ISSN 2415-3060 (print), ISSN 2522-4972 (online)
  • 2 of 58
Up
УЖМБС 2020, 5(6): 17–26
https://doi.org/10.26693/jmbs05.06.017
Medicine. Reviews

Activation of Purinergic Signaling Pathways during the Development of Inflammatory Bowel Disease

Zherebyatiev O. S., Voitovich O. V., Motilonok T. Yu., Egorov A. A., Burega I. Yu.
Abstract

Inflammatory bowel disease is an important illness of unclear pathogenesis associated with major defects in mucosal immunoregulation and develops in genetically susceptible individuals. These abnormalities often occur in association with microbial dysbiosis and result in unfettered inflammation of the intestine and extraintestinal tissues. Such events result in long-term morbidity and possibly even death, in otherwise healthy adults and children. Dampening inflammation and re-establishing immune tolerance in inflammatory bowel disease remain the major therapeutic goal. However, existing inflammatory bowel disease therapies albeit providing recent advances, still largely rely on broad-based immunosuppression. For example, only around half of the patients treated with anti-TNF agents show substantive clinical responses. These improvements are often self-limited, while unfortunately increasing the risk of opportunistic infections. The purpose of the study was to investigate the control of mucosal immune responses, which are based on fundamental signaling pathways. Long-term interests in the regulation of purinergic signaling are now being leveraged to develop innovative and hopefully non-toxic therapies for inflammatory bowel disease. This review and the accompanying articles in this special issue address new therapeutic concepts in inflammatory bowel disease, as based on recent, linked work in hypoxia and purinergic signaling, mucosal barrier functions and microRNA biology. In several recent, comprehensive reviews, have already addressed the biological functions of ectoenzymes, such as CD39, CD73, and CD38, in the regulation of purinergic signaling and control of extracellular adenosine levels. Others, have noted the importance of these mechanisms in immunomodulation, as in cancer and inflammation. The ectonucleotidases of the CD39 family, in particular, have major impacts on the dynamic equilibrium of proinflammatory extracellular ATP, ADP nucleotides vs. the immunosuppressive potential of adenosine nucleosides. CD39 plays a dominant role in purinergic regulation of vascular inflammation, thrombosis, and the immune response in such settings. The relevance and importance of these purinergic signaling pathways in selected neoplastic states (lymphoma and chronic leukemia) and inflammatory diseases (sepsis and autoimmunity) have been already alluded to in recent work. A brief synopsis of the major components of purinergic signaling; chiefly for those not familiar to this field, will focus on very recent work detailing the immunomodulation of CD39 on T cells and other immune cells by both genetic and environmental factors in the setting of inflammatory bowel disease and experimental colitis, inclusive of the new roles for natural metabolites such as bilirubin, and will also briefly cover the role of CD39 expression on exosomes and microparticles, in control of inflammation in the gut and touch on the relevance of the microbiome. Lastly, it will cover the emerging importance of other NTPDases of the CD39 family and speculate on their role in controlling gut inflammation. Conclusion. Review of the literature with own data is devoted to description of the recent advances in the study purinergic signaling pathways implicated in immune dysregulation, in the pathogenesis of inflammatory bowel disease. Our focus in this review is on novel aspects of the functions of CD39 and related nucleoside triphosphate diphosphohydrolases in inflammatory bowel disease

Keywords: adenosine, ATP, E-NTPDase (ecto-nucleoside triphosphate diphosphohydrolase family, inflammatory bowel disease, lymphocytes

Full text: PDF (Ukr) 296K

References
  1. Graziano L. Binding to DNA of the RNA-polymerase II C-terminal domain allows discrimination between Cdk7 and Cdk9 phosphorylation. Nucleic Acids Res. 2009 Mar; 37(4): 1260-1268. https://doi.org/10.1093/nar/gkn1061
  2. Burnstock G, Knight GE. Cellular distribution and functions of P2 receptor subtypes in different systems. Int Rev Cytol. 2004; 240: 31-304. https://doi.org/10.1016/S0074-7696(04)40002-3
  3. Burnstock G. Purinergic signaling and vascular cell proliferation and death. Arterioscler Thromb Vasc Biol. 2002 Mar 1; 22(3): 364-73. https://doi.org/10.1161/hq0302.105360
  4. Resta R, Yamashita Y, Thompson LF. Ecto-enzyme and signaling functions of lymphocyte CD73. Immunol Rev. 1998 Feb; 161: 95-109. https://doi.org/10.1111/j.1600-065x.1998.tb01574.x
  5. Holger K.E, Michail V.S, Simon C.R. Purinergic Signaling during Inflammation. N Engl J Med. 2012 Dec 13; 367(24): 2322-2333. https://doi.org/10.1056/NEJMra1205750
  6. Beldi G, Enjyoji K, Wu Y, Miller L, Banz Y, Sun X, et al. The role of purinergic signaling in the liver and in transplantation: effects of extracellular nucleotides on hepatic graft vascular injury, rejection and metabolism. Front Biosci. 2008 Jan 1; 13: 2588-603. https://doi.org/10.2741/2868
  7. Robson SC, Sévigny J, Zimmermann H. The E-NTPDase family of ectonucleotidases: Structure function relationships and pathophysiological significance. Purinergic Signal. 2006 Jun; 2(2): 409-30. https://doi.org/10.1007/s11302-006-9003-5
  8. Alberto LH, Antonella C, Gianluca Z, Santina B, Valeria Q, Andrea ZA. CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology. 2013 Sep 1; 2(9): e26246. https://doi.org/10.4161/onci.26246
  9. Cekic C, Linden J. Purinergic regulation of the immune system. Nat Rev Immunol. 2016 Mar; 16(3): 177-92. https://doi.org/10.1038/nri.2016.4
  10. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007 Jun 11; 204(6): 1257–1265. https://doi.org/10.1084/jem.20062512
  11. Dwyer KM, Hanidziar D, Putheti P, Hill PA, Pommey S, McRae JL, et al. Expression of CD39 by human peripheral blood CD4+ CD25+ T cells denotes a regulatory memory phenotype. Am J Transplant. 2010 Nov; 10(11): 2410-20. https://doi.org/10.1111/j.1600-6143.2010.03291.x
  12. Cohen HB, Briggs KT, Marino JP, Ravid K, Robson SC, Mosser DM. TLR stimulation initiates a CD39-based autoregulatory mechanism that limits macrophage inflammatory responses. Blood. 2013 Sep 12; 122(11): 1935-45. https://doi.org/10.1182/blood-2013-04-496216
  13. Kamyshnyy AM, Grynevych YV, Degen AS, Topol YA, Buga TM. Th17-kletki i ikh rol v razvitii autoimmunnykh zabolevaniy [Th17 cells and their role in the development of autoimmune diseases]. Zaporozhskiy meditsinskiy zhurnal. 2011; 6: 81-87. [Russian]
  14. Jin X, Shepherd RK, Duling BR, Linden J. Inosine binds to A3 adenosine receptors and stimulates mast cell degranulation. J Clin Invest. 1997; 100(11): 2849-2857. doi:10.1172/JCI119833
  15. Di Virgilio F, Boeynaems JM, Robson SC. Extracellular nucleotides as negative modulators of immunity. Curr Opin Pharmacol. 2009; 9(4): 507-513. doi:10.1016/j.coph.2009.06.021
  16. Gao L, Dong L, Whitlock JP Jr. A novel response to dioxin. Induction of ecto-ATPase gene expression. J Biol Chem. 1998 Jun 19; 273(25): 15358-65. https://doi.org/10.1074/jbc.273.25.15358
  17. Gandhi R, Kumar D, Burns EJ, Nadeau M, Dake B, Laroni A, et al. Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3(+) regulatory T cells. Nat Immunol. 2010 Sep; 11(9): 846-53. https://doi.org/10.1038/ni.1915
  18. Mascanfroni ID, Yeste A, Vieira SM, Burns EJ, Patel B, Sloma I, et al. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39. Nat Immunol. 2013 Oct; 14(10): 1054-63. https://doi.org/10.1038/ni.2695
  19. Hart ML, Gorzolla IC, Schittenhelm J, Robson SC, Eltzschig HK. SP1-dependent induction of CD39 facilitates hepatic ischemic preconditioning. J Immunol. 2010; 184(7): 4017-4024. doi:10.4049/jimmunol.0901851
  20. Hart ML, Grenz A, Gorzolla IC, Schittenhelm J, Dalton JH, Eltzschig HK. Hypoxia-inducible factor-1α-dependent protection from intestinal ischemia/reperfusion injury involves ecto-5'-nucleotidase (CD73) and the A2B adenosine receptor. J Immunol. 2011 Apr 1; 186(7):4367-74. https://doi.org/10.4049/jimmunol.0903617
  21. Eckle T, Hartmann K, Bonney S, Reithel S, Mittelbronn M, Walker LA, et al. Adora2b-elicited Per2 stabilization promotes a HIF-dependent metabolic switch crucial for myocardial adaptation to ischemia. Nat Med. 2012 Apr 15; 18(5): 774-82. https://doi.org/10.1038/nm.2728
  22. Stein AC, Gaetano JN, Jacobs J, Kunnavakkam R, Bissonnette M, Pekow J. Northern Latitude but Not Season Is Associated with Increased Rates of Hospitalizations Related to Inflammatory Bowel Disease: Results of a Multi-Year Analysis of a National Cohort. PLoS One. 2016; 11(8): e0161523. https://doi.org/10.1371/journal.pone.0161523
  23. Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, et al. Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature. 2008 May 1; 453(7191): 65-71. https://doi.org/10.1038/nature06880
  24. Goettel JA, Gandhi R, Kenison JE, Yeste A, Murugaiyan G, Sambanthamoorthy S, et al. AHR Activation Is Protective against Colitis Driven by T Cells in Humanized Mice. Cell Rep. 2016 Oct 25; 17(5): 1318-1329. https://doi.org/10.1016/j.celrep.2016.09.082
  25. Naganuma M, Wiznerowicz EB, Lappas CM, Linden J, Worthington MT, Ernst PB. Cutting edge: Critical role for A2A adenosine receptors in the T cell-mediated regulation of colitis. J Immunol. 2006 Sep 1; 177(5): 2765-9. https://doi.org/10.4049/jimmunol.177.5.2765
  26. Frick JS, MacManus CF, Scully M, Glover LE, Eltzschig HK, Colgan SP. Contribution of adenosine A2B receptors to inflammatory parameters of experimental colitis. J Immunol. 2009; 182(8): 4957-4964. doi:10.4049/jimmunol.0801324
  27. Doherty GA, Bai A, Hanidziar D. CD73 is a phenotypic marker of effector memory Th17 cells in inflammatory bowel disease. Eur J Immunol. 2012; 42(11): 3062-3072. https://doi.org/10.1002/eji.201242623
  28. Friedman DJ, Künzli BM, A-Rahim YI, Sevigny J, Berberat PO, Enjyoji K, et al. From the Cover: CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease. Proc Natl Acad Sci USA. 2009 Sep 29; 106(39): 16788-93. https://doi.org/10.1073/pnas.0902869106
  29. Mangino M, Roederer M, Beddall M. Innate and adaptive immune traits are differentially affected by genetic and environmental factors. Nat Commun. 2017; 8: 13850. doi: 10.1038/ncomms13850
  30. Mitsuhashi S, Feldbrügge L, Csizmadia E, Mitsuhashi M, Robson SC, Moss AC. Luminal Extracellular Vesicles (EVs) in Inflammatory Bowel Disease (IBD) Exhibit Proinflammatory Effects on Epithelial Cells and Macrophages. Inflamm Bowel Dis. 2016 Jul; 22(7): 1587-95. https://doi.org/10.1097/MIB.0000000000000840
  31. Jiang ZG, Wu Y, Csizmadia E, Feldbrügge L, Enjyoji K, Tigges J, et al. Characterization of circulating microparticle-associated CD39 family ecto-nucleotidases in human plasma. Purinergic Signal. 2014 Dec; 10(4): 611-8. https://doi.org/10.1007/s11302-014-9423-6
  32. Banz Y, Beldi G, Wu Y, Atkinson B, Usheva A, Robson SC. CD39 is incorporated into plasma microparticles where it maintains functional properties and impacts endothelial activation. Br J Haematol. 2008 Aug; 142(4): 627-37. https://doi.org/10.1111/j.1365-2141.2008.07230.x
  33. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell. 2015 Oct 8; 163(2): 367-80. https://doi.org/10.1016/j.cell.2015.08.058
  34. Baron L, Gombault A, Fanny M. The NLRP3 inflammasome is activated by nanoparticles through ATP, ADP and adenosine. Cell Death Dis. 2015; 6: e1629. doi: 10.1038/cddis.2014.576
  35. Ouyang X, Ghani A, Malik A, Wilder T, Colegio OR, Flavell RA, et al. Adenosine is required for sustained inflammasome activation via the A₂A receptor and the HIF-1α pathway. Nat Commun. 2013; 4: 2909. https://doi.org/10.1038/ncomms3909
  36. Wang Y, Telesford KM, Ochoa-Repáraz J, Haque-Begum S, Christy M, Kasper EJ, et al. An intestinal commensal symbiosis factor controls neuroinflammation via TLR2-mediated CD39 signalling. Nat Commun. 2014 Jul 21; 5:4432. https://doi.org/10.1038/ncomms5432
  37. Sansom FM, Robson SC, Hartland EL. Possible effects of microbial ecto-nucleoside triphosphate diphosphohydrolases on host-pathogen interactions. Microbiol Mol Biol Rev. 2008 Dec; 72(4): 765-81. https://doi.org/10.1128/MMBR.00013-08
  38. Vaughn BP, Vatanen T, Allegretti JR, Bai A, Xavier RJ, Korzenik J, et al. Increased Intestinal Microbial Diversity Following Fecal Microbiota Transplant for Active Crohn's Disease. Inflamm Bowel Dis. 2016 Sep; 22(9): 2182-90. https://doi.org/10.1097/MIB.0000000000000893
  39. Chiaro TR, Soto R, Zac Stephens W. A member of the gut mycobiota modulates host purine metabolism exacerbating colitis in mice [published correction appears in Sci Transl Med. 2017; 9(380): eaaf9044. https://doi.org/10.1126/scitranslmed.aaf9044
  40. Sivignon A, de Vallée A, Barnich N, Denizot J, Darcha C, Pignède G, et al. Saccharomyces cerevisiae CNCM I-3856 prevents colitis induced by AIEC bacteria in the transgenic mouse model mimicking Crohn's disease. Inflamm Bowel Dis. 2015 Feb; 21(2): 276-86. https://doi.org/10.1097/MIB.0000000000000280
  41. Lavoie EG, Gulbransen BD, Martín-Satué M, Aliagas E, Sharkey KA, Sévigny J. Ectonucleotidases in the digestive system: focus on NTPDase3 localization. Am J Physiol Gastrointest Liver Physiol. 2011 Apr; 300(4): G608-20. https://doi.org/10.1152/ajpgi.00207.2010
  42. Kusu T, Kayama H, Kinoshita M, Jeon SG, Ueda Y, Goto Y, et al. Ecto-nucleoside triphosphate diphosphohydrolase 7 controls Th17 cell responses through regulation of luminal ATP in the small intestine. J Immunol. 2013 Jan 15; 190(2): 774-83. https://doi.org/10.4049/jimmunol.1103067
  43. Zimmermann H, Zebisch M, Sträter N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal. 2012; 8(3): 437-502. https://doi.org/10.1007/s11302-012-9309-4
  44. Heine P, Braun N, Heilbronn A, Zimmermann H. Functional characterization of rat ecto-ATPase and ecto-ATP diphosphohydrolase after heterologous expression in CHO cells. Eur J Biochem. 1999 May; 262(1): 102-7. https://doi.org/10.1046/j.1432-1327.1999.00347.x
  45. Shi JD, Kukar T, Wang CY, Li QZ, Cruz PE, Davoodi-Semiromi A, et al. Molecular cloning and characterization of a novel mammalian endo-apyrase (LALP1). J Biol Chem. 2001 May 18; 276(20): 17474-8. https://doi.org/10.1074/jbc.M011569200
  46. Wink MR, Braganhol E, Tamajusuku AS, Lenz G, Zerbini LF, Libermann TA, et al. Nucleoside triphosphate diphosphohydrolase-2 (NTPDase2/CD39L1) is the dominant ectonucleotidase expressed by rat astrocytes. Neuroscience. 2006; 138(2): 421-32. https://doi.org/10.1016/j.neuroscience.2005.11.039
  47. Gallego D, Gil V, Martínez-Cutillas M, Mañé N, Martín MT, Jiménez M. Purinergic neuromuscular transmission is absent in the colon of P2Y(1) knocked out mice. J Physiol. 2012; 590(8): 1943-1956. https://doi.org/10.1113/jphysiol.2011.224345
  48. Gombault A, Baron L, Couillin I. ATP release and purinergic signaling in NLRP3 inflammasome activation. Front Immunol. 2013 Jan 8; 3: 414. https://doi.org/10.3389/fimmu.2012.00414
  49. Fields RD, Stevens B. ATP: an extracellular signaling molecule between neurons and glia. Trends Neurosci. 2000 Dec; 23(12): 625-33. https://doi.org/10.1016/s0166-2236(00)01674-x
  50. Gabanyi I, Muller PA, Feighery L, Oliveira TY, Costa-Pinto FA, Mucida D. Neuro-immune Interactions Drive Tissue Programming in Intestinal Macrophages. Cell. 2016 Jan 28; 164(3): 378-91. https://doi.org/10.1016/j.cell.2015.12.023
  51. Kusu T, Kayama H, Kinoshita M, Jeon SG, Ueda Y, Goto Y, et al. Ecto-nucleoside triphosphate diphosphohydrolase 7 controls Th17 cell responses through regulation of luminal ATP in the small intestine. J Immunol. 2013 Jan 15; 190(2): 774-83. https://doi.org/10.4049/jimmunol.1103067