期刊文献+

A20 inhibits lipopolysaccharide-induced inflammation in enterocytes 被引量:3

A20 inhibits lipopolysaccharide-induced inflammation in enterocytes
下载PDF
导出
摘要 AIM To examine the role of A20 in the regulation of intestinal epithelial cells(IECs) inflammation.METHODS Using gene transfection,both stable overexpression and knockdown A20-expressed HT-29 cell lines were established.Accordingly,the cells were divided into the following groups:The control group,the A20 overexpression group,the A20 knockdown group and the respective controls.A20 was stimulated with lipo-polysaccharide(LPS) in a dose- and time-dependent manner and was detected using western blotting and real-time polymerase chain reaction(PCR) analyses.Immunofluorescence and western blotting analyses were performed to investigate the role of A20 in the regulation of nuclear factor(NF)-κB activation and translocation into the nucleus.ELISA and real-time PCR were performed to examine A20 in regulating the release of the following inflammatory cytokines:Tumor necrosis factor(TNF)-a,interleukin(IL)-1b,IL-6 and IL-8.RESULTS The expression of A20 in IECs was inducible.When intestinal epithelial cells were subjected to the stimulation of LPS,the expression of A20 was increased,and the expression of A20 was induced in a dose-and timedependent manner.The expression of A20 was very low in HT-29 cells without LPS stimulation but rapidly increased and was maintained at a high level 2-4 h after stimulationwith LPS.These levels gradually declined with a change in time-course,and the expression of A20 increased with increasing LPS stimulation.Western blotting and immunofluorescence revealed that overexpression of A20 can inhibit NF-κB activation and its translocation to the nucleus.The overexpression of A20 can reduce the levels of proinflammatory cytokines involved in the pathophysiology of inflammatory bowel disease.There was no significant difference in the expression of IL-8 mR NA in the control group,A20 overexpression group or A20 knockdown group without LPS stimulation(P > 0.05);however,while after 2 h,4 h and 8 h stimulation with LPS,the expression of IL-8 in the A20 overexpression group was lower than the control group and the A20 knockdown group(P < 0.05 or P < 0.01).The expression of TNF-a was different at different time points after 8 h of LPS stimulation(F = 31.33,DF = 5,P < 0.001),and the expression of TNF-a increased as the LPS stimulation time increased.Upon LPS stimulation,lower levels of TNF-a were detected in the A20 overexpression cell lines(P < 0.05).There were no significant differences in the induction of IL-6 and IL-1b among the control group,A20 overexpression group and A20 knockdown group(P > 0.05).CONCLUSION A20 plays an important role in limiting inflammation by inhibiting LPS-induced NF-κB responses in the gut luminal.A20 may be a potential therapeutic tool for inflammatory diseases. AIMTo examine the role of A20 in the regulation of intestinal epithelial cells (IECs) inflammation. METHODSUsing gene transfection, both stable overexpression and knockdown A20-expressed HT-29 cell lines were established. Accordingly, the cells were divided into the following groups: The control group, the A20 overexpression group, the A20 knockdown group and the respective controls. A20 was stimulated with lipopolysaccharide (LPS) in a dose- and time-dependent manner and was detected using western blotting and real-time polymerase chain reaction (PCR) analyses. Immunofluorescence and western blotting analyses were performed to investigate the role of A20 in the regulation of nuclear factor (NF)-&kappa;B activation and translocation into the nucleus. ELISA and real-time PCR were performed to examine A20 in regulating the release of the following inflammatory cytokines: Tumor necrosis factor (TNF)-&alpha;, interleukin (IL)-1&beta;, IL-6 and IL-8. RESULTSThe expression of A20 in IECs was inducible. When intestinal epithelial cells were subjected to the stimulation of LPS, the expression of A20 was increased, and the expression of A20 was induced in a dose- and time-dependent manner. The expression of A20 was very low in HT-29 cells without LPS stimulation but rapidly increased and was maintained at a high level 2-4 h after stimulation with LPS. These levels gradually declined with a change in time-course, and the expression of A20 increased with increasing LPS stimulation. Western blotting and immunofluorescence revealed that overexpression of A20 can inhibit NF-&kappa;B activation and its translocation to the nucleus. The overexpression of A20 can reduce the levels of proinflammatory cytokines involved in the pathophysiology of inflammatory bowel disease. There was no significant difference in the expression of IL-8 mRNA in the control group, A20 overexpression group or A20 knockdown group without LPS stimulation (P > 0.05); however, while after 2 h, 4 h and 8 h stimulation with LPS, the expression of IL-8 in the A20 overexpression group was lower than the control group and the A20 knockdown group (P P P P P > 0.05). CONCLUSIONA20 plays an important role in limiting inflammation by inhibiting LPS-induced NF-&kappa;B responses in the gut luminal. A20 may be a potential therapeutic tool for inflammatory diseases.
出处 《World Journal of Gastrointestinal Pharmacology and Therapeutics》 CAS 2016年第4期540-549,共10页 世界胃肠药理与治疗学杂志(英文版)(电子版)
基金 Supported by The National Natural Science Foundation of China,No.81300291
关键词 A20 (TNFAIP3 ) LIPOPOLYSACCHARIDE 原子 factor-&kappa B 煽动性的肠疾病 A20 (TNFAIP3) Lipopolysaccharide Nuclear factor-&kappa B Inflammatory bowel disease
  • 相关文献

参考文献2

二级参考文献91

  • 1[1]Strober W,Nakamura K,Kitani A.The SAMP1/Yit mouse:another step closer to modeling human inflammatory bowel disease.J Clin Invest 2001; 107:667-670
  • 2[2]Bouma G,Strober W.The immunological and genetic basis of inflammatory bowel disease.Nat Rev Immunol 2003; 3:521-533
  • 3[3]Fiocchi C.Inflammatory bowel disease:etiology and pathogenesis.Gastroenterology 1998; 115:182-205
  • 4[4]Murch SH,Braegger CP,Walker-Smith JA,MacDonald TT.Location of tumour necrosis factor alpha by immunohistochemistry in chronic inflammatory bowel disease.Gut 1993;34:1705-1709
  • 5[5]Breese EJ,Michie CA,Nicholls SW,Murch SH,Williams CB,Domizio P,Walker-Smith JA,MacDonald TT.Tumor necrosis factor alpha-producing cells in the intestinal mucosa of children with inflammatory bowel disease.Gastroenterology 1994;106:1455-1466
  • 6[6]Braegger CP,Nicholls S,Murch SH,Stephens S,MacDonald TT.Tumour necrosis factor alpha in stool as a marker of intestinal inflammation.Lancet 1992; 339:89-91
  • 7[7]Powrie F,Leach MW,Mauze S,Menon S,Caddie LB,Coffman RL.Inhibition of Th1 responses prevents inflammatory bowel disease in scid mice reconstituted with CD45RBhi CD4+ T cells.Immunity 1994; 1:553-562
  • 8[8]Kosiewicz MM,Nast CC,Krishnan A,Rivera-Nieves J,Moskaluk CA,Matsumoto S,Kozaiwa K,Cominelli F.Th1-type responses mediate spontaneous ileitis in a novel murine model of Crohn's disease.J Clin Invest 2001; 107:695-702
  • 9[9]Van den Brande JM,Braat H,van den Brink GR,Versteeg HH,Bauer CA,Hoedemaeker I,van Montfrans C,Hommes DW,Peppelenbosch MP,van Deventer SJ.Infliximab but not etanercept induces apoptosis in lamina propria T-lymphocytes from patients with Crohn's disease.Gastroenterology 2003; 124:1774-1785
  • 10[10]Targan SR,Hanauer SB,van Deventer SJ,Mayer L,Present DH,Braakman T,DeWoody KL,Schaible TF,Rutgeerts PJ.A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn's disease.Crohn's Disease cA2 Study Group.N Engl JMed 1997; 337:1029-1035

共引文献25

同被引文献5

引证文献3

二级引证文献7

相关作者

内容加载中请稍等...

相关机构

内容加载中请稍等...

相关主题

内容加载中请稍等...

浏览历史

内容加载中请稍等...
;
使用帮助 返回顶部