Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update
Abstract
:1. Introduction
2. Etiology—Genetics Factors
2.1. Genetic Factors and the Microbiome in Inflammatory Bowel Disease
2.1.1. NOD2 (Nucleotide Binding Oligomerization Domain Containing 2)
2.1.2. ATG16L1 (Autophagy-Related 16-like 1)
2.1.3. CARD9 (Caspase Recruitment Domain 9)
2.1.4. CLEC7A (C-Type Lectin Domain Containing 7A)
2.2. Inheritance of the Microbiota
2.3. Genetic Factors and the Immune System in IBD
2.3.1. NOD2
2.3.2. ATG16L1 T300A
2.3.3. CARD9
2.3.4. IL23R (Interleukin 23 Receptor)
2.3.5. Interleukin 10 (IL-10)
2.3.6. TNFSF15/TL1A (Tumor Necrosis Factor Superfamily Member 15)
2.4. Practical Implementation of Genetics in IBD
2.4.1. Different Degrees of Relatedness in IBD
2.4.2. Inflammatory Bowel Disease in Twins
2.5. Epigenetics
2.5.1. DNA Methylation
2.5.2. Histone Modifications
2.5.3. Expression of Noncoding RNAs
2.6. Pharmacogenetics in Inflammatory Bowel Disease
3. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Dudley, M.; Kojinkov, M.; Baraga, D. ECCO-EFCCA Patient Guidelines on Crohn’s Disease (CD), European Crohn’s and Colitis Organisation: 2016. Available online: https://efcca.org/projects/ecco-efcca-patient-guidelines, (accessed on 13 September 2022).
- Gomollón, F.; Dignass, A.; Annese, V.; Tilg, H.; Van Assche, G.; Lindsay, J.O.; Peyrin-Biroulet, L.; Cullen, G.J.; Daperno, M.; Kucharzik, T.; et al. 3rd European Evidence-based Consensus on the Diagnosis and Management of Crohn’s Disease 2016: Part 1: Diagnosis and Medical Management. J. Crohn’s Colitis 2017, 11, 3–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Windsor, J.W.; Kaplan, G.G. Evolving Epidemiology of IBD. Curr. Gastroenterol. Rep. 2019, 21, 40. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, G.G.; Windsor, J.W. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 56–66. [Google Scholar] [CrossRef] [PubMed]
- Mak, W.Y.; Zhao, M.; Ng, S.C.; Burisch, J. The epidemiology of inflammatory bowel disease: East meets west. J. Gastroenterol. Hepatol. 2020, 35, 380–389. [Google Scholar] [CrossRef] [PubMed]
- Mak, J.W.Y.; Ho, C.L.T.; Wong, K.; Cheng, T.Y.; Yip, T.C.F.; Leung, W.K.; Li, M.; Lo, F.H.; Ng, K.M.; Sze, S.F.; et al. Epidemiology and Natural History of Elderly-onset Inflammatory Bowel Disease: Results From a Territory-wide Hong Kong IBD Registry. J. Crohn’s Colitis 2021, 15, 401–408. [Google Scholar] [CrossRef]
- Burisch, J.; Jess, T.; Martinato, M.; Lakatos, P.L. The burden of inflammatory bowel disease in Europe. J. Crohn’′s Colitis 2013, 7, 322–337. [Google Scholar] [CrossRef] [Green Version]
- de Souza, H.; Fiocchi, C.; Iliopoulos, D. The IBD interactome: An integrated view of aetiology, pathogenesis and therapy. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 739–749. [Google Scholar] [CrossRef]
- Cooney, R.; Baker, J.; Brain, O.; Danis, B.; Pichulik, T.; Allan, P.; Ferguson, D.J.P.; Campbell, B.J.; Jewell, D.; Simmons, A. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat. Med. 2010, 16, 90–97. [Google Scholar] [CrossRef]
- Eckmann, L.; Karin, M. NOD2 and Crohn’s Disease: Loss or Gain of Function? Immunity 2005, 22, 661–667. [Google Scholar] [CrossRef] [Green Version]
- Foerster, E.G.; Mukherjee, T.; Cabral-Fernandes, L.; Rocha, J.D.; Girardin, S.E.; Philpott, D.J. How autophagy controls the intestinal epithelial barrier. Autophagy 2022, 18, 86–103. [Google Scholar] [CrossRef]
- Lauro, M.L.; Burch, J.M.; Grimes, C.L. The effect of NOD2 on the microbiota in Crohn′s disease. Curr. Opin. Biotechnol. 2016, 40, 97–102. [Google Scholar] [CrossRef] [Green Version]
- Matsuzawa-Ishimoto, Y.; Shono, Y.; Gomez, L.E.; Hubbard-Lucey, V.M.; Cammer, M.; Neil, J.; Dewan, M.Z.; Lieberman, S.R.; Lazrak, A.; Marinis, J.M.; et al. Autophagy protein ATG16L1 prevents necroptosis in the intestinal epithelium. J. Exp. Med. 2017, 214, 3687–3705. [Google Scholar] [CrossRef]
- Pigneur, B.; Escher, J.; Elawad, M.; Lima, R.; Buderus, S.; Kierkus, J.; Guariso, G.; Canioni, D.; Lambot, K.; Talbotec, C.; et al. Phenotypic Characterization of Very Early-onset IBD Due to Mutations in the IL10, IL10 Receptor Alpha or Beta Gene. Inflamm. Bowel Dis. 2013, 19, 2820–2828. [Google Scholar] [CrossRef]
- Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef]
- Guan, Q. A Comprehensive Review and Update on the Pathogenesis of Inflammatory Bowel Disease. J. Immunol. Res. 2019, 2019, 7247238. [Google Scholar] [CrossRef] [Green Version]
- Annese, V. Genetics and epigenetics of IBD. Pharmacol. Res. 2020, 159, 104892. [Google Scholar] [CrossRef]
- Glassner, K.L.; Abraham, B.P.; Quigley, E.M. The microbiome and inflammatory bowel disease. J. Allergy Clin. Immunol. 2020, 145, 16–27. [Google Scholar] [CrossRef] [Green Version]
- Aschard, H.; Laville, V.; Tchetgen, E.T.; Knights, D.; Imhann, F.; Seksik, P.; Zaitlen, N.; Silverberg, M.S.; Cosnes, J.; Weersma, R.K.; et al. Genetic effects on the commensal microbiota in inflammatory bowel disease patients. PLOS Genet. 2019, 15, e1008018. [Google Scholar] [CrossRef] [Green Version]
- Øyri, S.F.; Műzes, G.; Sipos, F. Dysbiotic gut microbiome: A key element of Crohn’s disease. Comp. Immunol. Microbiol. Infect. Dis. 2015, 43, 36–49. [Google Scholar] [CrossRef]
- Wright, E.K.; Kamm, M.A.; Teo, S.M.; Inouye, M.; Wagner, J.; Kirkwood, C.D. Recent Advances in Characterizing the Gastrointestinal Microbiome in Crohnʼs Disease: A Systematic Review. Inflamm. Bowel Dis. 2015, 21, 1219–1228. [Google Scholar] [CrossRef]
- Quraishi, M.N.; Shaheen, W.; Oo, Y.H.; Iqbal, T.H. Immunological mechanisms underpinning faecal microbiota transplantation for the treatment of inflammatory bowel disease. Clin. Exp. Immunol. 2020, 199, 24–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gevers, D.; Kugathasan, S.; Denson, L.A.; Vázquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; et al. The Treatment-Naive Microbiome in New-Onset Crohn’s Disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mentella, M.C.; Scaldaferri, F.; Pizzoferrato, M.; Gasbarrini, A.; Miggiano, G.A.D. Nutrition, IBD and Gut Microbiota: A Review. Nutrients 2020, 12, 944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nell, S.; Suerbaum, S.; Josenhans, C. The impact of the microbiota on the pathogenesis of IBD: Lessons from mouse infection models. Nat. Rev. Genet. 2010, 8, 564–577. [Google Scholar] [CrossRef] [PubMed]
- Pinho, R.M.; Maga, E.A. DNA methylation as a regulator of intestinal gene expression. Br. J. Nutr. 2021, 126, 1611–1625. [Google Scholar] [CrossRef]
- Cohen, L.J.; Cho, J.H.; Gevers, D.; Chu, H. Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases. Gastroenterology 2019, 156, 2174–2189. [Google Scholar] [CrossRef] [Green Version]
- Lavoie, S.; Conway, K.L.; Lassen, K.G.; Jijon, H.B.; Pan, H.; Chun, E.; Michaud, M.; Lang, J.K.; Comeau, C.A.G.; Dreyfuss, J.; et al. The Crohn’s disease polymorphism, ATG16L1 T300A, alters the gut microbiota and enhances the local Th1/Th17 response. Elife 2019, 8, 39982. [Google Scholar] [CrossRef]
- Santana, P.T.; Rosas, S.L.B.; Ribeiro, B.E.; Marinho, Y.; de Souza, H.S.P. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int. J. Mol. Sci. 2022, 23(7), 3464. [Google Scholar] [CrossRef]
- Zhang, Q.; Pan, Y.; Yan, R.; Zeng, B.; Wang, H.; Zhang, X.; Li, W.; Wei, H.; Liu, Z. Commensal bacteria direct selective cargo sorting to promote symbiosis. Nat. Immunol. 2015, 16, 918–926. [Google Scholar] [CrossRef]
- Negroni, A.; Pierdomenico, M.; Cucchiara, S.; Stronati, L. NOD2 and inflammation: Current insights. J. Inflamm. Res. 2018, 11, 49–60. [Google Scholar] [CrossRef]
- Venema, W.T.U.; Voskuil, M.D.; Dijkstra, G.; Weersma, R.K.; Festen, E.A. The genetic background of inflammatory bowel disease: From correlation to causality. J. Pathol. 2017, 241, 146–158. [Google Scholar] [CrossRef]
- Kobayashi, K.S.; Chamaillard, M.; Ogura, Y.; Henegariu, O.; Inohara, N.; Nuñez, G.; Flavell, R.A. Nod2-Dependent Regulation of Innate and Adaptive Immunity in the Intestinal Tract. Science 2005, 307, 731–734. [Google Scholar] [CrossRef]
- Philpott, D.J.; Sorbara, M.T.; Robertson, S.J.; Croitoru, K.; Girardin, S.E. NOD proteins: Regulators of inflammation in health and disease. Nat. Rev. Immunol. 2014, 14, 9–23. [Google Scholar] [CrossRef]
- Ni, J.; Wu, G.D.; Albenberg, L.; Tomov, V.T. Gut microbiota and IBD: Causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 573–584. [Google Scholar] [CrossRef] [Green Version]
- Al Nabhani, Z.; Lepage, P.; Mauny, P.; Montcuquet, N.; Roy, M.; Le Roux, K.; Dussaillant, M.; Berrebi, D.; Hugot, J.-P.; Barreau, F. Nod2 Deficiency Leads to a Specific and Transmissible Mucosa-associated Microbial Dysbiosis Which Is Independent of the Mucosal Barrier Defect. J. Crohn’s Colitis 2016, 10, 1428–1436. [Google Scholar] [CrossRef] [Green Version]
- Butera, A.; Di Paola, M.; Pavarini, L.; Strati, F.; Pindo, M.; Sanchez, M.; Cavalieri, D.; Boirivant, M.; De Filippo, C. Nod2 Deficiency in mice is Associated with Microbiota Variation Favouring the Expansion of mucosal CD4+ LAP+ Regulatory Cells. Sci. Rep. 2018, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Knights, D.; Silverberg, M.S.; Weersma, R.K.; Gevers, D.; Dijkstra, G.; Huang, H.; Tyler, A.D.; Van Sommeren, S.; Imhann, F.; Stempak, J.M.; et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014, 6, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Turpin, W.; Bedrani, L.; Espin-Garcia, O.; Xu, W.; Silverberg, M.S.; Smith, M.I.; Garay, J.A.R.; Lee, S.-H.; Guttman, D.S.; Griffiths, A.; et al. Associations of NOD2 polymorphisms with Erysipelotrichaceae in stool of in healthy first degree relatives of Crohn’s disease subjects. BMC Med Genet. 2020, 21, 1–8. [Google Scholar] [CrossRef]
- Li, E.; Zhang, Y.; Tian, X.; Wang, X.; Gathungu, G.; Wolber, A.; Shiekh, S.S.; Sartor, R.B.; Davidson, N.O.; Ciorba, M.A.; et al. Influence of Crohn’s disease related polymorphisms in innate immune function on ileal microbiome. PLoS ONE 2019, 14, e0213108. [Google Scholar] [CrossRef]
- Bonder, M.J.; Kurilshikov, A.; Tigchelaar, E.F.; Mujagic, Z.; Imhann, F.; Vila, A.V.; Deelen, P.; Vatanen, T.; Schirmer, M.; Smeekens, S.P.; et al. The effect of host genetics on the gut microbiome. Nat. Genet. 2016, 48, 1407–1412. [Google Scholar] [CrossRef]
- A Kennedy, N.; A Lamb, C.; Berry, S.H.; Walker, A.W.; Mansfield, J.; Parkes, M.; Simpkins, R.; Tremelling, M.; Nutland, S.; Parkhill, J.; et al. The Impact of NOD2 Variants on Fecal Microbiota in Crohn’s Disease and Controls Without Gastrointestinal Disease. Inflamm. Bowel Dis. 2018, 24, 583–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chu, H.; Khosravi, A.; Kusumawardhani, I.P.; Kwon, A.H.K.; Vasconcelos, A.C.; Cunha, L.D.; Mayer, A.E.; Shen, Y.; Wu, W.-L.; Kambal, A.; et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 2016, 352, 1116–1120. [Google Scholar] [CrossRef] [Green Version]
- Nelson, A.; Stewart, C.J.; A Kennedy, N.; Lodge, J.K.; Tremelling, M.; Probert, C.S.; Parkes, M.; Mansfield, J.C.; Smith, D.L.; Hold, G.L.; et al. The Impact of NOD2 Genetic Variants on the Gut Mycobiota in Crohn’s Disease Patients in Remission and in Individuals Without Gastrointestinal Inflammation. J. Crohn’s Colitis 2021, 15, 800–812. [Google Scholar] [CrossRef] [PubMed]
- AL Nabhani, Z.; Dietrich, G.; Hugot, J.-P.; Barreau, F. Nod2: The intestinal gate keeper. PLOS Pathog. 2017, 13, e1006177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turpin, W.; Goethel, A.; Bedrani, L.; Croitoru, M.K. Determinants of IBD Heritability: Genes, Bugs, and More. Inflamm. Bowel Dis. 2018, 24, 1133–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khor, B.; Gardet, A.; Xavier, R.J. Genetics and pathogenesis of inflammatory bowel disease. Nature 2011, 474, 307–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bel, S.; Pendse, M.; Wang, Y.; Li, Y.; Ruhn, K.A.; Hassell, B.; Leal, T.; Winter, S.E.; Xavier, R.J.; Hooper, L.V. Paneth cells secrete lysozyme via secretory autophagy during bacterial infection of the intestine. Science 2017, 357, 1047–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Travassos, L.H.; Carneiro, L.A.M.; Ramjeet, M.; Hussey, S.; Kim, Y.-G.; Magalhães, J.G.; Yuan, L.; Soares, F.; Chea, E.; Le Bourhis, L.; et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat. Immunol. 2010, 11, 55–62. [Google Scholar] [CrossRef]
- Nguyen, H.T.T.; Lapaquette, P.; Bringer, M.-A.; Darfeuille-Michaud, A. Autophagy and Crohn’s Disease. J. Innate Immun. 2013, 5, 434–443. [Google Scholar] [CrossRef]
- Yin, H.; Wu, H.; Chen, Y.; Zhang, J.; Zheng, M.; Chen, G.; Li, L.; Lu, Q. The Therapeutic and Pathogenic Role of Autophagy in Autoimmune Diseases. Front. Immunol. 2018, 9, 1512. [Google Scholar] [CrossRef]
- Glick, D.; Barth, S.; MacLeod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef] [Green Version]
- Lassen, K.G.; Kuballa, P.; Conway, K.L.; Patel, K.K.; Becker, C.E.; Peloquin, J.M.; Villablanca, E.J.; Norman, J.M.; Liu, T.-C.; Heath, R.J.; et al. Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense. Proc. Natl. Acad. Sci. USA 2014, 111, 7741–7746. [Google Scholar] [CrossRef] [Green Version]
- Chesney, K.L.; Men, H.; Hankins, M.A.; Bryda, E.C. The Atg16l1 gene: Characterization of wild type, knock-in, and knock-out phenotypes in rats. Physiol. Genom. 2021, 53, 269–281. [Google Scholar] [CrossRef]
- Liu, H.; Gao, P.; Jia, B.; Lu, N.; Zhu, B.; Zhang, F. IBD-Associated Atg16L1T300A Polymorphism Regulates Commensal Microbiota of the Intestine. Front. Immunol. 2022, 12, 772189. [Google Scholar] [CrossRef]
- Sadabad, M.S.; Regeling, A.; de Goffau, M.C.; Blokzijl, T.; Weersma, R.K.; Penders, J.; Faber, K.N.; Harmsen, H.J.M.; Dijkstra, G. The ATG16L1–T300A allele impairs clearance of pathosymbionts in the inflamed ileal mucosa of Crohn′s disease patients. Gut 2015, 64, 1546–1552. [Google Scholar] [CrossRef] [Green Version]
- Imhann, F.; Vila, A.V.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; Van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119. [Google Scholar] [CrossRef]
- Tsianos, V.E.; Kostoulas, C.; Gazouli, M.; Frillingos, S.; Georgiou, I.; Christodoulou, D.K.; Katsanos, K.H.; Tsianos, E.V. ATG16L1 T300A polymorphism is associated with Crohn’s disease in a Northwest Greek cohort, but ECM1 T130M and G290S polymorphisms are not associated with ulcerative colitis. Ann. Gastroenterol. 2020, 33, 38–44. [Google Scholar] [CrossRef]
- Larabi, A.; Barnich, N.; Nguyen, H.T.T. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy 2020, 16, 38–51. [Google Scholar] [CrossRef] [Green Version]
- Drummond, R.A.; Franco, L.M.; Lionakis, M.S. Human CARD9: A Critical Molecule of Fungal Immune Surveillance. Front. Immunol. 2018, 9, 1836. [Google Scholar] [CrossRef] [Green Version]
- Lamas, B.; Richard, M.L.; Leducq, V.; Pham, H.-P.; Michel, M.-L.; DA Costa, G.; Bridonneau, C.; Jegou, S.; Hoffmann, T.W.; Natividad, J.M.; et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat. Med. 2016, 22, 598–605. [Google Scholar] [CrossRef]
- Luo, P.; Yang, Z.; Chen, B.; Zhong, X. The multifaceted role of CARD9 in inflammatory bowel disease. J. Cell. Mol. Med. 2020, 24, 34–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, S.; Rezende, R.M.; Moreira, T.G.; Tankou, S.K.; Cox, L.M.; Wu, M.; Song, A.; Dhang, F.H.; Wei, Z.; Costamagna, G.; et al. Oral Administration of miR-30d from Feces of MS Patients Suppresses MS-like Symptoms in Mice by Expanding Akkermansia muciniphila. Cell Host Microbe 2019, 26, 779–794.e8. [Google Scholar] [CrossRef] [PubMed]
- Lamas, B.; Michel, M.-L.; Waldschmitt, N.; Pham, H.-P.; Zacharioudaki, V.; Dupraz, L.; Delacre, M.; Natividad, J.M.; Da Costa, G.; Planchais, J.; et al. Card9 mediates susceptibility to intestinal pathogens through microbiota modulation and control of bacterial virulence. Gut 2018, 67, 1836–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Spatz, M.; Da Costa, G.; Michaudel, C.; Lapiere, A.; Danne, C.; Agus, A.; Michel, M.-L.; Netea, M.G.; Langella, P.; et al. Deletion of both Dectin-1 and Dectin-2 affects the bacterial but not fungal gut microbiota and susceptibility to colitis in mice. Microbiome 2022, 10, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Sovran, B.; Planchais, J.; Jegou, S.; Straube, M.; Lamas, B.; Natividad, J.M.; Agus, A.; Dupraz, L.; Glodt, J.; DA Costa, G.; et al. Enterobacteriaceae are essential for the modulation of colitis severity by fungi. Microbiome 2018, 6, 152. [Google Scholar] [CrossRef]
- Zuo, T.; Lu, X.-J.; Zhang, Y.; Cheung, C.P.; Lam, S.; Zhang, F.; Tang, W.; Ching, J.Y.L.; Zhao, R.; Chan, P.K.S.; et al. Gut mucosal virome alterations in ulcerative colitis. Gut 2019, 68, 1169–1179. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Mao, B.; Gu, J.; Wu, J.; Cui, S.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W.W. Blautia—A new functional genus with potential probiotic properties? Gut Microbes 2021, 13, 1–21. [Google Scholar] [CrossRef]
- Chen, L.; Wang, W.; Zhou, R.; Ng, S.C.; Li, J.; Huang, M.; Zhou, F.; Wang, X.; Shen, B.; Kamm, M.A.; et al. Characteristics of Fecal and Mucosa-Associated Microbiota in Chinese Patients With Inflammatory Bowel Disease. Medicine 2014, 93, e51. [Google Scholar] [CrossRef]
- Goodrich, J.K.; Davenport, E.R.; Beaumont, M.; Jackson, M.A.; Knight, R.; Ober, C.; Spector, T.D.; Bell, J.T.; Clark, A.G.; Ley, R.E. Genetic Determinants of the Gut Microbiome in UK Twins. Cell Host Microbe 2016, 19, 731–743. [Google Scholar] [CrossRef] [Green Version]
- Goodrich, J.K.; Davenport, E.R.; Waters, J.L.; Clark, A.G.; Ley, R.E. Cross-species comparisons of host genetic associations with the microbiome. Science 2016, 352, 532–535. [Google Scholar] [CrossRef]
- Grigg, J.B.; Sonnenberg, G.F. Host-Microbiota Interactions Shape Local and Systemic Inflammatory Diseases. J. Immunol. 2017, 198, 564–571. [Google Scholar] [CrossRef] [Green Version]
- Schirmer, M.; Garner, A.; Vlamakis, H.; Xavier, R.J. Microbial genes and pathways in inflammatory bowel disease. Nat. Rev. Microbiol. 2019, 17(8), 497–511. [Google Scholar] [CrossRef]
- Goethel, A.; Croitoru, K.; Philpott, D. The interplay between microbes and the immune response in inflammatory bowel disease. J. Physiol. 2018, 596, 3869–3882. [Google Scholar] [CrossRef] [Green Version]
- Mizushima, N. A brief history of autophagy from cell biology to physiology and disease. Nature 2018, 20, 521–527. [Google Scholar] [CrossRef]
- Zhang, H.; Zheng, L.; McGovern, D.P.B.; Hamill, A.M.; Ichikawa, R.; Kanazawa, Y.; Luu, J.; Kumagai, K.; Cilluffo, M.; Fukata, M.; et al. Myeloid ATG16L1 Facilitates Host-Bacteria Interactions in Maintaining Intestinal Homeostasis. J. Immunol. 2017, 198, 2133–2146. [Google Scholar] [CrossRef] [Green Version]
- Takagawa, T.; Kitani, A.; Fuss, I.; Levine, B.; Brant, S.R.; Peter, I.; Tajima, M.; Nakamura, S.; Strober, W. An increase in LRRK2 suppresses autophagy and enhances Dectin-1–induced immunity in a mouse model of colitis. Sci. Transl. Med. 2018, 10, eaan8162. [Google Scholar] [CrossRef] [Green Version]
- Kim, S.; Eun, H.S.; Jo, E.K. Roles of Autophagy-Related Genes in the Pathogenesis of Inflammatory Bowel Disease. Cells 2019, 8(1), 77. [Google Scholar] [CrossRef] [Green Version]
- Iida, T.; Onodera, K.; Nakase, H. Role of autophagy in the pathogenesis of inflammatory bowel disease. World J. Gastroenterol. 2017, 23, 1944–1953. [Google Scholar] [CrossRef]
- Spalinger, M.R.; Manzini, R.; Hering, L.; Riggs, J.B.; Gottier, C.; Lang, S.; Atrott, K.; Fettelschoss, A.; Olomski, F.; Kündig, T.M.; et al. PTPN2 Regulates Inflammasome Activation and Controls Onset of Intestinal Inflammation and Colon Cancer. Cell Rep. 2018, 22, 1835–1848. [Google Scholar] [CrossRef] [Green Version]
- Spalinger, M.R.; Kasper, S.; Chassard, C.; Raselli, T.; Freywagner, I.; Gottier, C.; Lang, S.; Atrott, K.; Vavricka, S.R.; Mair, F.; et al. PTPN2 controls differentiation of CD4+ T cells and limits intestinal inflammation and intestinal dysbiosis. Mucosal Immunol. 2015, 8, 918–929. [Google Scholar] [CrossRef]
- Hoffmann, P.; Lamerz, D.; Hill, P.; Kirchner, M.; Gauss, A. Gene Polymorphisms of NOD2, IL23R, PTPN2 and ATG16L1 in Patients with Crohn’s Disease: On the Way to Personalized Medicine? Genes 2021, 12, 866. [Google Scholar] [CrossRef] [PubMed]
- Drummond, R.; Lionakis, M.S. Mechanistic Insights into the Role of C-Type Lectin Receptor/CARD9 Signaling in Human Antifungal Immunity. Front. Cell. Infect. Microbiol. 2016, 6, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, W.; Zhang, R.; Wang, X.; Song, Y.; Liu, Z.; Han, W.; Li, R. Impairment of Immune Response against Dematiaceous Fungi in Card9 Knockout Mice. Mycopathologia 2016, 181, 631–642. [Google Scholar] [CrossRef] [PubMed]
- Bergmann, H.; Roth, S.; Pechloff, K.; Kiss, E.A.; Kuhn, S.; Heikenwälder, M.; Diefenbach, A.; Greten, F.R.; Ruland, J. Card9-dependent IL-1β regulates IL-22 production from group 3 innate lymphoid cells and promotes colitis-associated cancer. Eur. J. Immunol. 2017, 47, 1342–1353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartjes, L.; Ruland, J. CARD9 Signaling in Intestinal Immune Homeostasis and Oncogenesis. Front. Immunol. 2019, 10, 419. [Google Scholar] [CrossRef] [Green Version]
- Sun, R.; Hedl, M.; Abraham, C. IL23 induces IL23R recycling and amplifies innate receptor-induced signalling and cytokines in human macrophages, and the IBD-protective IL23R R381Q variant modulates these outcomes. Gut 2020, 69, 264–273. [Google Scholar] [CrossRef]
- Schmitt, H.; Neurath, M.F.; Atreya, R. Role of the IL23/IL17 Pathway in Crohn’s Disease. Front. Immunol. 2021, 12, 622934. [Google Scholar] [CrossRef]
- Liu, Z.; Yadav, P.K.; Xu, X.; Su, J.; Chen, C.; Tang, M.; Lin, H.; Yu, J.; Qian, J.; Yang, P.-C.; et al. The increased expression of IL-23 in inflammatory bowel disease promotes intraepithelial and lamina propria lymphocyte inflammatory responses and cytotoxicity. J. Leukoc. Biol. 2011, 89, 597–606. [Google Scholar] [CrossRef]
- Xu, W.-D.; Xie, Q.-B.; Zhao, Y.; Liu, Y. Association of Interleukin-23 receptor gene polymorphisms with susceptibility to Crohn’s disease: A meta-analysis. Sci. Rep. 2015, 5, 18584. [Google Scholar] [CrossRef] [Green Version]
- Peng, L.-L.; Wang, Y.; Zhu, F.-L.; Xu, W.-D.; Ji, X.-L.; Ni, J. IL-23R mutation is associated with ulcerative colitis: A systemic review and meta-analysis. Oncotarget 2017, 8, 4849–4863. [Google Scholar] [CrossRef]
- Krawiec, P.; Pawłowska-Kamieniak, A.; Pac-Kożuchowska, E. Interleukin 10 and interleukin 10 receptor in paediatric inflammatory bowel disease: From bench to bedside lesson. J. Inflamm. 2021, 18, 1–5. [Google Scholar] [CrossRef]
- Lin, Z.; Wang, Z.; Hegarty, J.; Lin, T.R.; Wang, Y.; Deiling, S.; Wu, R.; Thomas, N.J.; Floros, J. Genetic association and epistatic interaction of the interleukin-10 signaling pathway in pediatric inflammatory bowel disease. World J. Gastroenterol. 2017, 23, 4897–4909. [Google Scholar] [CrossRef]
- Neumann, C.; Scheffold, A.; Rutz, S. Functions and regulation of T cell-derived interleukin-10. Semin. Immunol. 2019, 44, 101344. [Google Scholar] [CrossRef]
- Engelhardt, K.R.; Grimbacher, B. IL-10 in Humans: Lessons from the Gut, IL-10/IL-10 Receptor Deficiencies, and IL-10 Polymorphisms. Curr. Top. Microbiol. Immunol. 2014, 380, 1–18. [Google Scholar] [CrossRef]
- Papierska, K.; Krajka-Kuźniak, V. STAT3 as a therapeutic target. Farm. Współczesna 2020, 13, 29–34. [Google Scholar]
- Wei, H.-X.; Wang, B.; Li, B. IL-10 and IL-22 in Mucosal Immunity: Driving Protection and Pathology. Front. Immunol. 2020, 11, 1315. [Google Scholar] [CrossRef]
- Franke, A.; Balschun, T.; Karlsen, T.H.; Sventoraityte, J.; Nikolaus, S.; Mayr, G.; Domingues, F.; Albrecht, M.; Nothnagel, M.; et al.; the IBSEN study group Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat. Genet. 2008, 40, 1319–1323. [Google Scholar] [CrossRef]
- Zhu, L.; Shi, T.; Zhong, C.; Wang, Y.; Chang, M.; Liu, X. IL-10 and IL-10 Receptor Mutations in Very Early Onset Inflammatory Bowel Disease. Gastroenterol. Res. 2017, 10, 65–69. [Google Scholar] [CrossRef]
- Richard, A.C.; Peters, J.E.; Savinykh, N.; Lee, J.C.; Hawley, E.T.; Meylan, F.; Siegel, R.M.; Lyons, P.A.; Smith, K. Reduced monocyte and macrophage TNFSF15/TL1A expression is associated with susceptibility to inflammatory bowel disease. PLoS Genet. 2018, 14, e1007458. [Google Scholar] [CrossRef]
- Zhang, J.; Zhang, J.; Wu, D.; Wang, J.; Dong, W. Associations between TNFSF15 polymorphisms and susceptibility to ulcerative colitis and Crohn′s disease: A meta-analysis. Autoimmunity 2014, 47, 512–518. [Google Scholar] [CrossRef]
- Wang, I.C.Y.; Kitson, J.; Thern, A.; Williamson, J.; Farrow, S.N.; Owen, M.J. Genomic structure, expression, and chromosome mapping of the mouse homologue for the WSL-1 ( DR3, Apo3, TRAMP, LARD, TR3, TNFRSF12 ) gene. Immunogenetics 2001, 53, 59–63. [Google Scholar] [CrossRef] [PubMed]
- Valatas, V.; Kolios, G.; Bamias, G. TL1A (TNFSF15) and DR3 (TNFRSF25): A Co-stimulatory System of Cytokines With Diverse Functions in Gut Mucosal Immunity. Front. Immunol. 2019, 10, 583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.Z.; Van Sommeren, S.; Huang, H.; Ng, S.C.; Alberts, R.; Takahashi, A.; Ripke, S.; Lee, J.C.; Jostins, L.; Shah, T.; et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 2015, 47, 979–986. [Google Scholar] [CrossRef] [PubMed]
- Lan, X.; Lan, X.; Chang, Y.; Zhang, X.; Liu, J.; Vikash, V.; Wang, W.; Huang, M.; Wang, X.; Zhou, F.; et al. Identification of Two Additional Susceptibility Loci for Inflammatory Bowel Disease in a Chinese Populationy. Cell. Physiol. Biochem. 2017, 41, 2077–2090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Migone, T.-S.; Zhang, J.; Luo, X.; Zhuang, L.; Chen, C.; Hu, B.; Hong, J.S.; Perry, J.W.; Chen, S.-F.; Zhou, J.X.; et al. TL1A Is a TNF-like Ligand for DR3 and TR6/DcR3 and Functions as a T Cell Costimulator. Immunity 2002, 16, 479–492. [Google Scholar] [CrossRef] [Green Version]
- Chizzolini, C.; Dufour, A.M.; Brembilla, N.C. Is there a role for IL-17 in the pathogenesis of systemic sclerosis? Immunol. Lett. 2018, 195, 61–67. [Google Scholar] [CrossRef]
- Bamias, G.; Mishina, M.; Nyce, M.; Ross, W.G.; Kollias, G.; Rivera-Nieves, J.; Pizarro, T.T.; Cominelli, F. Role of TL1A and its receptor DR3 in two models of chronic murine ileitis. Proc. Natl. Acad. Sci. USA 2006, 103, 8441–8446. [Google Scholar] [CrossRef] [Green Version]
- Bamias, G.; Filidou, E.; Goukos, D.; Valatas, V.; Arvanitidis, K.; Panagopoulou, M.; Kouklakis, G.; Daikos, G.L.; Ladas, S.D.; Kolios, G. Crohn′s disease-associated mucosal factors regulate the expression of TNF-like cytokine 1A and its receptors in primary subepithelial intestinal myofibroblasts and intestinal epithelial cells. Transl. Res. 2016, 180, 118–130.e2. [Google Scholar] [CrossRef]
- Strisciuglio, C.; Giugliano, F.; Martinelli, M.; Cenni, S.; Greco, L.; Staiano, A.; Miele, E. Impact of Environmental and Familial Factors in a Cohort of Pediatric Patients With Inflammatory Bowel Disease. J. Craniofacial Surg. 2017, 64, 569–574. [Google Scholar] [CrossRef]
- Amarapurkar, A.D.; Amarapurkar, D.N.; Rathi, P.; Sawant, P.; Patel, N.; Kamani, P.; Rawal, K.; Baijal, R.; Sonawane, A.; Narawane, N.; et al. Risk factors for inflammatory bowel disease: A prospective multi-center study. Indian J. Gastroenterol. 2018, 37, 189–195. [Google Scholar] [CrossRef]
- Santos, M.P.C.; Gomes, C.; Torres, J. Familial and ethnic risk in inflammatory bowel disease. Ann. Gastroenterol. 2018, 31, 14–23. [Google Scholar] [CrossRef]
- Moller, F.T.; Andersen, V.; Wohlfahrt, J.; Jess, T. Familial Risk of Inflammatory Bowel Disease: A Population-Based Cohort Study 1977–2011. Am. J. Gastroenterol. 2015, 110, 564–571. [Google Scholar] [CrossRef]
- Kedia, S.; Ahuja, V. Does the road to primary prevention of inflammatory bowel disease start from childhood? JGH Open. 2022, 6(6), 365–368. [Google Scholar] [CrossRef]
- Andreu, M.; Márquez, L.; Domènech, E.; Gisbert, J.; García, V.; Marín-Jiménez, I.; Peñalva, M.; Gomollón, F.; Calvet, X.; Merino, O.; et al. Disease severity in familial cases of IBD. J. Crohn’s Colitis 2014, 8, 234–239. [Google Scholar] [CrossRef] [Green Version]
- Chao, C.-Y.; Bessissow, T. Does Familial IBD Have its Own Signature? J. Crohn’s Colitis 2018, 12, 515–516. [Google Scholar] [CrossRef] [Green Version]
- Boaz, E.; Shitrit, A.B.-G.; Schechter, M.; Goldin, E.; Reissman, P.; Yellinek, S.; Koslowsky, B. Inflammatory bowel disease in families with four or more affected first-degree relatives. Scand. J. Gastroenterol. 2022, 1–5. [Google Scholar] [CrossRef]
- Ruban, M.; Slavick, A.; Amir, A.; Ben-Tov, A.; Moran-Lev, H.; Weintraub, Y.; Anafy, A.; Cohen, S.; Yerushalmy-Feler, A. Increasing rate of a positive family history of inflammatory bowel disease (IBD) in pediatric IBD patients. Eur. J. Pediatr. 2022, 181, 1–7. [Google Scholar] [CrossRef]
- Kuenzig, M.E.; Fung, S.G.; Marderfeld, L.; Mak, J.W.; Kaplan, G.G.; Ng, S.C.; Wilson, D.C.; Cameron, F.; Henderson, P.; Kotze, P.G.; et al. Twenty-first Century Trends in the Global Epidemiology of Pediatric-Onset Inflammatory Bowel Disease: Systematic Review. Gastroenterology 2022, 162, 1147–1159.e4. [Google Scholar] [CrossRef]
- Mosli, M.; Alzahrani, A.; Showlag, S.; AlShehri, A.; Hejazi, A.; Alnefaie, M.; Almaymuni, A.; Abdullahi, M.; Albeshir, M.; Alsulais, E.; et al. A cross-sectional survey of multi-generation inflammatory bowel disease consanguinity and its relationship with disease onset. Saudi J. Gastroenterol. 2017, 23, 337–340. [Google Scholar] [CrossRef]
- Bell, J.T.; Spector, T.D. A twin approach to unraveling epigenetics. Trends Genet. 2011, 27, 116–125. [Google Scholar] [CrossRef] [Green Version]
- Hwang, S.W.; Kwak, M.S.; Kim, W.S.; Lee, J.-M.; Park, S.H.; Lee, H.-S.; Yang, D.-H.; Kim, K.-J.; Ye, B.D.; Byeon, J.-S.; et al. Influence of a Positive Family History on the Clinical Course of Inflammatory Bowel Disease. J. Crohn’s Colitis 2016, 10, 1024–1032. [Google Scholar] [CrossRef] [PubMed]
- Moller, F.T.; Andersen, V.; Andersson, M.; Jess, T. Hospital Admissions, Biological Therapy, and Surgery in Familial and Sporadic Cases of Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2015, 21, 2825–2832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kevans, D.; Silverberg, M.S.; Borowski, K.; Griffiths, A.; Xu, W.; Onay, V.; Paterson, A.D.; Knight, J.; Croitoru, K.; Project, O.B.O.T.G. IBD Genetic Risk Profile in Healthy First-Degree Relatives of Crohn’s Disease Patients. J. Crohn′s Colitis 2016, 10, 209–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gabbani, T.; Deiana, S.; Annese, A.L.; Lunardi, S.; Annese, V. The genetic burden of inflammatory bowel diseases: Implications for the clinic? Expert Rev. Gastroenterol. Hepatol. 2016, 10, 1109–1117. [Google Scholar] [CrossRef] [PubMed]
- Capone, K.; Rosenberg, H.J.; Wroblewski, K.; Gokhale, R.; Kirschner, B.S. Change in Prevalence of Family History During Long-term Follow-up of Patients With Pediatric-onset Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 829–834. [Google Scholar] [CrossRef]
- Xu, S.; Zou, H.; Zhang, H.; Zhu, S.; Zhou, R.; Li, J. Investigation of inflammatory bowel disease risk factors in 4 families in central China. Exp. Ther. Med. 2018, 15, 1367–1375. [Google Scholar] [CrossRef] [Green Version]
- Ballester, M.P.; Marti-Aguado, D.; Tosca, J.; Bosca-Watts, M.M.; Sanahuja, A.; Navarro, P.; Pascual, I.; Antón, R.; Mora, F.; Mínguez, M. Disease severity and treatment requirements in familial inflammatory bowel disease. Int. J. Color. Dis. 2017, 32, 1197–1205. [Google Scholar] [CrossRef]
- Halfvarson, J.; Ludvigsson, J.F.; Bresso, F.; Askling, J.; Sachs, M.C.; Olén, O. Age determines the risk of familial inflammatory bowel disease—A nationwide study. Aliment. Pharmacol. Ther. 2022, 56, 491–500. [Google Scholar] [CrossRef]
- Borren, N.; Conway, G.; Garber, J.J.; Khalili, H.; Budree, S.; Mallick, H.; Yajnik, V.; Xavier, R.J.; Ananthakrishnan, A.N. Differences in Clinical Course, Genetics, and the Microbiome Between Familial and Sporadic Inflammatory Bowel Diseases. J. Crohn’s Colitis 2018, 12, 525–531. [Google Scholar] [CrossRef]
- Uhlig, H.H.; Schwerd, T.; Koletzko, S.; Shah, N.; Kammermeier, J.; Elkadri, A.; Ouahed, J.; Wilson, D.C.; Travis, S.P.; Turner, D.; et al. The Diagnostic Approach to Monogenic Very Early Onset Inflammatory Bowel Disease. Gastroenterology 2014, 147, 990–1007.e3. [Google Scholar] [CrossRef] [Green Version]
- Park, S.H.; Hwang, S.W.; Ye, B.D.; Noh, S.; Park, J.C.; Kim, J.Y.; Kim, J.; Ham, N.S.; Oh, E.H.; Yang, D.; et al. Concordance regarding disease type and phenotypic characteristics among patients with familial inflammatory bowel disease. J. Gastroenterol. Hepatol. 2020, 35, 988–993. [Google Scholar] [CrossRef]
- Ek, W.; D’Amat, M.; Halfvarson, J. The history of genetics in inflammatory bowel disease. Ann. Gastroenterol. 2014, 27, 294–303. [Google Scholar]
- El Mouzan, M.; Al-Mofarreh, M.; Assiri, A.; Hamid, Y.; Saeed, A. Consanguinity and Inflammatory Bowel Diseases. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 182–185. [Google Scholar] [CrossRef]
- Wang, P.-Q.; Hu, J.; Al Kazzi, E.; Akhuemonkhan, E.; Zhi, M.; Gao, X.; Pessoa, R.H.D.P.; Ghazaleh, S.; Cornelius, T.; Sabunwala, S.A.; et al. Family history and disease outcomes in patients with Crohn’s disease: A comparison between China and the United States. World J. Gastrointest. Pharmacol. Ther. 2016, 7, 556–563. [Google Scholar] [CrossRef]
- Jacobs, J.P.; Goudarzi, M.; Singh, N.; Tong, M.; McHardy, I.; Ruegger, P.; Asadourian, M.; Moon, B.-H.; Ayson, A.; Borneman, J.; et al. A Disease-Associated Microbial and Metabolomics State in Relatives of Pediatric Inflammatory Bowel Disease Patients. Cell. Mol. Gastroenterol. Hepatol. 2016, 2, 750–766. [Google Scholar] [CrossRef] [Green Version]
- Mosli, M.; Zou, G.; Garg, S.K.; Feagan, S.G.; MacDonald, J.K.; Chande, N.; Sandborn, W.J.; Feagan, B.G. C-Reactive Protein, Fecal Calprotectin, and Stool Lactoferrin for Detection of Endoscopic Activity in Symptomatic Inflammatory Bowel Disease Patients: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 2015, 110, 802–819. [Google Scholar] [CrossRef]
- Cho, J.; Nicolae, D.; Gold, L.; Fields, C.; LaBuda, M.; Rohal, P.; Pickles, M.; Qin, L.; Fu, Y.; Mann, J.; et al. Identification of novel susceptibility loci for inflammatory bowel disease on chromosomes 1p, 3q, and 4q: Evidence for epistasis between 1p and IBD1. Proc. Natl. Acad. Sci. USA 1998, 95, 7502–7507. [Google Scholar] [CrossRef] [Green Version]
- Barmada, M.M.; Brant, S.R.; Nicolae, D.; Achkar, J.-P.; Panhuysen, C.I.; Bayless, T.M.; Cho, J.H.; Duerr, R.H. A Genome Scan in 260 Inflammatory Bowel Disease-Affected Relative Pairs. Inflamm. Bowel Dis. 2004, 10, 513–520. [Google Scholar] [CrossRef]
- Vermeire, S.; Rutgeerts, P.; Van Steen, K.; Joossens, S.; Claessens, G.; Pierik, M.; Peeters, M.; Vlietinck, R. Genome wide scan in a Flemish inflammatory bowel disease population: Support for the IBD4 locus, population heterogeneity, and epistasis. Gut 2004, 53, 980–986. [Google Scholar] [CrossRef] [Green Version]
- Paavola, P.; Helio, T.; Kiuru, M.; Halme, L.; Turunen, U.; Terwilliger, J.; Karvonen, A.-L.; Julkunen, R.; Niemelä, S.; Nurmi, H.; et al. Genetic analysis in Finnish families with inflammatory bowel disease supports linkage to chromosome 3p21. Eur. J. Hum. Genet. 2001, 9, 328–334. [Google Scholar] [CrossRef] [Green Version]
- Rioux, J.; Daly, M.; Green, T.; Stone, V.; Lander, E.; Hudson, T.; Steinhart, A.; Bull, S.; Cohen, Z.; Greenberg, G. Absence of linkage between inflammatory bowel disease and selected loci on chromosomes 3, 7, 12, and 16. Gastroenterology 1998, 115, 1062–1065. [Google Scholar] [CrossRef] [PubMed]
- Williams, C.N.; Kocher, K.; Lander, E.S.; Daly, M.J.; Rioux, J.D. Using a Genome-Wide Scan and Meta-analysis to Identify a Novel IBD Locus and Confirm Previously Identified IBD Loci. Inflamm. Bowel Dis. 2002, 8, 375–381. [Google Scholar] [CrossRef] [PubMed]
- Duerr, R.H.; Barmada, M.M.; Zhang, L.; Pfützer, R.; Weeks, D.E. High-Density Genome Scan in Crohn Disease Shows Confirmed Linkage to Chromosome 14q11-12. Am. J. Hum. Genet. 2000, 66, 1857–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, C.; Hu, G.; Zeng, Z.; Lin, L.; Gu, G. Progress in searching for susceptibility gene for inflammatory bowel disease by positional cloning. World J. Gastroenterol. 2003, 9, 1646–1656. [Google Scholar] [CrossRef] [PubMed]
- Tamboli, C.; Cortot, A.; Colombel, J. What are the major arguments in favour of the genetic susceptibility for inflammatory bowel disease? Eur. J. Gastroenterol. Hepatol. 2003, 15, 587–592. [Google Scholar] [CrossRef]
- McGovern, D.P.B.; Gardet, A.; Törkvist, L.; Goyette, P.; Essers, J.; Taylor, K.D.; Neale, B.M.; Ong, R.T.H.; Lagacé, C.; et al.; The NIDDK IBD Genetics Consortium Genome-wide association identifies multiple ulcerative colitis susceptibility loci. Nat. Genet. 2010, 42, 332–337. [Google Scholar] [CrossRef] [Green Version]
- Fisher, S.; Tremelling, M.; Anderson, C.; Gwilliam, R.; Bumpstead, S.; Prescott, N.; Nimmo, E.; Massey, D.; Berzuini, C.; Johnson, C.; et al. Genetic determinants of ulcerative colitis include the ECM1 locus and five loci implicated in Crohn’s disease. Nat. Genet. 2008, 40, 710–712. [Google Scholar] [CrossRef]
- Silverberg, M.S.; Cho, J.H.; Rioux, J.D.; McGovern, D.P.B.; Wu, J.; Annese, V.; Achkar, J.-P.; Goyette, P.; Scott, R.; Xu, W.; et al. Ulcerative colitis–risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study. Nat. Genet. 2009, 41, 216–220. [Google Scholar] [CrossRef]
- de Lange, K.M.; Barrett, J.C. Understanding inflammatory bowel disease via immunogenetics. J. Autoimmun. 2015, 64, 91–100. [Google Scholar] [CrossRef] [Green Version]
- Gordon, H.; Moller, F.T.; Andersen, V.; Harbord, M. Heritability in inflammatory bowel disease: From the first twin study to genome-wide association studies. Inflamm. Bowel Dis. 2015, 21, 1428–1434. [Google Scholar] [CrossRef] [Green Version]
- Moller, F.T.; Knudsen, L.A.; Harbord, M.; Satsangi, J.; Gordon, H.; Christiansen, L.; Christensen, K.; Jess, T.; Andersen, V. Danish cohort of monozygotic inflammatory bowel disease twins: Clinical characteristics and inflammatory activity. World J. Gastroenterol. 2016, 22, 5050–5059. [Google Scholar] [CrossRef] [Green Version]
- Bengtson, M.-B.; Aamodt, G.; Vatn, M.H.; Harris, J.R. Concordance for IBD among twins compared to ordinary siblings—A Norwegian population-based study. J. Crohn′s Colitis 2010, 4, 312–318. [Google Scholar] [CrossRef]
- Gordon, H.; Blad, W.; Møller, F.T.; Orchard, T.; Steel, A.; Trevelyan, G.; Ng, S.; Harbord, M. UK IBD Twin Registry: Concordance and Environmental Risk Factors of Twins with IBD. Am. J. Dig. Dis. 2022, 67, 2444–2450. [Google Scholar] [CrossRef]
- Brand, E.C.; Klaassen, M.A.; Gacesa, R.; Vila, A.V.; Ghosh, H.; de Zoete, M.R.; Boomsma, D.I.; Hoentjen, F.; Horje, C.S.H.T.; van de Meeberg, P.C.; et al. Healthy Cotwins Share Gut Microbiome Signatures With Their Inflammatory Bowel Disease Twins and Unrelated Patients. Gastroenterology 2021, 160, 1970–1985. [Google Scholar] [CrossRef]
- Lepage, P.; Häsler, R.; Spehlmann, M.E.; Rehman, A.; Zvirbliene, A.; Begun, A.; Ott, S.; Kupcinskas, L.; Doré, J.; Raedler, A.; et al. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Nat. Commun. 2020, 11, 1512. [Google Scholar] [CrossRef]
- Brodin, P.; Jojic, V.; Gao, T.; Bhattacharya, S.; Angel, C.J.L.; Furman, D.; Shen-Orr, S.; Dekker, C.L.; Swan, G.E.; Butte, A.J.; et al. Variation in the Human Immune System Is Largely Driven by Non-Heritable Influences. Cell 2015, 160, 37–47. [Google Scholar] [CrossRef] [Green Version]
- Du, J.; Yin, J.; Du, H.; Zhang, J. Revisiting an Expression Dataset of Discordant Inflammatory Bowel Disease Twin Pairs Using a Mutation Burden Test Reveals CYP2C18 as a Novel Marker. Front. Genet. 2021, 12, 680125. [Google Scholar] [CrossRef]
- Natasha, G.; Zilbauer, M. Epigenetics in IBD: A conceptual framework for disease pathogenesis. Front. Gastroenterol. 2022, 13, e22–e27. [Google Scholar] [CrossRef]
- Xu, J.; Xu, H.-M.; Yang, M.-F.; Liang, Y.-J.; Peng, Q.-Z.; Zhang, Y.; Tian, C.-M.; Wang, L.-S.; Yao, J.; Nie, Y.-Q.; et al. New Insights Into the Epigenetic Regulation of Inflammatory Bowel Disease. Front. Pharmacol. 2022, 13, 813659. [Google Scholar] [CrossRef]
- Howell, K.J.; Kraiczy, J.; Nayak, K.M.; Gasparetto, M.; Ross, A.; Lee, C.; Mak, T.N.; Koo, B.-K.; Kumar, N.; Lawley, T.; et al. DNA Methylation and Transcription Patterns in Intestinal Epithelial Cells From Pediatric Patients With Inflammatory Bowel Diseases Differentiate Disease Subtypes and Associate With Outcome. Gastroenterology 2018, 154, 585–598. [Google Scholar] [CrossRef] [Green Version]
- Somineni, H.K.; Venkateswaran, S.; Kilaru, V.; Marigorta, U.M.; Mo, A.; Okou, D.T.; Kellermayer, R.; Mondal, K.; Cobb, D.; Walters, T.D.; et al. Blood-Derived DNA Methylation Signatures of Crohn′s Disease and Severity of Intestinal Inflammation. Gastroenterology 2019, 156, 2254–2265.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gasparetto, M.; Payne, F.; Nayak, K.; Kraiczy, J.; Glemas, C.; Philip-McKenzie, Y.; Ross, A.; Edgar, R.D.; Zerbino, D.R.; Salvestrini, C.; et al. Transcription and DNA Methylation Patterns of Blood-Derived CD8+ T Cells Are Associated With Age and Inflammatory Bowel Disease But Do Not Predict Prognosis. Gastroenterology 2021, 160, 232–244.e7. [Google Scholar] [CrossRef] [PubMed]
- Moret-Tatay, I.; Cerrillo, E.; Sáez-González, E.; Hervás, D.; Iborra, M.; Sandoval, J.; Busó, E.; Tortosa, L.; Nos, P.; Beltrán, B. Identification of Epigenetic Methylation Signatures With Clinical Value in Crohnʼs Disease. Clin. Transl. Gastroenterol. 2019, 10, e00083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rapozo, D.C.M.; Bernardazzi, C.; De Souza, H.S.P. Diet and microbiota in inflammatory bowel disease: The gut in disharmony. World J. Gastroenterol. 2017, 23, 2124–2140. [Google Scholar] [CrossRef] [PubMed]
- Silva, J.P.B.; Navegantes-Lima, K.C.; de Oliveira, A.L.B.; Rodrigues, D.V.S.; Gaspar, S.L.F.; Monteiro, V.V.S.; Moura, D.P.; Monteiro, M.C. Protective Mechanisms of Butyrate on Inflammatory Bowel Disease. Curr. Pharm. Des. 2018, 24, 4154–4166. [Google Scholar] [CrossRef]
- Zhang, M.; Zhou, L.; Zhang, S.; Yang, Y.; Xu, L.; Hua, Z.; Zou, X. Bifidobacterium longum affects the methylation level of forkhead box P3 promoter in 2, 4, 6-trinitrobenzenesulphonic acid induced colitis in rats. Microb. Pathog. 2017, 110, 426–430. [Google Scholar] [CrossRef]
- Mannino, G.; Caradonna, F.; Cruciata, I.; Lauria, A.; Perrone, A.; Gentile, C. Melatonin reduces inflammatory response in human intestinal epithelial cells stimulated by interleukin-1β. J. Pineal Res. 2019, 67, e12598. [Google Scholar] [CrossRef]
- Pan, W.-H.; Sommer, F.; Falk-Paulsen, M.; Ulas, T.; Best, P.; Fazio, A.; Kachroo, P.; Luzius, A.; Jentzsch, M.; Rehman, A.; et al. Exposure to the gut microbiota drives distinct methylome and transcriptome changes in intestinal epithelial cells during postnatal development. Genome Med. 2018, 10, 1–15. [Google Scholar] [CrossRef]
- Ansari, I.; Raddatz, G.; Gutekunst, J.; Ridnik, M.; Cohen, D.; Abu-Remaileh, M.; Tuganbaev, T.; Shapiro, H.; Pikarsky, E.; Elinav, E.; et al. The microbiota programs DNA methylation to control intestinal homeostasis and inflammation. Nat. Microbiol. 2020, 5, 610–619. [Google Scholar] [CrossRef]
- Ventham, N.; Kennedy, N.; Adams, A.; Kalla, R.; Heath, S.; O’Leary, K.; Drummond, H. IBD BIOM consortium; IBD CHARACTER consortium, Wilson DC, Gut IG, Nimmo ER, Satsangi J. Integrative epigenome-wide analysis demonstrates that DNA methylation may mediate genetic risk in inflammatory bowel disease. Nat. Commun. 2016, 7, 13507. [Google Scholar] [CrossRef] [Green Version]
- Kraiczy, J.; Nayak, K.; Ross, A.; Raine, T.; Mak, T.N.; Gasparetto, M.; Cario, E.; Rakyan, V.; Heuschkel, R.; Zilbauer, M. Assessing DNA methylation in the developing human intestinal epithelium: Potential link to inflammatory bowel disease. Mucosal Immunol. 2016, 9, 647–658. [Google Scholar] [CrossRef] [Green Version]
- Yim, A.Y.F.L.; Duijvis, N.W.; Zhao, J.; de Jonge, W.J.; D’Haens, G.R.A.M.; Mannens, M.M.A.M.; Mul, A.N.P.M.; Velde, A.A.T.; Henneman, P. Peripheral blood methylation profiling of female Crohn’s disease patients. Clin. Epigenetics 2016, 8, 1–13. [Google Scholar] [CrossRef]
- McDermott, E.; Ryan, E.J.; Tosetto, M.; Gibson, D.; Burrage, J.; Keegan, D.; Byrne, K.; Crowe, E.; Sexton, G.; Malone, K.; et al. DNA Methylation Profiling in Inflammatory Bowel Disease Provides New Insights into Disease Pathogenesis. J. Crohn′s Colitis 2016, 10, 77–86. [Google Scholar] [CrossRef] [Green Version]
- Venkateswaran, S.; Somineni, H.K.; Kilaru, V.; Katrinli, S.; Prince, J.; Okou, D.T.; Hyams, J.S.; A Denson, L.; Kellermayer, R.; Gibson, G.; et al. Methylation quantitative trait loci are largely consistent across disease states in Crohn’s disease. G3 Genes|Genomes|Genetics 2022, 12, jkac041. [Google Scholar] [CrossRef]
- Yi, J. DNA Methylation Change Profiling of Colorectal Disease: Screening towards Clinical Use. Life 2021, 11, 412. [Google Scholar] [CrossRef]
- Rajamäki, K.; Taira, A.; Katainen, R.; Välimäki, N.; Kuosmanen, A.; Plaketti, R.-M.; Seppälä, T.T.; Ahtiainen, M.; Wirta, E.-V.; Vartiainen, E.; et al. Genetic and Epigenetic Characteristics of Inflammatory Bowel Disease–Associated Colorectal Cancer. Gastroenterology 2021, 161, 592–607. [Google Scholar] [CrossRef]
- Joustra, V.; Hageman, I.L.; Satsangi, J.; Adams, A.; Ventham, N.T.; de Jonge, W.J.; Henneman, P.; D’Haens, G.R.; Yim, A.Y.F.L. Systematic Review and Meta-analysis of Peripheral Blood DNA methylation studies in Inflammatory Bowel Disease. J. Crohn′s Colitis 2022, jjac119. [Google Scholar] [CrossRef]
- Zhang, T.; Cooper, S.; Brockdorff, N. The interplay of histone modifications–writers that read. EMBO Rep. 2015, 16, 1467–1481. [Google Scholar] [CrossRef]
- Zeng, Z.; Mukherjee, A.; Zhang, H. From Genetics to Epigenetics, Roles of Epigenetics in Inflammatory Bowel Disease. Front. Genet. 2019, 10, 1017. [Google Scholar] [CrossRef] [Green Version]
- Chen, P.; Zhu, H.; Mao, Y.; Zhuo, M.; Yu, Y.; Chen, M.; Zhao, Q.; Li, L.; Wu, M.; Ye, M. SETD8 involved in the progression of inflammatory bowel disease via epigenetically regulating p62 expression. J. Gastroenterol. Hepatol. 2021, 36, 2850–2863. [Google Scholar] [CrossRef]
- Chan, S.-N.; Low, E.N.D.; Ali, R.A.R.; Mokhtar, N.M. Delineating inflammatory bowel disease through transcriptomic studies: Current review of progress and evidence. Intest. Res. 2018, 16, 374–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- James, J.; Riis, L.B.; Malham, M.; Høgdall, E.; Langholz, E.; Nielsen, B.S. MicroRNA Biomarkers in IBD—Differential Diagnosis and Prediction of Colitis-Associated Cancer. Int. J. Mol. Sci. 2020, 21, 7893. [Google Scholar] [CrossRef] [PubMed]
- He, C.; Shi, Y.; Wu, R.; Sun, M.; Fang, L.; Wu, W.; Liu, C.; Tang, M.; Li, Z.; Wang, P.; et al. miR-301a promotes intestinal mucosal inflammation through induction of IL-17A and TNF-α in IBD. Gut 2016, 65, 1938–1950. [Google Scholar] [CrossRef] [PubMed]
- Gruszka, R.; Zakrzewska, M. The Oncogenic Relevance of miR-17-92 Cluster and Its Paralogous miR-106b-25 and miR-106a-363 Clusters in Brain Tumors. Int. J. Mol. Sci. 2018, 19, 879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perconti, G.; Rubino, P.; Contino, F.; Bivona, S.; Bertolazzi, G.; Tumminello, M.; Feo, S.; Giallongo, A.; Coronnello, C. RIP-Chip analysis supports different roles for AGO2 and GW182 proteins in recruiting and processing microRNA targets. BMC Bioinform. 2019, 20, 1–13. [Google Scholar] [CrossRef]
- Neudecker, V.; Haneklaus, M.; Jensen, O.; Khailova, L.; Masterson, J.C.; Tye, H.; Biette, K.; Jedlicka, P.; Brodsky, K.S.; Gerich, M.E.; et al. Myeloid-derived miR-223 regulates intestinal inflammation via repression of the NLRP3 inflammasome. J. Exp. Med. 2017, 214, 1737–1752. [Google Scholar] [CrossRef] [Green Version]
- Wei, M.; Gao, X.; Liu, L.; Li, Z.; Wan, Z.; Dong, Y.; Chen, X.; Niu, Y.; Zhang, J.; Yang, G. Visceral Adipose Tissue Derived Exosomes Exacerbate Colitis Severity via Pro-inflammatory MiRNAs in High Fat Diet Fed Mice. ACS Nano 2020, 14, 5099–5110. [Google Scholar] [CrossRef]
- Sanchez, H.N.; Moroney, J.B.; Gan, H.; Shen, T.; Im, J.L.; Li, T.; Taylor, J.R.; Zan, H.; Casali, P. B cell-intrinsic epigenetic modulation of antibody responses by dietary fiber-derived short-chain fatty acids. Nat. Commun. 2020, 11, 1–19. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.; Fan, R.; Wang, L.; Zhou, J.; Zheng, S.; Hu, S.; Chen, M.; Zhang, T.; Lin, Y.; Zhang, M.; et al. Genetic association between CARD9 variants and inflammatory bowel disease was not replicated in a Chinese Han population. Int. J. Clin. Exp. Pathol. 2015, 8, 13465–13470. [Google Scholar]
- Garo, L.P.; Ajay, A.K.; Fujiwara, M.; Gabriely, G.; Raheja, R.; Kuhn, C.; Kenyon, B.; Skillin, N.; Kadowaki-Saga, R.; Saxena, S.; et al. MicroRNA-146a limits tumorigenic inflammation in colorectal cancer. Nat. Commun. 2021, 12, 1–16. [Google Scholar] [CrossRef]
- Wang, J.; Dong, L.; Wang, M.; Gu, J.; Zhao, Y. MiR-146a regulates the development of ulcerative colitis via mediating the TLR4/MyD88/NF-κB signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2151–2157. [Google Scholar] [CrossRef]
- Schönauen, K.; Le, N.; Von Arnim, U.; Schulz, C.; Malfertheiner, P.; Link, A. Circulating and Fecal microRNAs as Biomarkers for Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2018, 24, 1547–1557. [Google Scholar] [CrossRef]
- Kalla, R.; Adams, A.T.; Ventham, N.T.; A Kennedy, N.; White, R.; Clarke, C.; Ivens, A.; Bergemalm, D.; Vatn, S.; Lopez-Jimena, B.; et al. Whole Blood Profiling of T-cell-Derived microRNA Allows the Development of Prognostic models in Inflammatory Bowel Disease. J. Crohn’s Colitis 2020, 14, 1724–1733. [Google Scholar] [CrossRef]
- Rankin, C.R.; Theodorou, E.; Law, I.K.M.; Rowe, L.; Kokkotou, E.; Pekow, J.; Wang, J.; Martin, M.G.; Pothoulakis, C.; Padua, D.M. Identification of novel mRNAs and lncRNAs associated with mouse experimental colitis and human inflammatory bowel disease. Am. J. Physiol. Liver Physiol. 2018, 315, G722–G733. [Google Scholar] [CrossRef]
- Liu, H.; Li, T.; Zhong, S.; Yu, M.; Huang, W. Intestinal epithelial cells related lncRNA and mRNA expression profiles in dextran sulphate sodium-induced colitis. J. Cell. Mol. Med. 2021, 25, 1060–1073. [Google Scholar] [CrossRef]
- Li, N.; Shi, R. Expression alteration of long non-coding RNAs and their target genes in the intestinal mucosa of patients with Crohn’s disease. Clin. Chim. Acta 2019, 494, 14–21. [Google Scholar] [CrossRef]
- Yarani, R.; Mirza, A.H.; Kaur, S.; Pociot, F. The emerging role of lncRNAs in inflammatory bowel disease. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Rankin, C.R.; Shao, L.; Elliott, J.; Rowe, L.; Patel, A.; Videlock, E.J.; Benhammou, J.N.; Sauk, J.S.; Ather, N.; Corson, M.; et al. The IBD-associated long noncoding RNA IFNG-AS1 regulates the balance between inflammatory and anti-inflammatory cytokine production after T-cell stimulation. Am. J. Physiol. Liver Physiol. 2020, 318, G34–G40. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Arijs, I.; Li, K.; Toedter, G.; Quintens, R.; Van Lommel, L.; Van Steen, K.; Leemans, P.; De Hertogh, G.; Lemaire, K.; Ferrante, M.; et al. Mucosal gene signatures to predict response to infliximab in patients with ulcerative colitis. Gut 2009, 58, 1612–1619. [Google Scholar] [CrossRef] [Green Version]
- McGovern, D.P.B.; Kugathasan, S.; Cho, J.H. Genetics of Inflammatory Bowel Diseases. Gastroenterology 2015, 149, 1163–1176.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lauro, R.; Mannino, F.; Irrera, N.; Squadrito, F.; Altavilla, D.; Squadrito, G.; Pallio, G.; Bitto, A. Pharmacogenetics of Biological Agents Used in Inflammatory Bowel Disease: A Systematic Review. Biomedicines 2021, 9, 1748. [Google Scholar] [CrossRef] [PubMed]
- Bosch, B.J.V.D.; Coenen, M.J. Pharmacogenetics of inflammatory bowel disease. Pharmacogenomics 2021, 22, 55–66. [Google Scholar] [CrossRef]
- Zhang, X.; Myers, J.M.B.; Yadagiri, V.K.; Ulm, A.; Chen, X.; Weirauch, M.T.; Hershey, G.K.K.; Ji, H. Nasal DNA methylation differentiates corticosteroid treatment response in pediatric asthma: A pilot study. PLoS ONE 2017, 12, e0186150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucafò, M.; Di Silvestre, A.; Romano, M.; Avian, A.; Antonelli, R.; Martelossi, S.; Naviglio, S.; Tommasini, A.; Stocco, G.; Ventura, A.; et al. Role of the Long Non-Coding RNA Growth Arrest-Specific 5 in Glucocorticoid Response in Children with Inflammatory Bowel Disease. Basic Clin. Pharmacol. Toxicol. 2018, 122, 87–93. [Google Scholar] [CrossRef] [Green Version]
- Yamamoto-Furusho, J.K. Pharmacogenetics in inflammatory bowel disease: Understanding treatment response and personalizing therapeutic strategies. Pharmacogenomics Pers. Med. 2017, 10, 197–204. [Google Scholar] [CrossRef] [Green Version]
- Park, S.C.; Jeen, Y.T. Genetic Studies of Inflammatory Bowel Disease-Focusing on Asian Patients. Cells 2019, 8, 404. [Google Scholar] [CrossRef] [Green Version]
- Chouchana, L.; Narjoz, C.; Beaune, P.; Loriot, M.-A.; Roblin, X. Review article: The benefits of pharmacogenetics for improving thiopurine therapy in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2012, 35, 15–36. [Google Scholar] [CrossRef]
- Cascorbi, I. The Pharmacogenetics of Immune-Modulating Therapy. Adv. Pharmacol. 2018, 83, 275–296. [Google Scholar] [CrossRef]
- Ye, B.; McGovern, D. Genetic variation in IBD: Progress, clues to pathogenesis and possible clinical utility. Expert Rev. Clin. Immunol. 2016, 12, 1091–1107. [Google Scholar] [CrossRef] [Green Version]
- Moon, W.; Loftus, E.J. Review article: Recent advances in pharmacogenetics and pharmacokinetics for safe and effective thiopurine therapy in inflammatory bowel disease. Aliment Pharmacol Ther. 2016, 43, 863–883. [Google Scholar] [CrossRef] [Green Version]
- Tew, G.W.; Hackney, J.; Gibbons, D.; Lamb, C.; Luca, D.; Egen, J.G.; Diehl, L.; Anderson, J.E.; Vermeire, S.; Mansfield, J.C.; et al. Association Between Response to Etrolizumab and Expression of Integrin αE and Granzyme A in Colon Biopsies of Patients With Ulcerative Colitis. Gastroenterology 2016, 150, 477–487.e9. [Google Scholar] [CrossRef] [Green Version]
- Di Paolo, A.; Luci, G. Personalized Medicine of Monoclonal Antibodies in Inflammatory Bowel Disease: Pharmacogenetics, Therapeutic Drug Monitoring, and Beyond. Front Pharmacol. 2021, 11, 610806. [Google Scholar] [CrossRef]
Genes Studied | Conclusions of the Study | |
---|---|---|
Animal studies | NOD2 gene |
|
Research involving humans | NOD2 gene |
|
Genes Studied | Conclusions of the Study | |
---|---|---|
Animal studies | ATG16L1 gene |
|
Research involving humans | ATG16L1 gene |
|
Genes Studied | Conclusions of the Study | |
---|---|---|
Aminal studies | CARD9 gene |
|
Gene Group | Name of the Gene | Function |
---|---|---|
Genes associated with molecular pattern recognition of pathogens | NOD2/CARD15, CARD9 | Activation of pro-inflammatory and anti-inflammatory cytokines and regulation of inflammation and cell apoptosis. |
Genes associated with autophagy | ATG16L1, IRGM, LRRK2 | Key role in regulating the interaction between the gut microbiota and innate and acquired immunity, and in host defense against intestinal pathogens. |
Genes associated with lymphocyte differentiation | IL23R | Activation and development of the Th17 lineage and its effects on dendritic cells and macrophages leading to the production of various pro-inflammatory molecules. |
Genes encoding interleukins | IL-10 | An anti-inflammatory cytokine that inhibits the production of pro-inflammatory cytokines. |
Genes encoding the protein | TNFSF15 | The TNFSF15 gene product (TL1A) is a TNF-like factor that is expressed in endothelial cells, macrophages and lymphocytes of the intestinal lamina propria. |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jarmakiewicz-Czaja, S.; Zielińska, M.; Sokal, A.; Filip, R. Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update. Genes 2022, 13, 2388. https://doi.org/10.3390/genes13122388
Jarmakiewicz-Czaja S, Zielińska M, Sokal A, Filip R. Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update. Genes. 2022; 13(12):2388. https://doi.org/10.3390/genes13122388
Chicago/Turabian StyleJarmakiewicz-Czaja, Sara, Magdalena Zielińska, Aneta Sokal, and Rafał Filip. 2022. "Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update" Genes 13, no. 12: 2388. https://doi.org/10.3390/genes13122388