Inherited determinants of Crohn's disease and ulcerative colitis phenotypes: a genetic association study

Isabelle Cleynen, Gabrielle Boucher, Luke Jostins, L Philip Schumm, Sebastian Zeissig, Tariq Ahmad, Vibeke Andersen, Jane M Andrews, Vito Annese, Stephan Brand, Steven R Brant, Judy H Cho, Mark J Daly, Marla Dubinsky, Richard H Duerr, Lynnette R Ferguson, Andre Franke, Richard B Gearry, Philippe Goyette, Hakon Hakonarson, Jonas Halfvarson, Johannes R Hov, Hailang Huang, Nicholas A Kennedy, Limas Kupcinskas, Ian C Lawrance, James C Lee, Jack Satsangi, Stephan Schreiber, Emilie Théâtre, Andrea E van der Meulen-de Jong, Rinse K Weersma, David C Wilson, International Inflammatory Bowel Disease Genetics Consortium, Miles Parkes, Severine Vermeire, John D Rioux, John Mansfield, Mark S Silverberg, Graham Radford-Smith, Dermot P B McGovern, Jeffrey C Barrett, Charlie W Lees, Clara Abraham, Jean-Paul Achkar, Tariq Ahmad, Leila Amininejad, Ashwin N Ananthakrishnan, Vibeke Andersen, Carl A Anderson, Jane M Andrews, Vito Annese, Guy Aumais, Leonard Baidoo, Robert N Baldassano, Peter A Bampton, Murray Barclay, Jeffrey C Barrett, Theodore M Bayless, Johannes Bethge, Joshua C Bis, Alain Bitton, Gabrielle Boucher, Stephan Brand, Berenice Brandt, Steven R Brant, Carsten Büning, Angela Chew, Judy H Cho, Isabelle Cleynen, Ariella Cohain, Anthony Croft, Mark J Daly, Mauro D'Amato, Silvio Danese, Dirk De Jong, Martine De Vos, Goda Denapiene, Lee A Denson, Kathy Devaney, Olivier Dewit, Renata D'Inca, Marla Dubinsky, Richard H Duerr, Cathryn Edwards, David Ellinghaus, Jonah Essers, Lynnette R Ferguson, Eleonora A Festen, Philip Fleshner, Tim Florin, Denis Franchimont, Andre Franke, Karin Fransen, Richard Gearry, Michel Georges, Christian Gieger, Jürgen Glas, Philippe Goyette, Todd Green, Anne M Griffiths, Stephen L Guthery, Hakon Hakonarson, Jonas Halfvarson, Katherine Hanigan, Talin Haritunians, Ailsa Hart, Chris Hawkey, Nicholas K Hayward, Matija Hedl, Paul Henderson, Xinli Hu, Hailiang Huang, Ken Y Hui, Marcin Imielinski, Andrew Ippoliti, Laimas Jonaitis, Luke Jostins, Tom H Karlsen, Nicholas A Kennedy, Mohammed Azam Khan, Gediminas Kiudelis, Krupa Krishnaprasad, Subra Kugathasan, Limas Kupcinskas, Anna Latiano, Debby Laukens, Ian C Lawrance, James C Lee, Charlie W Lees, Marcis Leja, Johan Van Limbergen, Paolo Lionetti, Jimmy Z Liu, Edouard Louis, Gillian Mahy, John Mansfield, Dunecan Massey, Christopher G Mathew, Dermot P B McGovern, Raquel Milgrom, Mitja Mitrovic, Grant W Montgomery, Craig Mowat, William Newman, Aylwin Ng, Siew C Ng, Sok Meng Evelyn Ng, Susanna Nikolaus, Kaida Ning, Markus Nöthen, Ioannis Oikonomou, Orazio Palmieri, Miles Parkes, Anne Phillips, Cyriel Y Ponsioen, Urõs Potocnik, Natalie J Prescott, Deborah D Proctor, Graham Radford-Smith, Jean-Francois Rahier, Soumya Raychaudhuri, Miguel Regueiro, Florian Rieder, John D Rioux, Stephan Ripke, Rebecca Roberts, Richard K Russell, Jeremy D Sanderson, Miquel Sans, Jack Satsangi, Eric E Schadt, Stefan Schreiber, L Philip Schumm, Regan Scott, Mark Seielstad, Yashoda Sharma, Mark S Silverberg, Lisa A Simms, Jurgita Skieceviciene, Sarah L Spain, A Hillary Steinhart, Joanne M Stempak, Laura Stronati, Jurgita Sventoraityte, Stephan R Targan, Kirstin M Taylor, Anje ter Velde, Emilie Theatre, Leif Torkvist, Mark Tremelling, Andrea van der Meulen, Suzanne van Sommeren, Eric Vasiliauskas, Severine Vermeire, Hein W Verspaget, Thomas Walters, Kai Wang, Ming-Hsi Wang, Rinse K Weersma, Zhi Wei, David Whiteman, Cisca Wijmenga, David C Wilson, Juliane Winkelmann, Ramnik J Xavier, Sebastian Zeissig, Bin Zhang, Clarence K Zhang, Hu Zhang, Wei Zhang, Hongyu Zhao, Zhen Z Zhao, Isabelle Cleynen, Gabrielle Boucher, Luke Jostins, L Philip Schumm, Sebastian Zeissig, Tariq Ahmad, Vibeke Andersen, Jane M Andrews, Vito Annese, Stephan Brand, Steven R Brant, Judy H Cho, Mark J Daly, Marla Dubinsky, Richard H Duerr, Lynnette R Ferguson, Andre Franke, Richard B Gearry, Philippe Goyette, Hakon Hakonarson, Jonas Halfvarson, Johannes R Hov, Hailang Huang, Nicholas A Kennedy, Limas Kupcinskas, Ian C Lawrance, James C Lee, Jack Satsangi, Stephan Schreiber, Emilie Théâtre, Andrea E van der Meulen-de Jong, Rinse K Weersma, David C Wilson, International Inflammatory Bowel Disease Genetics Consortium, Miles Parkes, Severine Vermeire, John D Rioux, John Mansfield, Mark S Silverberg, Graham Radford-Smith, Dermot P B McGovern, Jeffrey C Barrett, Charlie W Lees, Clara Abraham, Jean-Paul Achkar, Tariq Ahmad, Leila Amininejad, Ashwin N Ananthakrishnan, Vibeke Andersen, Carl A Anderson, Jane M Andrews, Vito Annese, Guy Aumais, Leonard Baidoo, Robert N Baldassano, Peter A Bampton, Murray Barclay, Jeffrey C Barrett, Theodore M Bayless, Johannes Bethge, Joshua C Bis, Alain Bitton, Gabrielle Boucher, Stephan Brand, Berenice Brandt, Steven R Brant, Carsten Büning, Angela Chew, Judy H Cho, Isabelle Cleynen, Ariella Cohain, Anthony Croft, Mark J Daly, Mauro D'Amato, Silvio Danese, Dirk De Jong, Martine De Vos, Goda Denapiene, Lee A Denson, Kathy Devaney, Olivier Dewit, Renata D'Inca, Marla Dubinsky, Richard H Duerr, Cathryn Edwards, David Ellinghaus, Jonah Essers, Lynnette R Ferguson, Eleonora A Festen, Philip Fleshner, Tim Florin, Denis Franchimont, Andre Franke, Karin Fransen, Richard Gearry, Michel Georges, Christian Gieger, Jürgen Glas, Philippe Goyette, Todd Green, Anne M Griffiths, Stephen L Guthery, Hakon Hakonarson, Jonas Halfvarson, Katherine Hanigan, Talin Haritunians, Ailsa Hart, Chris Hawkey, Nicholas K Hayward, Matija Hedl, Paul Henderson, Xinli Hu, Hailiang Huang, Ken Y Hui, Marcin Imielinski, Andrew Ippoliti, Laimas Jonaitis, Luke Jostins, Tom H Karlsen, Nicholas A Kennedy, Mohammed Azam Khan, Gediminas Kiudelis, Krupa Krishnaprasad, Subra Kugathasan, Limas Kupcinskas, Anna Latiano, Debby Laukens, Ian C Lawrance, James C Lee, Charlie W Lees, Marcis Leja, Johan Van Limbergen, Paolo Lionetti, Jimmy Z Liu, Edouard Louis, Gillian Mahy, John Mansfield, Dunecan Massey, Christopher G Mathew, Dermot P B McGovern, Raquel Milgrom, Mitja Mitrovic, Grant W Montgomery, Craig Mowat, William Newman, Aylwin Ng, Siew C Ng, Sok Meng Evelyn Ng, Susanna Nikolaus, Kaida Ning, Markus Nöthen, Ioannis Oikonomou, Orazio Palmieri, Miles Parkes, Anne Phillips, Cyriel Y Ponsioen, Urõs Potocnik, Natalie J Prescott, Deborah D Proctor, Graham Radford-Smith, Jean-Francois Rahier, Soumya Raychaudhuri, Miguel Regueiro, Florian Rieder, John D Rioux, Stephan Ripke, Rebecca Roberts, Richard K Russell, Jeremy D Sanderson, Miquel Sans, Jack Satsangi, Eric E Schadt, Stefan Schreiber, L Philip Schumm, Regan Scott, Mark Seielstad, Yashoda Sharma, Mark S Silverberg, Lisa A Simms, Jurgita Skieceviciene, Sarah L Spain, A Hillary Steinhart, Joanne M Stempak, Laura Stronati, Jurgita Sventoraityte, Stephan R Targan, Kirstin M Taylor, Anje ter Velde, Emilie Theatre, Leif Torkvist, Mark Tremelling, Andrea van der Meulen, Suzanne van Sommeren, Eric Vasiliauskas, Severine Vermeire, Hein W Verspaget, Thomas Walters, Kai Wang, Ming-Hsi Wang, Rinse K Weersma, Zhi Wei, David Whiteman, Cisca Wijmenga, David C Wilson, Juliane Winkelmann, Ramnik J Xavier, Sebastian Zeissig, Bin Zhang, Clarence K Zhang, Hu Zhang, Wei Zhang, Hongyu Zhao, Zhen Z Zhao

Abstract

Background: Crohn's disease and ulcerative colitis are the two major forms of inflammatory bowel disease; treatment strategies have historically been determined by this binary categorisation. Genetic studies have identified 163 susceptibility loci for inflammatory bowel disease, mostly shared between Crohn's disease and ulcerative colitis. We undertook the largest genotype association study, to date, in widely used clinical subphenotypes of inflammatory bowel disease with the goal of further understanding the biological relations between diseases.

Methods: This study included patients from 49 centres in 16 countries in Europe, North America, and Australasia. We applied the Montreal classification system of inflammatory bowel disease subphenotypes to 34,819 patients (19,713 with Crohn's disease, 14,683 with ulcerative colitis) genotyped on the Immunochip array. We tested for genotype-phenotype associations across 156,154 genetic variants. We generated genetic risk scores by combining information from all known inflammatory bowel disease associations to summarise the total load of genetic risk for a particular phenotype. We used these risk scores to test the hypothesis that colonic Crohn's disease, ileal Crohn's disease, and ulcerative colitis are all genetically distinct from each other, and to attempt to identify patients with a mismatch between clinical diagnosis and genetic risk profile.

Findings: After quality control, the primary analysis included 29,838 patients (16,902 with Crohn's disease, 12,597 with ulcerative colitis). Three loci (NOD2, MHC, and MST1 3p21) were associated with subphenotypes of inflammatory bowel disease, mainly disease location (essentially fixed over time; median follow-up of 10·5 years). Little or no genetic association with disease behaviour (which changed dramatically over time) remained after conditioning on disease location and age at onset. The genetic risk score representing all known risk alleles for inflammatory bowel disease showed strong association with disease subphenotype (p=1·65 × 10(-78)), even after exclusion of NOD2, MHC, and 3p21 (p=9·23 × 10(-18)). Predictive models based on the genetic risk score strongly distinguished colonic from ileal Crohn's disease. Our genetic risk score could also identify a small number of patients with discrepant genetic risk profiles who were significantly more likely to have a revised diagnosis after follow-up (p=6·8 × 10(-4)).

Interpretation: Our data support a continuum of disorders within inflammatory bowel disease, much better explained by three groups (ileal Crohn's disease, colonic Crohn's disease, and ulcerative colitis) than by Crohn's disease and ulcerative colitis as currently defined. Disease location is an intrinsic aspect of a patient's disease, in part genetically determined, and the major driver to changes in disease behaviour over time.

Funding: International Inflammatory Bowel Disease Genetics Consortium members funding sources (see Acknowledgments for full list).

Copyright © 2016 Cleynen et al. Open Access article distributed under the terms of CC BY. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Evolution of clinical subphenotypes (A) Proportion of patients with Crohn's disease who have inflammatory (Montreal classification B1), stricturing (B2), or penetrating (B3) disease over time from diagnosis to most recent follow-up. (B) Proportion of patients with Crohn's disease who have ileal (L1), colonic (L2), or ileocolonic (L3) disease over time from diagnosis to most recent follow-up. (C) Survival plot of time from diagnosis of Crohn's disease to resectional surgery stratified by disease location. (D) Survival plot of time from diagnosis of ulcerative colitis to colectomy stratified by disease extent (extensive disease, E3; non-extensive disease, E1 and E2).
Figure 2
Figure 2
Effect of single nucleotide polymorphisms, HLA alleles, and polygenic risk scores on phenotypes of inflammatory bowel disease (A) Effect sizes for genotype–phenotype associations for risk of Crohn's disease and ulcerative colitis (odds ratio relative to controls), Crohn's disease location (odds ratio of ileal vs colonic disease), Crohn's disease behaviour (proportional odds ratio), disease extent of ulcerative colitis (odds ratio of extensive vs non-extensive disease), and age at diagnosis (linear coefficients) for MST1, MHC, and NOD2 variants. All effect sizes are per allele, and are adjusted for associations with correlated phenotypes by including them as additional predictors in the regression model, along with principal components to control for stratification. See appendix A for more details on these regression models. Genome-wide significant associations are depicted by filled circles, and error bars depict 95% CIs. (B) Effect sizes of genetic risk scores for disease location, disease behaviour, and age at diagnosis including all 163 susceptibility loci. Effect sizes are calculated by linear regression of the risk score against the phenotype, adjusted for the effect of the other phenotypes and for principal components, and error bars depict 95% CIs. Filled circles represent effects that are significant after correcting for 15 phenotype-score combinations (p<0·003). Effect sizes are measured on scales standardised to unit variance (and thus represent the number of standard deviations that the mean phenotype increases by per standard deviation increase in the risk score).
Figure 3
Figure 3
Violin plot showing the genetic substructure of inflammatory bowel disease location The violin represents the range of the log CD versus UC score for the indicated subphenotype (calculated with the R package “vioplot”), with dots representing the mean of that group and error bars the 95% CIs. Although the effects are small compared with the variation within groups, the mean effects can still be measured accurately (right side of the figure). It can be seen on this figure that the Crohn's disease versus ulcerative colitis (CD vs UC) risk score placed colonic Crohn's disease between ileal Crohn's disease and ulcerative colitis. The plot also shows the positioning of the intermediate phenotypes (ileocolonic Crohn's disease and inflammatory bowel disease unclassified [IBD-U]) in between ileal and colonic Crohn's disease, and ulcerative colitis and colonic Crohn's disease, respectively.
Figure 4
Figure 4
Histograms of Crohn's disease versus ulcerative colitis (CD vs UC) genetic risk score in patients with inflammatory bowel disease Risk scores created from the 163 known inflammatory bowel disease risk loci with per-locus contributions estimated to maximally distinguish all Crohn's disease from ulcerative colitis. Distributions of ulcerative colitis samples are shown in blue, ileal Crohn's disease samples in green, and colonic Crohn's disease with hatched lines (middle area in dark green shows overlap of blue and green distributions). The overlap of all three distributions shows the shared genetic aetiology of inflammatory bowel disease, and the intermediate position of colonic Crohn's disease between ulcerative colitis and ileal Crohn's disease shows that it is genetically distinct from the others. Vertical dashed lines show boundaries for outlier analysis: ulcerative colitis cases above 2 were selected as being likely Crohn's disease and Crohn's disease cases below −2 as likely ulcerative colitis.

References

    1. Molodecky NA, Soon IS, Rabi DM. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46–54. e42.
    1. Neovius M, Arkema EV, Blomqvist P. Patients with ulcerative colitis miss more days of work than the general population, even following colectomy. Gastroenterology. 2013;144:536–543.
    1. Everhart JE, Ruhl CE. Burden of digestive diseases in the United States part I: overall and upper gastrointestinal diseases. Gastroenterology. 2009;136:376–386.
    1. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature. 2011;474:307–317.
    1. Kirschner BS. Ulcerative colitis in children. Pediatr Clin North Am. 1996;43:235–254.
    1. Moum B, Ekbom A, Vatn MH. Inflammatory bowel disease: re-evaluation of the diagnosis in a prospective population based study in south eastern Norway. Gut. 1997;40:328–332.
    1. Pera A, Bellando P, Caldera D. Colonoscopy in inflammatory bowel disease. Diagnostic accuracy and proposal of an endoscopic score. Gastroenterology. 1987;92:181–185.
    1. Satsangi J, Silverberg MS, Vermeire S. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006;55:749–753.
    1. Silverberg MS, Satsangi J, Ahmad T. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a working party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol. 2005;19(suppl A):5–36.
    1. Crohn BB, Ginzburg L, Oppenheimer GD. Regional ileitis; a pathologic and clinical entity. JAMA. 1932;99:1323–1329.
    1. Dalziel TK. Thomas Kennedy Dalziel 1861–1924. Chronic interstitial enteritis. Dis Colon Rectum. 1989;32:1076–1078.
    1. Silverberg MS, Mirea L, Bull SB. A population- and family-based study of Canadian families reveals association of HLA DRB1*0103 with colonic involvement in inflammatory bowel disease. Inflamm Bowel Dis. 2003;9:1–9.
    1. Cuthbert AP, Fisher SA, Mirza MM. The contribution of NOD2 gene mutations to the risk and site of disease in inflammatory bowel disease. Gastroenterology. 2002;122:867–874.
    1. Ahmad T, Armuzzi A, Bunce M. The molecular classification of the clinical manifestations of Crohn's disease. Gastroenterology. 2002;122:854–866.
    1. Brant SR, Picco MF, Achkar JP. Defining complex contributions of NOD2/CARD15 gene mutations, age at onset, and tobacco use on Crohn's disease phenotypes. Inflamm Bowel Dis. 2003;9:281–289.
    1. Cleynen I, Gonzalez JR, Figueroa C. Genetic factors conferring an increased susceptibility to develop Crohn's disease also influence disease phenotype: results from the IBDchip European Project. Gut. 2013;62:1556–1565.
    1. de la Concha EG, Fernandez-Arquero M, Lopez-Nava G. Susceptibility to severe ulcerative colitis is associated with polymorphism in the central MHC gene IKBL. Gastroenterology. 2000;119:1491–1495.
    1. Economou M, Trikalinos TA, Loizou KT, Tsianos EV, Ioannidis JPA. Differential effects of NOD2 variants on Crohn's disease risk and phenotype in diverse populations: a meta analysis. Am J Gastroenterol. 2004;99:2393–2404.
    1. Newman B, Silverberg MS, Gu X. CARD15 and HLA DRB1 alleles influence susceptibility and disease localization in Crohn's disease. Am J Gastroenterol. 2004;99:306–315.
    1. Dignass A, Lindsay JO, Sturm A. Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 2: current management. J Crohns Colitis. 2012;6:991–1030.
    1. Dignass A, Van Assche G, Lindsay JO. The second European evidence-based consensus on the diagnosis and management of Crohn's disease: current management. J Crohns Colitis. 2010;4:28–62.
    1. Mowat C, Cole A, Windsor A. Guidelines for the management of inflammatory bowel disease in adults. Gut. 2011;60:571–607.
    1. Hanauer SB, Feagan BG, Lichtenstein GR. Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial. Lancet. 2002;359:1541–1549.
    1. Hanauer SB, Sandborn WJ, Rutgeerts P. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology. 2006;130:323–333.
    1. Reinisch W, Sandborn WJ, Hommes DW. Adalimumab for induction of clinical remission in moderately to severely active ulcerative colitis: results of a randomised controlled trial. Gut. 2011;60:780–787.
    1. Rutgeerts P, Sandborn WJ, Feagan BG. Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2005;353:2462–2476.
    1. Sandborn WJ, van Assche G, Reinisch W. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2012;142:257–265. e1–3.
    1. Feagan BG, Rutgeerts P, Sands BE. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013;369:699–710.
    1. Sandborn WJ, Feagan BG, Rutgeerts P. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2013;369:711–721.
    1. Frolkis AD, Dykeman J, Negron ME. Risk of surgery for inflammatory bowel diseases has decreased over time: a systematic review and meta-analysis of population-based studies. Gastroenterology. 2013;145:996–1006.
    1. Solberg IC, Lygren I, Jahnsen J. Clinical course during the first 10 years of ulcerative colitis: results from a population-based inception cohort (IBSEN Study) Scand J Gastroenterol. 2009;44:431–440.
    1. Solberg IC, Vatn MH, Hoie O. Clinical course in Crohn's disease: results of a Norwegian population-based ten-year follow-up study. Clin Gastroenterol Hepatol. 2007;5:1430–1438.
    1. Jostins L, Ripke S, Weersma RK. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–124.
    1. Lennard-Jones JE. Classification of inflammatory bowel disease. Scand J Gastroenterol Suppl. 1989;170:2–6. discussion 16–19.
    1. Goyette P, Boucher G, Mallon D. High-density mapping of the MHC identifies a shared role for HLA-DRB1*01:03 in inflammatory bowel diseases and heterozygous advantage in ulcerative colitis. Nat Genet. 2015;7:172–179.
    1. Purcell S, Neale B, Todd-Brown K. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–575.
    1. Jostins L, Levine AP, Barrett JC. Using genetic prediction from known complex disease loci to guide the design of next-generation sequencing experiments. PLoS One. 2013;8:e76328.
    1. Barrett JC, Hansoul S, Nicolae DL. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet. 2008;40:955–962.
    1. Faubion WA, Jr, Loftus EV, Jr, Harmsen WS. The natural history of corticosteroid therapy for inflammatory bowel disease: a population-based study. Gastroenterology. 2001;121:255–260.
    1. Halfvarson J, Bodin L, Tysk C. Inflammatory bowel disease in a Swedish twin cohort: a long-term follow-up of concordance and clinical characteristics. Gastroenterology. 2003;124:1767–1773.
    1. Henckaerts L, Van Steen K, Verstreken I. Genetic risk profiling and prediction of disease course in Crohn's disease patients. Clin Gastroenterol Hepatol. 2009;7:972–980. e2.
    1. Donaldson PT, Farrant JM, Wilkinson ML. Dual association of HLA DR2 and DR3 with primary sclerosing cholangitis. Hepatology. 1991;13:129–133.
    1. Spurkland A, Saarinen S, Boberg KM. HLA class II haplotypes in primary sclerosing cholangitis patients from five European populations. Tissue Antigens. 1999;53:459–469.
    1. Loftus EV, Jr, Harewood GC, Loftus CG. PSC-IBD: a unique form of inflammatory bowel disease associated with primary sclerosing cholangitis. Gut. 2005;54:91–96.
    1. Hamshere ML, O'Donovan MC, Jones IR. Polygenic dissection of the bipolar phenotype. Br J Psychiatry. 2011;198:284–288.
    1. Chen GB, Lee SH, Brion MJ. Estimation and partitioning of (co)heritability of inflammatory bowel disease from GWAS and immunochip data. Hum Mol Genet. 2014;23:4710–4720.
    1. Morgan XC, Tickle TL, Sokol H. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79.

Source: PubMed

3
購読する