Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1344 2022-07-24 14:46:49 |
2 format change Meta information modification 1344 2022-07-25 02:47:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Vieujean, S.;  Caron, B.;  Haghnejad, V.;  Jouzeau, J.;  Netter, P.;  Heba, A.;  Ndiaye, N.C.;  Moulin, D.;  Barreto, G.;  Danese, S.; et al. Epigenetics in Inflammatory Bowel Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/25468 (accessed on 02 July 2024).
Vieujean S,  Caron B,  Haghnejad V,  Jouzeau J,  Netter P,  Heba A, et al. Epigenetics in Inflammatory Bowel Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/25468. Accessed July 02, 2024.
Vieujean, Sophie, Bénédicte Caron, Vincent Haghnejad, Jean-Yves Jouzeau, Patrick Netter, Anne-Charlotte Heba, Ndeye Coumba Ndiaye, David Moulin, Guillermo Barreto, Silvio Danese, et al. "Epigenetics in Inflammatory Bowel Disease" Encyclopedia, https://encyclopedia.pub/entry/25468 (accessed July 02, 2024).
Vieujean, S.,  Caron, B.,  Haghnejad, V.,  Jouzeau, J.,  Netter, P.,  Heba, A.,  Ndiaye, N.C.,  Moulin, D.,  Barreto, G.,  Danese, S., & Peyrin-Biroulet, L. (2022, July 24). Epigenetics in Inflammatory Bowel Disease. In Encyclopedia. https://encyclopedia.pub/entry/25468
Vieujean, Sophie, et al. "Epigenetics in Inflammatory Bowel Disease." Encyclopedia. Web. 24 July, 2022.
Epigenetics in Inflammatory Bowel Disease
Edit

Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract that encompass two main phenotypes, namely Crohn’s disease and ulcerative colitis. These conditions occur in genetically predisposed individuals in response to environmental factors. Epigenetics, acting by DNA methylation, post-translational histones modifications or by non-coding RNAs, could explain how the exposome (or all environmental influences over the life course, from conception to death) could influence the gene expression to contribute to intestinal inflammation.

inflammatory bowel disease epigenetics exposome

1. Introduction

Inflammatory bowel diseases (IBD) are chronic relapsing-remitting inflammatory disorders of the gastrointestinal tract encompassing two main phenotypes: Crohn’s disease (CD) and ulcerative colitis (UC). The pathogenesis of IBD is not fully understood to date, but the most commonly accepted hypothesis is an inappropriate gut mucosal immune response towards the constituents of the gut microbiota, which cross an impaired epithelial barrier, in genetically predisposed individuals and under the influence of environmental factors [1]. Epidemiological studies (such as those carried out on monozygotic twins [2] and immigrants [3]), as well as the increase over time of the CD and UC incidence and prevalence (while the human gene pool is the same as before) [4], are all arguments that emphasize the importance of environmental factors in the occurrence of these inflammatory diseases. Epigenetics is a branch of life science that studies mechanisms regulating DNA-dependent processes (e.g., transcription, replication, recombination, repair, etc.) without primarily involving the nucleotide sequence of the DNA but, rather, the structure of how DNA is packed in the cell nucleus (chromatin structure), which can be inherited by daughter cells after cell division. Epigenetic mechanisms, including DNA methylation, post-translational histones modifications and non-coding ribonucleic acids (ncRNAs) [5], [6], regulating gene expression provide plausible explanations for the influence of the environment on gene expression profiles that favor intestinal inflammation [7]. Supporting this line of ideas, DNA methylation profiles observed in older monozygous twins with different environmental histories shows that epigenetic imprinting occurs mainly during crucial periods of development, whereas epigenomic changes can also occur day after day and accumulate over time in response to the exposome [8][9][10].
The term exposome has been proposed to encompass all environmental influences over the life course, from conception to death, that may influence disease emergence and clinical outcomes [11][12]. External environmental factors influencing the occurrence of IBD include the maternal lifestyle and in utero events [13], breastfeeding [14], diet [7], smoking habits [15][16], drugs [16][17][18], physical activity [16], stress [19], appendicectomy [16], vitamin D/UV exposure [16], infections [20] and hygiene [21]. While it is possible that these different factors directly induce epigenetic changes in the host, it is also possible that they influence the microbiome, an internal component of the exposome, and contribute to the occurrence of IBD through the exposome–microbiome–epigenome axis [22].

2. Epigenetics in IBD

Epigenetic mechanisms of gene expressions are involved in the intestinal epithelium homeostasis and in the development and differentiation of the immune cells, as well as in the modulation of responses generated by the immune system to defend against potential pathogens [23]. These epigenetic changes are reversible [24]. The genomic DNA in the eukaryotic cell nucleus is organized into chromatin. Chromatin consists of nucleic acids (genomic DNA and different types of RNAs); histone proteins (H2A, H2B, H3, H4 and H1) and non-histone chromatin-associated proteins [5][25][26]. Nucleosomes constitute the functional and structural units of chromatin. A nucleosome is built by around 146 bp of genomic DNA surrounding a histone octamer, which consists of two H2A–H2B dimers and one (H3–H4)2 tetramer [27][28]. In other words, chromatin is the physiological template for all DNA-dependent biological processes, including transcription. This fact increases the complexity of transcription regulation, since it implies that the chromatin structure has to be dynamic to grant or block access of transcription regulators to their respective binding elements on the DNA and to the transcription machinery to the genomic information in the nucleotide sequence. The epigenetic mechanisms of transcriptional regulation involve DNA methylation, histone modifications, nucleosome remodeling, interaction with the nuclear matrix and regulation via long non-coding RNAs (lncRNAs) and micro RNAs (miR) [26][29][30][31]. These mechanisms of transcription regulation establish cell-specific, heritable patterns of differential gene expression and silencing from the same genome and allow the cells to change these gene expression signatures in response to stimuli, such as changing conditions due to their environment [32][33].
DNA methylation in eukaryotes refers to the covalent transfer of a methyl group (-CH3) to the carbon atom at position 5 of cytosine forming 5-methylcytosine (5mC), most frequently at the dinucleotide sequence CG (mCG) [31][34][35][36][37]. DNA regions that are ≥200 bp long and show a CG:GC ratio ≥ 0.6 are defined as a CpG island [38]. The presence of DNA methylation prevents transcription factors from reaching gene promoters and generally leads to gene silencing [39][40]. DNA methylation in eukaryotes is catalyzed by DNA methyltransferases (DNMTs): DNMT1, which maintains DNA methylation patterns (during DNA replication and cell division), and DNMT3A/3B, which are responsible for de novo methylating DNMTs (during development or differentiation) [41][42][43]. These enzymes transfer methyl groups from S-adenosyl-L-methionine (SAM) to the cytosine residues in DNA [44]. On the contrary, DNA demethylation is mediated by the ten-eleven translocation (TET) enzymes, which add a hydroxyl group onto the methyl group of 5mC to form 5hmC (5-hydroxymethyl cytosine) [45]. Compared to healthy subjects, IBD patients show DNA methylation changes both at the cell (mainly immune) level and at the tissue level [46][47][48][49][50][51][52]. These changes also differ between UC and CD patients and involve several loci responsible for the regulation of immune responses [46][47][48][49][50][51][52].
Another epigenetic mechanism of transcriptional regulation involves post-translational modifications of histone proteins (further referred to as histone modifications). Histone proteins (H1, H2A, H2B H3 and H4) are relatively small and basic proteins that are abundant in the cell nucleus and are an essential part of the nucleosome, as described above. Due to structural characteristics of the nucleosome, histone proteins can undergo post-translational modifications at their N-terminal tails, which include acetylation, methylation, phosphorylation, ubiquitination and sumoylation, among others [53][54][55][56]. While DNA methylation is relatively stable in somatic cells, histone modifications are more diverse and dynamic, changing rapidly during the course of the cell cycle [6][30][53][54]. Acetylation at specific amino acids of histones (e.g., histone 3 lysine 9 acetylation; H3K9Ac) is generally associated with active chromatin and is mediated by histone acetyltransferases (HAT) and removed by histone deacetylases (HDAC). Histone methylation also occurs at specific amino acids of histone proteins and can be associated with both the repression (e.g., H3 lysine 27 trimethylation; H3K27me3) and activation (e.g., H3 lysine 4 trimethylation; H3K4me3) of gene expressions. There is a variety of enzymes mediating histone methylation (histone methyltransferases; HMT) and histone demethylation [57][58]. Similarly, the reactions leading to other histone modifications are catalyzed by a broad spectrum of enzymes in a regulated manner. Several environmental agents induce changes in histone modifications, thereby leading to changes in gene expression signatures.
In addition to these mechanisms, epigenetic regulation can also involve ncRNA, which are RNAs not translated into proteins, including miRs and lncRNAs. If miRs have a length of 18–25 nucleotides, lncRNAs are over 200 bases long [59]. These nucleic acid molecules can regulate gene expressions by interfering with messenger RNA (mRNA) translations by degrading them or through interactions with protein complexes involved in the regulation of gene expression [59][60]. The ncRNAs are differentially expressed between the control and IBD subjects, and there is also a difference in expression between CD and UC patients [61][62][63]. In IBD, miRs are involved in the regulation of the intestinal mucosal barrier, T-cell differentiation, the Th17 signaling pathway and autophagy [63]. In UC patients, miR-21, miR-16 and let-7 expressions are significantly increased in inflamed mucosa, while miR-192, miR-375 and miR-422b expressions are significantly reduced [61]. In CD patients, miR-23b, miR-106 and miR-191 are significantly increased in the inflamed mucosa, while miR-19b and miR-629 expressions are significantly decreased [61].
All these epigenetic mechanisms contribute to the development, progression and maintenance of IBD. They are usually triggered by a range of environmental factors. Some authors have mentioned three critical periods during which the environment can favor the onset of the disease: (1) during the prenatal period (in response to the maternal lifestyle), (2) in the early postnatal period (during gut microbiota colonization) and (3) just before the disease onset [64].

References

  1. Danese, S.; Fiocchi, C. Etiopathogenesis of inflammatory bowel diseases. World J. Gastroenterol. 2006, 12, 4807–4812.
  2. Park, J.H.; Wacholder, S.; Gail, M.H.; Peters, U.; Jacobs, K.B.; Chanock, S.J.; Chatterjee, N. Estimation of effect size distribution from genome-wide association studies and implications for future discoveries. Nat. Genet. 2010, 42, 570–575.
  3. Shanahan, F. The gut microbiota-A clinical perspective on lessons learned. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 609–614.
  4. Bernstein, C.N.; Shanahan, F. Disorders of a modern lifestyle: Reconciling the epidemiology of inflammatory bowel diseases. Gut 2008, 57, 1185–1191.
  5. Rubio, K.; Dobersch, S.; Barreto, G. Functional interactions between scaffold proteins, noncoding RNAs, and genome loci induce liquid-liquid phase separation as organizing principle for 3-dimensional nuclear architecture: Implications in cancer. FASEB J. 2019, 33, 5814–5822.
  6. Dobersch, S.; Rubio, K.; Singh, I.; Günther, S.; Graumann, J.; Cordero, J.; Castillo-Negrete, R.; Huynh, M.B.; Mehta, A.; Braubach, P.; et al. Positioning of nucleosomes containing γ-H2AX precedes active DNA demethylation and transcription initiation. Nat. Commun. 2021, 12, 1072.
  7. Rogler, G.; Vavricka, S. Exposome in IBD: Recent insights in environmental factors that influence the onset and course of IBD. Inflamm. Bowel Dis. 2015, 21, 400–408.
  8. Fraga, M.F.; Ballestar, E.; Paz, M.F.; Ropero, S.; Setien, F.; Ballestar, M.L.; Heine-Suñer, D.; Cigudosa, J.C.; Urioste, M.; Benitez, J.; et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 2005, 102, 10604–10609.
  9. Christensen, B.C.; Houseman, E.A.; Marsit, C.; Zheng, S.; Wrensch, M.R.; Wiemels, J.L.; Nelson, H.; Karagas, M.R.; Padbury, J.F.; Bueno, R.; et al. Aging and environmental exposures alter tissue-specific DNA methylation dependent upon CPG island context. PLoS Genet. 2009, 5, e1000602.
  10. Szarc Vel Szic, K.; Declerck, K.; Vidaković, M.; Vanden Berghe, W. From inflammaging to healthy aging by dietary lifestyle choices: Is epigenetics the key to personalized nutrition? Clin. Epigenetics 2015, 7, 33.
  11. Wild, C.P. Complementing the genome with an “exposome”: The outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1847–1850.
  12. Rappaport, S.M. Implications of the exposome for exposure science. J. Expo. Sci. Environ. Epidemiol. 2011, 21, 5–9.
  13. Waterland, R.A.; Michels, K.B. Epigenetic epidemiology of the developmental origins hypothesis. Annu. Rev. Nutr. 2007, 27, 363–388.
  14. Barclay, A.R.; Russell, R.K.; Wilson, M.L.; Gilmour, W.H.; Satsangi, J.; Wilson, D.C. Systematic Review: The Role of Breastfeeding in the Development of Pediatric Inflammatory Bowel Disease. J. Pediatr. 2009, 155, 421–426.
  15. Tysk, C.; Lindberg, E.; Jarnerot, G.; Floderus-Myrhed, B. Ulcerative colitis and Crohn’s disease in an unselected population of monozygotic and dizygotic twins. A study of heritability and the influence of smoking. Gut 1988, 29, 990–996.
  16. Piovani, D.; Danese, S.; Peyrin-Biroulet, L.; Nikolopoulos, G.K.; Lytras, T.; Bonovas, S. Environmental Risk Factors for Inflammatory Bowel Diseases: An Umbrella Review of Meta-Analyses. Gastroenterology 2019, 157, 647–659.e4.
  17. Boyko, E.J.; Theis, M.K.; Vaughan, T.L.; Nicol-blades, B. Increased risk of inflammatory bowel disease associated with oral contraceptive use. Am. J. Epidemiol. 1994, 140, 268–278.
  18. Shaw, S.Y.; Blanchard, J.F.; Bernstein, C.N. Association between the use of antibiotics and new diagnoses of Crohn’s disease and ulcerative colitis. Am. J. Gastroenterol. 2011, 106, 2133–2142.
  19. Bernstein, C.N. Psychological Stress and Depression: Risk Factors for IBD? Dig. Dis. 2016, 34, 58–63.
  20. Singh, S.; Graff, L.A.; Bernstein, C.N. Do NSAIDs, antibiotics, infections, or stress trigger flares in IBD? Am. J. Gastroenterol. 2009, 104, 1298–1313.
  21. DeFilippis, E.M.; Longman, R.; Harbus, M.; Dannenberg, K.; Scherl, E.J. Crohn’s Disease: Evolution, Epigenetics, and the Emerging Role of Microbiome-Targeted Therapies. Curr. Gastroenterol. Rep. 2016, 18, 13.
  22. Fiocchi, C. Inflammatory bowel disease pathogenesis: Where are we? J. Gastroenterol. Hepatol. 2015, 30 (Suppl. 1), 12–18.
  23. Ray, G.; Longworth, M.S. Epigenetics, DNA Organization, and Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2019, 25, 235–247.
  24. Bernstein, B.E.; Meissner, A.; Lander, E.S. The Mammalian Epigenome. Cell 2007, 128, 669–681.
  25. Zhang, Y. Recent progress in the epigenetics and chromatin field. Cell Res. 2011, 21, 373–374.
  26. Ozturk, N.; Singh, I.; Mehta, A.; Braun, T.; Barreto, G. HMGA proteins as modulators of chromatin structure during transcriptional activation. Front. Cell Dev. Biol. 2014, 2, 5.
  27. Richmond, T.J.; Finch, J.T.; Rushton, B.; Rhodes, D.; Klug, A. Structure of the nucleosome core particle at 7 resolution. Nature 1984, 311, 532–537.
  28. Bentley, G.A.; Lewit-Bentley, A.; Finch, J.T.; Podjarny, A.D.; Roth, M. Crystal structure of the nucleosome core particle at 16 Å resolution. J. Mol. Biol. 1984, 176, 55–75.
  29. Singh, I.; Contreras, A.; Cordero, J.; Rubio, K.; Dobersch, S.; Günther, S.; Jeratsch, S.; Mehta, A.; Krüger, M.; Graumann, J.; et al. MiCEE is a ncRNA-protein complex that mediates epigenetic silencing and nucleolar organization. Nat. Genet. 2018, 50, 990–1001.
  30. Cedar, H.; Bergman, Y. Linking DNA methylation and histone modification: Patterns and paradigms. Nat. Rev. Genet. 2009, 10, 295–304.
  31. Deaton, A.M.; Bird, A. CpG islands and the regulation of transcription. Genes Dev. 2011, 25, 1010–1022.
  32. Skinner, M.K. Environmental epigenetics and a unified theory of the molecular aspects of evolution: A neo-Lamarckian concept that facilitates neo-Darwinian evolution. Genome Biol. Evol. 2015, 7, 1296–1302.
  33. Dobersch, S.; Rubio, K.; Barreto, G. Pioneer Factors and Architectural Proteins Mediating Embryonic Expression Signatures in Cancer. Trends Mol. Med. 2019, 25, 287–302.
  34. Esteller, M. Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet. 2007, 8, 286–298.
  35. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002, 16, 6–21.
  36. Schübeler, D. Function and information content of DNA methylation. Nature 2015, 517, 321–326.
  37. Bird, A.P.; Wolffe, A.P. Methylation-induced repression-belts, braces, and chromatin. Cell 1999, 99, 451–454.
  38. Gardiner-Garden, M.; Frommer, M. CpG Islands in vertebrate genomes. J. Mol. Biol. 1987, 196, 261–282.
  39. Lim, D.H.K.; Maher, E.R. DNA methylation: A form of epigenetic control of gene expression. Obstet. Gynaecol. 2010, 12, 37–42.
  40. Yang, I.V.; Schwartz, D.A. Epigenetic control of gene expression in the lung. Am. J. Respir. Crit. Care Med. 2011, 183, 1295–1301.
  41. Reik, W.; Dean, W.; Walter, J. Epigenetic Reprogramming in Mammalian Development. Science 2001, 293, 1089–1093.
  42. Jones, P.A.; Liang, G. Rethinking how DNA methylation patterns are maintained. Nat. Rev. Genet. 2009, 10, 805–811.
  43. Law, J.A.; Jacobsen, S.E. Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nat. Rev. Genet. 2010, 11, 204–220.
  44. Jin, B.; Robertson, K.D. DNA methyltransferases, DNA damage repair, and cancer. Adv. Exp. Med. Biol. 2013, 754, 3–29.
  45. Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38.
  46. Nimmo, E.R.; Prendergast, J.G.; Aldhous, M.C.; Kennedy, N.A.; Henderson, P.; Drummond, H.E.; Ramsahoye, B.H.; Wilson, D.C.; Semple, C.A.; Satsangi, J. Genome-wide methylation profiling in Crohn’s disease identifies altered epigenetic regulation of key host defense mechanisms including the Th17 pathway. Inflamm. Bowel Dis. 2012, 18, 889–899.
  47. Häsler, R.; Feng, Z.; Bäckdahl, L.; Spehlmann, M.E.; Franke, A.; Teschendorff, A.; Rakyan, V.K.; Down, T.A.; Wilson, G.A.; Feber, A.; et al. A functional methylome map of ulcerative colitis. Genome Res. 2012, 22, 2130–2137.
  48. Cooke, J.; Zhang, H.; Greger, L.; Silva, A.L.; Massey, D.; Dawson, C. Mucosal genome-wide methylation changes in inflammatory bowel disease. Inflamm. Bowel Dis. 2012, 18, 2128–2137.
  49. Saito, S.; Kato, J.; Hiraoka, S.; Horii, J.; Suzuki, H.; Higashi, R.; Kaji, E.; Kondo, Y.; Yamamoto, K. DNA methylation of colon mucosa in ulcerative colitis patients: Correlation with inflammatory status. Inflamm. Bowel Dis. 2011, 17, 1955–1965.
  50. Harris, R.; Nagy-Szakal, R.; Mir, S.A.V.; Frank, E.; Szigeti, R.; Kaplan, J.L.; Bronsky, J.; Opekun, A.; Ferry, G.D.; Winter, H.; et al. DNA methylation-associated colonic mucosal immune and defense responses in treatmentnaïve pediatric ulcerative colitis. Epigenetics 2014, 9, 1131–1137.
  51. Koizumi, K.; Alonso, S.; Miyaki, Y.; Okada, S.; Ogura, H.; Shiiya, N.; Konishi, F.; Taya, T.; Perucho, M.; Suzuki, K. Array-based identification of common DNA methylation alterations in ulcerative colitis. Int. J. Oncol. 2012, 40, 983–994.
  52. Lin, Z.; Hegarty, J.P.; Yu, W.; Cappel, J.A.; Chen, X.; Faber, P.W.; Wang, Y.; Poritz, L.S.; Fan, J.-B.; Koltun, W.A. Identification of disease-associated DNA methylation in B cells from Crohn’s disease and ulcerative colitis patients. Dig. Dis. Sci. 2012, 57, 3145–3153.
  53. Wang, Y.; Fischle, W.; Cheung, W.; Jacobs, S.; Khorasanizadeh, S.; Allis, C.D. Beyond the double helix: Writing and reading the histone code. Novartis Found. Symp. 2004, 259, 3–17; discussion 17–21, 163–169.
  54. Singh, I.; Ozturk, N.; Cordero, J.; Mehta, A.; Hasan, D.; Cosentino, C.; Sebastian, C.; Krüger, M.; Looso, M.; Carraro, G.; et al. High mobility group protein-mediated transcription requires DNA damage marker γ-H2AX. Cell Res. 2015, 25, 837–850.
  55. Zhang, M.; Zhao, J.; Lv, Y.; Wang, W.; Feng, C.; Zou, W.; Su, L.; Jiao, J. Histone Variants and Histone Modifications in Neurogenesis. Trends Cell Biol. 2020, 30, 869–880.
  56. Nakayama, T.; Takami, Y. Participation of histones and histone-modifying enzymes in cell functions through alterations in chromatin structure. J. Biochem. 2001, 129, 491–499.
  57. Morgan, M.A.J.; Shilatifard, A. Reevaluating the roles of histone-modifying enzymes and their associated chromatin modifications in transcriptional regulation. Nat. Genet. 2020, 52, 1271–1281.
  58. Stillman, B. Histone Modifications: Insights into Their Influence on Gene Expression. Cell 2018, 175, 6–9.
  59. Nie, L.; Wu, H.J.; Hsu, J.M.; Chang, S.S.; LaBaff, A.M.; Li, C.W.; Wang, Y.; Hsu, J.L.; Hung, M.C. Long non-coding RNAs: Versatile master regulators of gene expression and crucial players in cancer. Am. J. Transl. Res. 2012, 4, 127–150.
  60. Aalto, A.P.; Pasquinelli, A.E. Small non-coding RNAs mount a silent revolution in gene expression. Curr. Opin. Cell Biol. 2012, 24, 333–340.
  61. Pekow, J.R.; Kwon, J.H. MicroRNAs in inflammatory bowel disease. Inflamm. Bowel Dis. 2012, 18, 187–193.
  62. Coskun, M.; Bjerrum, J.T.; Seidelin, J.B.; Nielsen, O.H. MicroRNAs in inflammatory bowel disease-pathogenesis, diagnostics and therapeutics. World J. Gastroenterol. 2012, 18, 4629–4634.
  63. Annese, V. Genetics and epigenetics of IBD. Pharmacol. Res. 2020, 159, 104892.
  64. Kellermayer, R.; Zilbauer, M. The Gut Microbiome and the Triple Environmental Hit Concept of Inflammatory Bowel Disease Pathogenesis. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 589–595.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , ,
View Times: 386
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 25 Jul 2022
1000/1000
Video Production Service