of interleukin-1 and tumor necrosis factor by complement. Kidney Int 1990; 37: 85-93.
54. Lonnemann, G., M. Bingel, J. Floege, K.M. Koch, S. Shaldon, and C.A. Dinarello. Detection of endotoxin-like interleukin-1-inducing activity during in vitro dialysis. Kidney Int 1988; 33: 29-35.
55. Woeltje, K.F., A. Mathew, M. Rothstein, S. Seiler, and V.J. Fraser. Tuberculosis infection and anergy in hemodialysis patients. Am J Kidney Dis 1998; 31: 848-52.
56. Matas, A.J., R.L. Simmons, C.M. Kjellstrand, T.J. Buselmeier, and J.S. Najarian. Increased incidence of malignancy during chronic renal failure. Lancet 1975; 1: 883-6.
57. Fleming, S.J., D.M. Moran, W.G. Cooksley, and J.L. Faoagali. Poor response to a recombinant hepatitis B vaccine in dialysis patients. J Infect 1991; 22: 251-7.
58. Kreft, B., M. Klouche, R. Kreft, H. Kirchner, and K. Sack. Low efficiency of active immunization against diphtheria in chronic hemodialysis patients. Kidney Int 1997; 52: 212-6.
59. Sester, U., M. Sester, M. Hauk, H. Kaul, H. Kohler, and M. Girndt. T-cell activation follows Th1 rather than Th2 pattern in haemodialysis patients. Nephrol Dial Transplant 2000; 15: 1217-23.
60. Meuer, S.C., M. Hauer, P. Kurz, K.H. Meyer zum Buschenfelde, and H. Kohler. Selective blockade of the antigen-receptor-mediated pathway of T cell activation in patients with impaired primary immune responses. J Clin Invest 1987; 80: 743-9.
61. Girndt, M., H. Kohler, E. Schiedhelm-Weick, K.H. Meyer zum Buschenfelde, and B. Fleischer. T cell activation defect in hemodialysis patients: evidence for a role of the B7/CD28 pathway. Kidney Int 1993; 44: 359-65.
62. Mosmann, T.R., H. Cherwinski, M.W. Bond, M.A. Giedlin, and R.L. Coffman. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol 1986; 136: 2348-57.
63. Vanholder, R., S. Ringoir, A. Dhondt, and R. Hakim. Phagocytosis in uremic and hemodialysis patients: a prospective and cross sectional study. Kidney Int 1991; 39: 320-7.
64. Gaudino, M., F. Andreotti, R. Zamparelli, et al. The -174G/C interleukin-6 polymorphism influences postoperative interleukin-6 levels and postoperative atrial fibrillation. Is atrial fibrillation an inflammatory complication? Circulation 2003; 108 Suppl 1: II195-9.
65. Libetta, C., T. Rampino, and A. Dal Canton. Polarization of T-helper lymphocytes toward the Th2 phenotype in uremic patients. Am J Kidney Dis 2001; 38: 286-95.
66. Alvarez-Lara, M.A., J. Carracedo, R. Ramirez, et al. The imbalance in the ratio of Th1 and Th2 helper lymphocytes in uraemia is mediated by an increased apoptosis of Th1 subset. Nephrol Dial Transplant 2004; 19: 3084-90.
67. Hertel, J., P.L. Kimmel, T.M. Phillips, and J.P. Bosch. Eosinophilia and cellular cytokine responsiveness in hemodialysis patients. J Am Soc Nephrol 1992; 3: 1244-52.
68. Cledes, J., M.P. Guillodo, J.P. Herve, et al. Hemodialysis hypereosinophilia. Contrib Nephrol 1988; 62: 109-17.
69. Olszyna, D.P., D. Pajkrt, F.N. Lauw, S.J. van Deventer, and T. van Der Poll. Interleukin 10 inhibits the release of CC chemokines during human endotoxemia. J Infect Dis 2000; 181: 613-20.
70. Girndt, M., H. Kaul, U. Sester, et al. Anti-inflammatory interleukin-10 genotype protects dialysis patients from cardiovascular events. Kidney Int 2002; 62: 949-55.
71. Stenvinkel, P., M. Ketteler, R.J. Johnson, et al. IL-10, IL-6, and TNF-alpha: central factors in the altered cytokine network of uremia--the good, the bad, and the ugly. Kidney Int 2005; 67: 1216-33.
72. Weiss, S.J. Tissue destruction by neutrophils. N Engl J Med 1989; 320: 365-76.
73. Kononen, J., L. Bubendorf, A. Kallioniemi, et al. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 1998; 4: 844-7.
74. Goding, J.W., Monoclonal Antibodies: Principles and Practice. . 1983, London: Academic Press
75. Bone, R.C., R.A. Balk, F.B. Cerra, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest 1992; 101: 1644-55.
76. Ohata, T., Y. Sawa, M. Takagi, et al. Hybrid artificial lung with interleukin-10 and endothelial constitutive nitric oxide synthase gene-transfected endothelial cells attenuates inflammatory reactions induced by cardiopulmonary bypass. Circulation 1998; 98: II269-74.
77. Ma, X., P. Ottino, H.E. Bazan, and N.G. Bazan. Platelet-activating factor (PAF) induces corneal neovascularization and upregulates VEGF expression in endothelial cells. Invest Ophthalmol Vis Sci 2004; 45: 2915-21.
78. Zhong, H., Y. Wu, L. Belardinelli, and D. Zeng. A2B adenosine receptors induce IL-19 from bronchial epithelial cells, resulting in TNF-alpha increase. Am J Respir Cell Mol Biol 2006; 35: 587-92.
79. Wuyts, W.A., B.M. Vanaudenaerde, L.J. Dupont, M.G. Demedts, and G.M. Verleden. Involvement of p38 MAPK, JNK, p42/p44 ERK and NF-kappaB in IL-1beta-induced chemokine release in human airway smooth muscle cells. Respir Med 2003; 97: 811-7.
80. Schwabe, R.F. and D.A. Brenner. Mechanisms of Liver Injury. I. TNF-alpha-induced liver injury: role of IKK, JNK, and ROS pathways. Am J Physiol Gastrointest Liver Physiol 2006; 290: G583-9.
81. Diehl, A.M. Cytokine regulation of liver injury and repair. Immunol Rev 2000; 174: 160-71.
82. Keel, M., U. Ungethum, U. Steckholzer, et al. Interleukin-10 counterregulates proinflammatory cytokine-induced inhibition of neutrophil apoptosis during severe sepsis. Blood 1997; 90: 3356-63.
83. Taneja, R., J. Parodo, S.H. Jia, A. Kapus, O.D. Rotstein, and J.C. Marshall. Delayed neutrophil apoptosis in sepsis is associated with maintenance of mitochondrial transmembrane potential and reduced caspase-9 activity. Crit Care Med 2004; 32: 1460-9.
84. Sayeed, M.M. Delay of neutrophil apoptosis can exacerbate inflammation in sepsis patients: cellular mechanisms. Crit Care Med 2004; 32: 1604-6.
85. Hsu, Y.H., H.H. Li, M.Y. Hsieh, et al. Function of interleukin-20 as a proinflammatory molecule in rheumatoid and experimental arthritis. Arthritis Rheum 2006; 54: 2722-33.
86. Hsing, C.H., M.Y. Hsieh, W.Y. Chen, E. Cheung So, B.C. Cheng, and M.S. Chang. Induction of interleukin-19 and interleukin-22 after cardiac surgery with cardiopulmonary bypass. Ann Thorac Surg 2006; 81: 2196-201.
87. Maggi, E., R. Bellazzi, F. Falaschi, et al. Enhanced LDL oxidation in uremic patients: an additional mechanism for accelerated atherosclerosis? Kidney Int 1994; 45: 876-83.
88. Drueke, T.B., T. Nguyen Khoa, Z.A. Massy, V. Witko-Sarsat, B. Lacour, and B. Descamps-Latscha. Role of oxidized low-density lipoprotein in the atherosclerosis of uremia. Kidney Int Suppl 2001; 78: S114-9.
89. Oral, H.B., S.V. Kotenko, M. Yilmaz, et al. Regulation of T cells and cytokines by the interleukin-10 (IL-10)-family cytokines IL-19, IL-20, IL-22, IL-24 and IL-26. Eur J Immunol 2006; 36: 380-8.
90. Bourbon, A., M. Vionnet, P. Leprince, et al. The effect of methylprednisolone treatment on the cardiopulmonary bypass-induced systemic inflammatory response. Eur J Cardiothorac Surg 2004; 26: 932-8.
91. Kunz, S., K. Wolk, E. Witte, et al. Interleukin (IL)-19, IL-20 and IL-24 are produced by and act on keratinocytes and are distinct from classical ILs. Exp Dermatol 2006; 15: 991-1004.
92. Butler, J., G.M. Rocker, and S. Westaby. Inflammatory response to cardiopulmonary bypass. Ann Thorac Surg 1993; 55: 552-9.
93. Oudemans-van Straaten, H.M., P.G. Jansen, F.J. Hoek, et al. Intestinal permeability, circulating endotoxin, and postoperative systemic responses in cardiac surgery patients. J Cardiothorac Vasc Anesth 1996; 10: 187-94.
94. Hennein, H.A., H. Ebba, J.L. Rodriguez, et al. Relationship of the proinflammatory cytokines to myocardial ischemia and dysfunction after uncomplicated coronary revascularization. J Thorac Cardiovasc Surg 1994; 108: 626-35.
95. Bubendorf, L., A. Nocito, H. Moch, and G. Sauter. Tissue microarray (TMA) technology: miniaturized pathology archives for high-throughput in situ studies. J Pathol 2001; 195: 72-9.
96. Wang, C., F. Yeung, P.-C. Liu, et al. Identification of a Novel Transcription Factor, GAGATA-binding Protein, Involved in Androgen-mediated Expression of Prostate-specific Antigen. J. Biol. Chem. 2003; 278: 32423-32430.
97. Schraml, P., J. Kononen, L. Bubendorf, et al. Tissue microarrays for gene amplification surveys in many different tumor types. Clin Cancer Res 1999; 5: 1966-75.
98. Kallioniemi, O.P., U. Wagner, J. Kononen, and G. Sauter. Tissue microarray technology for high-throughput molecular profiling of cancer. Hum Mol Genet 2001; 10: 657-62.
99. Hsing, C.H., C.L. Ho, L.Y. Chang, Y.L. Lee, S.S. Chuang, and M.S. Chang. Tissue microarray analysis of interleukin-20 expression. Cytokine 2006; 35: 44-52.
100. Wei, C.C., W.Y. Chen, Y.C. Wang, et al. Detection of IL-20 and its receptors on psoriatic skin. Clin Immunol 2005; 117: 65-72.
101. Sheikh, F., V.V. Baurin, A. Lewis-Antes, et al. Cutting edge: IL-26 signals through a novel receptor complex composed of IL-20 receptor 1 and IL-10 receptor 2. J Immunol 2004; 172: 2006-10.
102. Wegenka, U.M., N. Dikopoulos, J. Reimann, G. Adler, and C. Wahl. The murine liver is a potential target organ for IL-19, IL-20 and IL-24: Type I Interferons and LPS regulate the expression of IL-20R2. J Hepatol 2006;
103. Oberholzer, C., A. Oberholzer, M. Clare-Salzler, and L.L. Moldawer. Apoptosis in sepsis: a new target for therapeutic exploration. Faseb J 2001; 15: 879-92.
104. Qing, M., A. Nimmesgern, P.C. Heinrich, et al. Intrahepatic synthesis of tumor necrosis factor-alpha related to cardiac surgery is inhibited by interleukin-10 via the Janus kinase (Jak)/signal transducers and activator of transcription (STAT) pathway. Crit Care Med 2003; 31: 2769-75.
105. Matute-Bello, G., W.C. Liles, F. Radella, 2nd, et al. Neutrophil apoptosis in the acute respiratory distress syndrome. Am J Respir Crit Care Med 1997; 156: 1969-77.
106. Biffl, W.L., E.E. Moore, F.A. Moore, C.C. Barnett, Jr., V.S. Carl, and V.N. Peterson. Interleukin-6 delays neutrophil apoptosis. Arch Surg 1996; 131: 24-9; discussion 29-30.
107. Cox, G. IL-10 enhances resolution of pulmonary inflammation in vivo by promoting apoptosis of neutrophils. Am J Physiol 1996; 271: L566-71.
108. Daichou, Y., S. Kurashige, S. Hashimoto, and S. Suzuki. Characteristic cytokine products of Th1 and Th2 cells in hemodialysis patients. Nephron 1999; 83: 237-45.
109. Yokoyama, T., K. Nitta, K. Futatsuyama, et al. Identification of T helper cell subsets in continuous ambulatory peritoneal dialysis patients. Nephron 2001; 89: 215-8.
110. Nitta, K., T. Akiba, A. Kawashima, et al. Characterization of TH1/TH2 profile in uremic patients. Nephron 2002; 91: 492-5.
111. te Velde, A.A., R.J. Huijbens, K. Heije, J.E. de Vries, and C.G. Figdor. Interleukin-4 (IL-4) inhibits secretion of IL-1 beta, tumor necrosis factor alpha, and IL-6 by human monocytes. Blood 1990; 76: 1392-7.
112. Nagalakshmi, M.L., E. Murphy, T. McClanahan, and R. de Waal Malefyt. Expression patterns of IL-10 ligand and receptor gene families provide leads for biological characterization. Int Immunopharmacol 2004; 4: 577-92.
113. Fiorentino, D.F., A. Zlotnik, T.R. Mosmann, M. Howard, and A. O'Garra. IL-10 inhibits cytokine production by activated macrophages. J Immunol 1991; 147: 3815-22.
114. Poe, J.C., D.H. Wagner, Jr., R.W. Miller, R.D. Stout, and J. Suttles. IL-4 and IL-10 modulation of CD40-mediated signaling of monocyte IL-1beta synthesis and rescue from apoptosis. J Immunol 1997; 159: 846-52.