リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Pathological mechanisms in Crohn’s disease via dysbiosis triggered by Paneth cell α-defensin misfolding」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Pathological mechanisms in Crohn’s disease via dysbiosis triggered by Paneth cell α-defensin misfolding

清水, 由宇 北海道大学

2020.12.25

概要

In the human intestinal tract, forty trillion of commensal bacteria construct complex ecosystem termed intestinal microbiota1 . The human intestinal microbiota is thought to possess approximately 10 million genes which are more than several hundred times larger than that of the host2 , and involved in many aspects of host physiology, that includes energy metabolism3 , immune system regulation4 , and nervous system development5 . Recently, associations with imbalance of the intestinal microbiota, termed dysbiosis, and many diseases, including chronic lifestyle diseases such as obesity and diabetes, immunological disorders, and nervous system diseases have been reported6 . a-Defensin, a major family of mammalian antimicrobial peptides, are known regulators of the intestinal microbiota. These ~ 4 kDa, basic peptides are characterized by evolutionally conserved Cys residue positions that are invariantly spaced to form disulfide bonds between CysI -CysVI, CysII-CysIV, and CysIII-CysV 7 . In the intestinal epithelium, a-defensin occur only in intracellular dense-core secretory granules of Paneth cells, one of the major terminally differentiated lineages of the small intestine. Paneth cells, which reside at the base of the crypts of Lieberkühn, release secretory granules that are rich in a-defensin, termed cryptdins (Crps) in mice and HD5 and HD6 in human, in response to bacteria and other stimuli at effective concentrations, thereby contributing to enteric innate immunity8‒13. Also, Paneth cell a-defensin contribute to regulating the composition of the intestinal microbiota in an activity-dependent manner in vivo as well as affecting development of host adaptive immunity14. Furthermore, oral administration of Crp4 prevents severe dysbiosis in mouse graft-versushost disease15,16, indicating that Paneth cell a-defensin secreted into the intestinal lumen contribute not only to innate immunity but also to maintenance of intestinal homeostasis by regulating the intestinal microbiota17,18.

Recently, a relationship has been revealed between the intestinal microbiota and the pathophysiology of Crohn’s disease (CD)19. CD is a chronic inflammatory bowel disease that may affect the entire gastrointestinal tract, especially the terminal ileum, with chronic inflammation and ulceration20. The number of patients with CD has been increasing continuously worldwide, including Europe, The Americas and Asia20‒22. Because CD typically appears in younger people in their teens and 20s and any radical treatments have yet to be developed, the patient’s quality of life could be diminished for a lifetime. Although a complete picture of CD pathogenesis is lacking, there is consensus that dysbiosis and dysregulated immune responses to the intestinal microbiota play important roles20. Moreover, both genetic factors consisting of more than 160 susceptibility loci23, as well as environmental factors such as overuse of antibiotics24 and adoption of “westernized diets”25 have been reported as CD risk factors, and these factors are suggested to induce pathophysiology of CD via dysbiosis26.

Evidence shows that certain Paneth cell defects are involved in CD onset and pathophysiology. Paneth cells continuously synthesize high levels of secretory proteins in the endoplasmic reticulum (ER) and are susceptible to ER stress, a failure of maintaining ER homeostasis induced by accumulation of misfolded proteins27. Excessive ER stress induces series of signal cascades termed unfolded protein response (UPR), and UPR manages to recover ER homeostasis by inducing suppression of protein translation, induction of ER chaperones which help refolding of the misfolded proteins, and degradation of the misfolded proteins via autophagy and activation of ubiquitin-proteasome system28. Several genes involved in resolution of ER stress affect CD susceptibility, and deletions or mutations of such gene. For example, UPR-related genes XBP129 and AGR230, autophagy-related genes ATG16L131, IRGM132, and LRRK233 cause Paneth cell abnormalities in granule morphology and cellular localization in mouse models. In CD patients with mutations in XBP129 and ATG16L131, Paneth cell abnormalities occur which are similar to those observed in genetically deficient mice. Moreover, several studies have identified relationships between ER stress in Paneth cells and disruptions of the intestinal microbiota in CD. The appearance rate of Paneth cells with abnormal granule morphology in CD patients is associated with dysbiosis that is characterized by reduction of diversity and decrease of anti-inflammatory bacteria such as Faecalibacterium34. CD patients positive for the ER stress marker GRP78 in Paneth cells harbor greater numbers of CD associated enteroinvasive Escherichia coli compared to patients that lack GPR78-positive Paneth cells35. Although these findings suggest the involvement of ER stress, Paneth cell dysfunction, and dysbiosis in CD pathophysiology, causal relationships between these factors remain to be demonstrated.

参考文献

1. Sender, R., Fuchs, S. & Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 164, 337–340 (2016).

2. Nishijima, S. et al. The gut microbiome of healthy Japanese and its microbial and functional uniqueness. DNA Research 23, 125–133 (2016).

3. Bäckhed, F. et al. The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences 101, 15718–15723 (2004).

4. Atarashi, K. et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500, 232–236 (2013).

5. Ma, Q. et al. Impact of microbiota on central nervous system and neurological diseases: the gutbrain axis. Journal of Neuroinflammation 16, 53 (2019).

6. Kho, Z. Y. & Lal, S. K. The Human Gut Microbiome - A Potential Controller of Wellness and Disease. Frontiers in Microbiology 9, 1835 (2018).

7. Selsted, M. E. & Ouellette, A. J. Mammalian defensins in the antimicrobial immune response. Nature Immunology 6, 551–557 (2005).

8. Ouellette, A. J. et al. Developmental regulation of cryptdin, a corticostatin/defensin precursor mRNA in mouse small intestinal crypt epithelium. The Journal of Cell Biology 108, 1687–1695 (1989).

9. Jones, D. E. & Bevins, C. L. Defensin-6 mRNA in human Paneth cells: implications for antimicrobial peptides in host defense of the human bowel. FEBS Letters 315, 187–192 (1993).

10. Porter, E. M., Liu, L., Oren, A., Anton, P. A. & Ganz, T. Localization of human intestinal defensin 5 in Paneth cell granules. Infection and Immunity 65, 2389–2395 (1997).

11. Wilson, C. L. et al. Regulation of intestinal alpha-defensin activation by the metalloproteinase matrilysin in innate host defense. Science 286, 113–117 (1999).

12. Ayabe, T. et al. Secretion of microbicidal a-defensins by intestinal Paneth cells in response to bacteria. Nature Immunology 1, 113–118 (2000).

13. Salzman, N. H., Ghosh, D., Huttner, K. M., Paterson, Y. & Bevins, C. L. Protection against enteric salmonellosis in transgenic mice expressing a human intestinal defensin. Nature 422, 522–526 (2003).

14. Salzman, N. H. et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nature Immunology 11, 76–83 (2010).

15. Hayase, E. et al. R-Spondin1 expands Paneth cells and prevents dysbiosis induced by graft-versushost disease. The Journal of Experimental Medicine 214, 3507–3518 (2017).

16. Eriguchi, Y. et al. Graft-versus-host disease disrupts intestinal microbial ecology by inhibiting Paneth cell production of α-defensins. Blood 120, 223–231 (2012).

17. Nakamura, K., Sakuragi, N., Takakuwa, A. & Ayabe, T. Paneth cell α-defensins and enteric microbiota in health and disease. Bioscience of Microbiota, Food and Health 35, 57–67 (2016).

18. Eriguchi, Y. et al. Essential role of IFN-γ in T cell-associated intestinal inflammation. JCI Insight 3, e121886 (2018).

19. Zuo, T. & Ng, S. C. The Gut Microbiota in the Pathogenesis and Therapeutics of Inflammatory Bowel Disease. Frontiers in Microbiology 9, 2247 (2018).

20. Torres, J., Mehandru, S., Colombel, J.-F. & Peyrin-Biroulet, L. Crohn’s disease. Lancet 389, 1741– 1755 (2017).

21. Burisch, J., Jess, T., Martinato, M., Lakatos, P. L. & ECCO-EpiCom. The burden of inflammatory bowel disease in Europe. Journal of Crohn’s and Colitis 7, 322–337 (2013).

22. Brusaferro, A. et al. Gut dysbiosis and paediatric Crohn’s disease. The Journal of Infection 78, 1– 7 (2019).

23. Jostins, L. et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

24. Shaw, S. Y., Blanchard, J. F. & Bernstein, C. N. Association Between the Use of Antibiotics and New Diagnoses of Crohnʼs Disease and Ulcerative Colitis. American Journal of Gastroenterology 106, 2133–2142 (2011).

25. Hou, J. K., Abraham, B. & El-Serag, H. Dietary Intake and Risk of Developing Inflammatory Bowel Disease: A Systematic Review of the Literature. The American Journal of Gastroenterology 106, 563–573 (2011).

26. Somineni, H. K. & Kugathasan, S. The Microbiome in Patients With Inflammatory Diseases. Clinical Gastroenterology and Hepatology 17, 243–255 (2019).

27. Hosomi, S., Kaser, A. & Blumberg, R. S. Role of endoplasmic reticulum stress and autophagy as interlinking pathways in the pathogenesis of inflammatory bowel disease. Current Opinion in Gastroenterology 31, 81–88 (2015).

28. Bernales, S., Papa, F. R. & Walter, P. Intracellular Signaling by the Unfolded Protein Response. Annual Review of Cell and Developmental Biology 22, 487–508 (2006).

29. Kaser, A. et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 134, 743–756 (2008).

30. Zhao, F. et al. Disruption of Paneth and goblet cell homeostasis and increased endoplasmic reticulum stress in Agr2-/- mice. Developmental Biology 338, 270–279 (2010).

31. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 456, 259–263 (2008).

32. Liu, B. et al. Irgm1-deficient mice exhibit Paneth cell abnormalities and increased susceptibility to acute intestinal inflammation. American Journal of Physiology Gastrointestinal and Liver Physiology 305, G573-84 (2013).

33. Zhang, Q. et al. Commensal bacteria direct selective cargo sorting to promote symbiosis. Nature Immunology 16, 918–926 (2015).

34. Liu, T. C. et al. Paneth cell defects in Crohn’s disease patients promote dysbiosis. JCI Insight 1, e86907 (2016).

35. Deuring, J. J. et al. Genomic ATG16L1 risk allele-restricted Paneth cell ER stress in quiescent Crohn’s disease. Gut 63, 1081–1091 (2014).

36. Hamdan, N., Kritsiligkou, P. & Grant, C. M. ER stress causes widespread protein aggregation and prion formation. The Journal of Cell Biology 216, 2295–2304 (2017).

37. Tsuchiya, Y. et al. IRE1-XBP1 pathway regulates oxidative proinsulin folding in pancreatic β cells. The Journal of Cell Biology 217, 1287–1301 (2018).

38. Cook, K. M. & Hogg, P. J. Post-Translational Control of Protein Function by Disulfide Bond Cleavage. Antioxidant & Redox Signaling 18, 1987–2015 (2013).

39. Masuda, K., Sakai, N., Nakamura, K., Yoshioka, S. & Ayabe, T. Bactericidal activity of mouse αdefensin cryptdin-4 predominantly affects noncommensal bacteria. Journal of Innate Immunity 3, 315–326 (2011).

40. Tanabe, H. et al. Denatured human alpha-defensin attenuates the bactericidal activity and the stability against enzymatic digestion. Biochemical and Biophysical Research Communications 358, 349–355 (2007).

41. Rivera-Nieves, J. et al. Emergence of perianal fistulizing disease in the SAMP1/YitFc mouse, a spontaneous model of chronic ileitis. Gastroenterology 124, 972–982 (2003).

42. Pizarro, T. T. et al. SAMP1/YitFc mouse strain: a spontaneous model of Crohn’s disease-like ileitis. Inflammatory Bowel Diseases 17, 2566–2584 (2011).

43. Cominelli, F., Arseneau, K. O., Rodriguez-Palacios, A. & Pizarro, T. T. Uncovering Pathogenic Mechanisms of Inflammatory Bowel Disease Using Mouse Models of Crohn’s Disease-Like Ileitis: What is the Right Model? Cellular and Molecular Gastroenterology and Hepatology 4, 19–32 (2017).

44. Rath, H. C. et al. Normal luminal bacteria, especially Bacteroides species, mediate chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin transgenic rats. The Journal of Clinical Investigation 98, 945–953 (1996).

45. Yokoi, Y. et al. Paneth cell granule dynamics on secretory responses to bacterial stimuli in enteroids. Scientific Reports 9, 2710 (2019).

46. Selsted, M. E. Investigational approaches for studying the structures and biological functions of myeloid antimicrobial peptides. Genetic Engineering 15, 131–147 (1993).

47. Nakamura, K., Sakuragi, N. & Ayabe, T. A monoclonal antibody-based sandwich enzyme-linked immunosorbent assay for detection of secreted α-defensin. Analytical Biochemistry 443, 124–131 (2013).

48. Takakuwa, A. et al. Butyric Acid and Leucine Induce α-Defensin Secretion from Small Intestinal Paneth Cells. Nutrients 11, (2019).

49. Schägger, H. & Jagow, G. von. Tricine-sodium dodecyl sulfate-polyacrylamide gel electrophoresis for the separation of proteins in the range from 1 to 100 kDa. Analytical Biochemistry 166, 368– 379 (1987).

50. Herlemann, D. P. et al. Transitions in bacterial communities along the 2000 km salinity gradient of the Baltic Sea. ISME Journal 5, 1571–1579 (2011).

51. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nature Methods 7, 335–336 (2010).

52. Edgar, R. C., Haas, B. J., Clemente, J. C., Quince, C. & Knight, R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics 27, 2194–2200 (2011).

53. Edgar, R. C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461 (2010).

54. Reikvam, D. H. et al. Depletion of murine intestinal microbiota: effects on gut mucosa and epithelial gene expression. PLoS ONE 6, e17996 (2011).

55. Vidrich, A. et al. Altered epithelial cell lineage allocation and global expansion of the crypt epithelial stem cell population are associated with ileitis in SAMP1/YitFc mice. The American Journal of Pathology 166, 1055–1067 (2005).

56. Batlle, E. et al. Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell 111, 251–263 (2002).

57. Dekaney, C. M., King, S., Sheahan, B. & Cortes, J. E. Mist1 Expression Is Required for Paneth Cell Maturation. Cellular and Molecular Gastroenterology and Hepatology 8, 549–560 (2019).

58. Ouellette, A. J. et al. Mouse Paneth cell defensins: primary structures and antibacterial activities of numerous cryptdin isoforms. Infection and Immunity 62, 5040–5047 (1994).

59. Maemoto, A. et al. Functional analysis of the alpha-defensin disulfide array in mouse cryptdin-4. The Journal of Biological Chemistry 279, 44188–44196 (2004).

60. Walker, A. W. et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiology 11, 7 (2011).

61. Manichanh, C. et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 55, 205–211 (2006).

62. Nishino, K. et al. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. Journal of Gastroenterology 53, 95–106 (2017).

63. Frank, D. N. et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences 104, 13780–13785 (2007).

64. Gophna, U., Sommerfeld, K., Gophna, S., Doolittle, W. F. & Zanten, S. J. O. V. van. Differences between tissue-associated intestinal microfloras of patients with Crohn’s disease and ulcerative colitis. Journal of Clinical Microbiology 44, 4136–4141 (2006).

65. Marini, M. et al. TNF-alpha neutralization ameliorates the severity of murine Crohn’s-like ileitis by abrogation of intestinal epithelial cell apoptosis. Proceedings of the National Academy of Sciences 100, 8366–8371 (2003).

66. Odashima, M. et al. Activation of A2A Adenosine Receptor Attenuates Intestinal Inflammation in Animal Models of Inflammatory Bowel Disease. Gastroenterology 129, 26–33 (2005).

67. Million, M. et al. New insights in gut microbiota and mucosal immunity of the small intestine. Human Microbiome Journal 7–8, 23–32 (2018).

68. Wexler, H. M. Bacteroides: the good, the bad, and the nitty-gritty. Clinical Microbiology Reviews 20, 593–621 (2007).

69. Walker, A. et al. Sulfonolipids as novel metabolite markers of Alistipes and Odoribacter affected by high-fat diets. Scientific Reports 7, 1–10 (2017).

70. Ferguson, L. R., Laing, B., Marlow, G. & Bishop, K. The role of vitamin D in reducing gastrointestinal disease risk and assessment of individual dietary intake needs: Focus on genetic and genomic technologies. Molecular Nutrition & Food Research 60, 119–133 (2015).

71. Gombart, A. F., Borregaard, N. & Koeffler, H. P. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB Journal 19, 1067–1077 (2005).

72. Su, D. et al. Vitamin D Signaling through Induction of Paneth Cell Defensins Maintains Gut Microbiota and Improves Metabolic Disorders and Hepatic Steatosis in Animal Models. Frontiers in Physiology 7, 471–18 (2016).

73. Wu, S. et al. Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis. Gut 64, 1082–1094 (2015).

74. VanDussen, K. L. et al. Genetic variants synthesize to produce Paneth cell phenotypes that define subtypes of Crohn’s disease. Gastroenterology 146, 200–209 (2014).

75. Wehkamp, J. et al. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-defensin expression. Gut 53, 1658–1664 (2004).

76. Williams, A. D. et al. Human alpha defensin 5 is a candidate biomarker to delineate inflammatory bowel disease. PLoS ONE 12, e0179710 (2017).

77. Ananthakrishnan, A. N. et al. Zinc intake and risk of Crohn’s disease and ulcerative colitis: a prospective cohort study. International Journal of Epidemiology 44, 1995–2005 (2015).

78. Guo, X. et al. Rutin and Its Combination With Inulin Attenuate Gut Dysbiosis, the Inflammatory Status and Endoplasmic Reticulum Stress in Paneth Cells of Obese Mice Induced by High-Fat Diet. Frontiers in Microbiology 9, 2651 (2018).

79. Podany, A. B., Wright, J., Lamendella, R., Soybel, D. I. & Kelleher, S. L. ZnT2-Mediated Zinc Import Into Paneth Cell Granules Is Necessary for Coordinated Secretion and Paneth Cell Function in Mice. Cellular and Molecular Gastroenterology and Hepatology 2, 369–383 (2016).

80. Hess, D. A. et al. MIST1 Links Secretion and Stress as both Target and Regulator of the Unfolded Protein Response. Molecular and Cell Biology 36, 2931–2944 (2016).

81. Pin, C. L., Rukstalis, J. M., Johnson, C. & Konieczny, S. F. The bHLH transcription factor Mist1 is required to maintain exocrine pancreas cell organization and acinar cell identity. Journal of Cell Biology 155, 519–530 (2001).

82. Jing, W., Hunter, H. N., Tanabe, H., Ouellette, A. J. & Vogel, H. J. Solution structure of cryptdin4, a mouse Paneth cell a-defensin. Biochemistry 43, 15759–15766 (2004).

83. Szyk, A. et al. Crystal structures of human alpha-defensins HNP4, HD5, and HD6. Protein Science  15, 2749–2760 (2006).

84. Hadjicharalambous, C. et al. Mechanisms of alpha-defensin bactericidal action: comparative membrane disruption by Cryptdin-4 and its disulfide-null analogue. Biochemistry 47, 12626–12634 (2008).

85. Zhang, Y., Cougnon, F. B. L., Wanniarachchi, Y. A., Hayden, J. A. & Nolan, E. M. Reduction of human defensin 5 affords a high-affinity zinc-chelating peptide. ACS chemical biology 8, 1907– 1911 (2013).

86. Meyers, S. et al. Fecal alpha 1-antitrypsin measurement: an indicator of Crohn’s disease activity. Gastroenterology 89, 13–18 (1985).

87. Wanniarachchi, Y. A., Kaczmarek, P., Wan, A. & Nolan, E. M. Human defensin 5 disulfide array mutants: disulfide bond deletion attenuates antibacterial activity against Staphylococcus aureus. Biochemistry 50, 8005–8017 (2011).

88. Schroeder, B. O. et al. Paneth cell α-defensin 6 (HD-6) is an antimicrobial peptide. Mucosal Immunology 8, 661–671 (2015).

89. Wang, C. et al. Reduction Impairs the Antibacterial Activity but Benefits the LPS Neutralization Ability of Human Enteric Defensin 5. Scientific Reports 6, 22875 (2016).

90. Hodin, C. M. et al. Reduced Paneth cell antimicrobial protein levels correlate with activation of the unfolded protein response in the gut of obese individuals. The Journal of pathology 225, 276– 284 (2011).

91. Grootjans, J. et al. Level of activation of the unfolded protein response correlates with Paneth cell apoptosis in human small intestine exposed to ischemia/reperfusion. Gastroenterology 140, 529- 539.e3 (2011).

92. Gyongyosi, B. et al. Alcohol-induced IL-17A production in Paneth cells amplifies endoplasmic reticulum stress, apoptosis, and inflammasome-IL-18 activation in the proximal small intestine in mice. Mucosal Immunology 12, 930–944 (2019)

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る