Editorial Open Access
Copyright ©2006 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Aug 14, 2006; 12(30): 4784-4787
Published online Aug 14, 2006. doi: 10.3748/wjg.v12.i30.4784
Contribution of genetics to a new vision in the understanding of inflammatory bowel disease
AS Peña, Departments of Gastroenterology and Pathology, VU University Medical Centre, Amsterdam, The Netherlands
Author contributions: All authors contributed equally to the work.
Correspondence to: Professor AS Peña, MD, PhD, FRCP, AGAF, VU University Medical Center, Head, Laboratory of Immunogenetics, PO Box 7057, 1007 MB Amsterdam, The Netherlands. as.pena@vumc.nl
Telephone: +31-20-4444705 Fax: +31-20-4444737
Received: March 25, 2006
Revised: March 30, 2006
Accepted: April 24, 2006
Published online: August 14, 2006

Abstract

Inflammatory bowel diseases (IBD), such as Crohn’s disease (CD) and ulcerative colitis (UC), are chronic inflammatory autoimmune conditions of the gastrointestinal tract. Other organs, such as the eyes, skin and articulations, are often affected and IBD may be accompanied by other diseases of autoimmune origin. There is no single etiological factor responsible for the onset of IBD. Recent advances in genetics and in the molecular mechanisms of the proteins coded by these genes have given rise to a new vision in understanding these complex diseases. Activation of specific genes that affect antigen presentation and the handling of cells by innate immunity may lead to autoimmunity with the consequent activation of the major histocompatibility complex (MHC) and multiple cytokines involved in the regulation of acquired immunity. In this review IBD is described as a constellation of diseases that can best be classified as barrier diseases. This vision, developed by Kiel in Germany, includes the idea that changes in our environment due to the westernization of civilization have not been met with adaptation of the innate immune system, and this has given rise to autoimmune diseases. These diseases affect 1-5 of 1000 individuals and represent a major burden on the national health systems of many countries on different continents. On a world scale, a major challenge is to generate interventions to prevent the development of these diseases in Asia, Latin America and Africa.

Key Words: Inflammatory bowel disease, Crohn’s disease, Ulcerative colitis, Genetics, Autoimmunity, Major histocompatibility complex



INTRODUCTION

In less than five years after the discovery of the first gene involved in susceptibility to Crohn’s disease (CD), the initial concept that CD is a multifactorial and polygenic disease has been consolidated [1,2].

Although the detailed functions of the NOD2 or CARD15 gene of chromosome 16 is not entirely clear, their relationship to intestinal flora, Toll receptors (TLR), and other intra-cytoplasmatic receptors of the NOD family together with their relation with NF-κB, has made clear that the innate immune response is of paramount importance in the pathogenesis of CD[3-5].

Two years ago it was suggested that the relationship between TLR-2 and NOD2 genes could explain the balance between activation of superficial receptors of the epithelial and dendritic cells to stimulate the production of NF-κB and NOD2 as inhibitors in order to prevent chronic disease[6-8]. According to this concept, individuals with mutations in the CARD15 gene are not able to control the intestinal inflammation and this induces a TH1 immune response[9]. Recently, however, mutations in the terminal N of the gene in a region rich with leucine repeats have been transfected into the NOD2 region of mice[10]. Surprisingly, when these mice are challenged with muramil dipeptide (MDP) they generate considerable production of NF-κB and IL-1B[10]. Also the transfection of these mutations in HEK293 cells results in increased transcription of TNF-α, which suggests that different genes contribute to susceptibility to the disease and differences in manifestations of CD[11].

The fact that NOD2 is preferentially expressed in Paneth cells in the ileocaecal region probably explains the strong association between these mutations and this disease localization[12,13]. This region is rich in defensins, which are natural antibiotics that contribute to the mucosal barrier and innate immunity[14]. It is also now known that patients with mutations in the CARD15 gene also have less α-defensins. This defect probably contributes to the role of intestinal flora in inducing and/or maintaining inflammation[15]. Preliminary results of a study carried out by the Stange group in Germany suggest that defensin deficiency is genetically determined. The defensin family is more complex and richer than originally described and its distribution varies within the gastrointestinal tract. The β-defensins are localized in the colon and defective β-defensins could contribute to the colonic localization of IBD[16]. It is also possible that different defensins may be specific to CD and UC variations[17].

These observations demonstrate the fine-tuning of molecular biological responses of the gastrointestinal tract and the complexity of interactions among different genes on different chromosomes. Another example of complexity is gene polymorphisms of the gene inhibitor of plasminogen (PAI-1) in combination with CARD15 mutations have an influence in the development of CD. Those patients who have mutations of CARD15 and carry the PAI-14G/4G genotype develop a stricturizing phenotype (OR, 4.64; 95% CI, 1.26-17.05)[18].

Despite the well-demonstrated and replicated role of CARD15 mutations in the susceptibility for CD in the majority of Caucasian populations[19-23], no mutations have been found in Asian[24-27] and in several European populations. In Scotland, Ireland, Galicia, Sweden and Finland the carriership of mutations in the CARD15 gene is less frequent[28-32]. An interesting phenomenon was recently reported in monozygotic twins. In Sweden, the number of carriers of these mutations was as low as in the general population[32]. However, in Denmark 40% of the monozygotic twins carried CARD15 mutations, which was a higher rate than in the Danish population with CD[33]. This corroborates the observations in Finland where the 1007fs allele frequency was higher in familial CD than in non-familial cases with CD (10.9% vs 3.5%; P < 0.01)[30]. These observations underscore the incidence of genetic variability and the importance of studying healthy controls in the general population.

RELEVANCE OF OTHER GENES

Several other genes on other chromosomes are involved in determining susceptibility to CD. Two interesting genes, one on chromosome 5 and one on chromosome 10, contribute to the new vision of the genetics of CD. On chromosome 5, the SLC22A4/SLC22A5 haplotype codes for molecules involved in cationic transport, other solutes and carnitine (OCT-1 y OCT2)[34-36]. In some populations, an epistatic interaction has been found to exist between the CARD15 mutations and the 250 KB region of 5q31[37,38]. The other interesting gene in this context is the DLG5 on chromosome 10, which is a gene that is important in the scaffolding of the epithelial cell[39,40].

The 250 KB region of 5q31 and the HLA region of 6p21 contain several genes that are of paramount importance in the regulation of the immune response and probably contribute to the phenotype of the patient with IBD. The HLA-DRB1*0103 allele is associated with UC and with the colonic localization of CD[41,42].

Carriers of HLA-DRB1*0103, B*35 and B*27 have higher risk for arthralgias/arthritis in some of the greater articulations and HLA-B*44 carriers are at higher risk for symmetric poly-arthritis[43,44].

A meta-analysis of 1068 CD patients has also implicated the genes described above in the development of CD and this study identified other regions of potential relevance on chromosomes 2q, 3q, 17q and 19q[45].

THE CONTRIBUTION OF GENETICS TO A NEW VISION

In the new vision of CD, several genes are involved in the maintenance of the intestinal barrier, such as scaffolding genes (DLG5[46]), genes involved in the transport of key molecules for the homeostasis or exclusion of toxins (SLC[47] and MDR1[48,49]), genes involving the sensing of bacteria, both on the surface (TLR, CD14) and intracytoplamatically (CARD15, CARD4 and CARD8[50-52]. Whether a specific gene for regulating permeability exists, as has been suggested on chromosome 19, remains to be demonstrated. The combination of gene rich regions of chromosome 5 and 6 involved in regulating the immune response may contribute to the phenotype of the disease.

In summary, in less than a decade the genetics of IBD has evolved from epidemiology to molecular biology, and from observational studies to functional studies. The challenge for the coming years is the discovery of gene-gene interactions and gene-environment interactions.

A delineation of phenotypes based on genetic and molecular mechanisms will improve diagnoses and more accurate prognoses. This knowledge, together with advances in the understanding of the phenomenon of tolerance and the disruption of this mechanism in the understanding of chronic inflammation of the gastrointestinal tract, will lead to the design of better and novel therapeutic strategies. This should be the basis for effective drug development in addition to increasing knowledge generated by pharmacogenetics and pharmacogenomics.

Regarding the role of the environment in those individuals with a genetic susceptibility, new findings on epigenetic effects from long-term follow-up in monozygotic twins will open a new area of investigation. Investigators at the Spanish National Cancer Centre (CNIO) in Madrid have shown that older monozygotic twins exhibited differences in their overall content and genomic distribution of 5-methylcytosine DNA and histone acetylation. These epigenetic changes affect their gene-expression patterns and may explain the well-known disease discordance in these cases of monozygotic twins[53].

Footnotes

S- Editor Pan BR L- Editor Lutze M E- Editor Ma WH

References
1.  Hugot JP, Chamaillard M, Zouali H, Lesage S, Cézard JP, Belaiche J, Almer S, Tysk C, O'Morain CA, Gassull M. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature. 2001;411:599-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3986]  [Cited by in F6Publishing: 3811]  [Article Influence: 165.7]  [Reference Citation Analysis (0)]
2.  Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001;411:603-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3555]  [Cited by in F6Publishing: 3393]  [Article Influence: 147.5]  [Reference Citation Analysis (0)]
3.  Peñate M, Peña AS. Relevance of the innate immune system. Rev Esp Enferm Dig. 2001;93:721-739.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Peña AS, Peñate M. Genetic susceptibility and regulation of inflammation in Crohn's disease. Relationship with the innate immune system. Rev Esp Enferm Dig. 2002;94:351-360.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Inohara C, Nuñez G. NOD-LRR proteins: role in host-microbial interactions and inflammatory disease. Annu Rev Biochem. 2005;74:355-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 728]  [Cited by in F6Publishing: 762]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
6.  Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol. 2004;5:800-808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 615]  [Cited by in F6Publishing: 589]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
7.  Netea MG, Kullberg BJ, de Jong DJ, Franke B, Sprong T, Naber TH, Drenth JP, Van der Meer JW. NOD2 mediates anti-inflammatory signals induced by TLR2 ligands: implications for Crohn's disease. Eur J Immunol. 2004;34:2052-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 184]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
8.  O'Neill LA. How NOD-ing off leads to Crohn disease. Nat Immunol. 2004;5:776-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
9.  Neurath MF, Finotto S, Glimcher LH. The role of Th1/Th2 polarization in mucosal immunity. Nat Med. 2002;8:567-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 451]  [Cited by in F6Publishing: 474]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
10.  Maeda S, Hsu LC, Liu H, Bankston LA, Iimura M, Kagnoff MF, Eckmann L, Karin M. Nod2 mutation in Crohn's disease potentiates NF-kappaB activity and IL-1beta processing. Science. 2005;307:734-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 630]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
11.  Linderson Y, Bresso F, Buentke E, Pettersson S, D'Amato M. Functional interaction of CARD15/NOD2 and Crohn's disease-associated TNFalpha polymorphisms. Int J Colorectal Dis. 2005;20:305-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
12.  Cho JH. The Nod2 gene in Crohn's disease: implications for future research into the genetics and immunology of Crohn's disease. Inflamm Bowel Dis. 2001;7:271-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 33]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
13.  Cho JH. Significant role of genetics in IBD: the NOD2 gene. Rev Gastroenterol Disord. 2003;3 Suppl 1:S18-S22.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Wehkamp J, Schwind B, Herrlinger KR, Baxmann S, Schmidt K, Duchrow M, Wohlschläger C, Feller AC, Stange EF, Fellermann K. Innate immunity and colonic inflammation: enhanced expression of epithelial alpha-defensins. Dig Dis Sci. 2002;47:1349-1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
15.  Wehkamp J, Salzman NH, Porter E, Nuding S, Weichenthal M, Petras RE, Shen B, Schaeffeler E, Schwab M, Linzmeier R. Reduced Paneth cell alpha-defensins in ileal Crohn's disease. Proc Natl Acad Sci USA. 2005;102:18129-18134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 749]  [Cited by in F6Publishing: 706]  [Article Influence: 37.2]  [Reference Citation Analysis (0)]
16.  Wehkamp J, Fellermann K, Herrlinger KR, Baxmann S, Schmidt K, Schwind B, Duchrow M, Wohlschläger C, Feller AC, Stange EF. Human beta-defensin 2 but not beta-defensin 1 is expressed preferentially in colonic mucosa of inflammatory bowel disease. Eur J Gastroenterol Hepatol. 2002;14:745-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 154]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
17.  Wehkamp J, Harder J, Weichenthal M, Mueller O, Herrlinger KR, Fellermann K, Schroeder JM, Stange EF. Inducible and constitutive beta-defensins are differentially expressed in Crohn's disease and ulcerative colitis. Inflamm Bowel Dis. 2003;9:215-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 228]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
18.  Alvarez-Lobos M, Arostegui JI, Sans M, Tassies D, Plaza S, Delgado S, Lacy AM, Pique JM, Yagüe J, Panés J. Crohn's disease patients carrying Nod2/CARD15 gene variants have an increased and early need for first surgery due to stricturing disease and higher rate of surgical recurrence. Ann Surg. 2005;242:693-700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 144]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
19.  Hampe J, Cuthbert A, Croucher PJ, Mirza MM, Mascheretti S, Fisher S, Frenzel H, King K, Hasselmeyer A, MacPherson AJ. Association between insertion mutation in NOD2 gene and Crohn's disease in German and British populations. Lancet. 2001;357:1925-1928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 769]  [Cited by in F6Publishing: 762]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
20.  Murillo L, Crusius JB, van Bodegraven AA, Alizadeh BZ, Peña AS. CARD15 gene and the classification of Crohn's disease. Immunogenetics. 2002;54:59-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 64]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
21.  Mendoza JL, Murillo LS, Fernández L, Peña AS, Lana R, Urcelay E, Cruz-Santamaría DM, de la Concha EG, Díaz-Rubio M, García-Paredes J. Prevalence of mutations of the NOD2/CARD15 gene and relation to phenotype in Spanish patients with Crohn disease. Scand J Gastroenterol. 2003;38:1235-1240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
22.  Vermeire S, Wild G, Kocher K, Cousineau J, Dufresne L, Bitton A, Langelier D, Pare P, Lapointe G, Cohen A. CARD15 genetic variation in a Quebec population: prevalence, genotype-phenotype relationship, and haplotype structure. Am J Hum Genet. 2002;71:74-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 206]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
23.  Cho JH. Advances in the genetics of inflammatory bowel disease. Curr Gastroenterol Rep. 2004;6:467-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
24.  Inoue N, Tamura K, Kinouchi Y, Fukuda Y, Takahashi S, Ogura Y, Inohara N, Núñez G, Kishi Y, Koike Y. Lack of common NOD2 variants in Japanese patients with Crohn's disease. Gastroenterology. 2002;123:86-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 348]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
25.  Yamazaki K, Takazoe M, Tanaka T, Kazumori T, Nakamura Y. Absence of mutation in the NOD2/CARD15 gene among 483 Japanese patients with Crohn's disease. J Hum Genet. 2002;47:469-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 216]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
26.  Sugimura M, Kinouchi Y, Takahashi S, Aihara H, Takagi S, Negoro K, Obana N, Kojima Y, Matsumoto K, Kikuchi T. CARD15/NOD2 mutational analysis in Japanese patients with Crohn's disease. Clin Genet. 2003;63:160-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
27.  Economou M, Trikalinos TA, Loizou KT, Tsianos EV, Ioannidis JP. Differential effects of NOD2 variants on Crohn's disease risk and phenotype in diverse populations: a metaanalysis. Am J Gastroenterol. 2004;99:2393-2404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 334]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
28.  Arnott ID, Nimmo ER, Drummond HE, Fennell J, Smith BR, MacKinlay E, Morecroft J, Anderson N, Kelleher D, O'Sullivan M. NOD2/CARD15, TLR4 and CD14 mutations in Scottish and Irish Crohn's disease patients: evidence for genetic heterogeneity within Europe? Genes Immun. 2004;5:417-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 168]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
29.  Núñez C, Barreiro M, Domínguez-Muñoz JE, Lorenzo A, Zapata C, Peña AS. CARD15 mutations in patients with Crohn's disease in a homogeneous Spanish population. Am J Gastroenterol. 2004;99:450-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
30.  Heliö T, Halme L, Lappalainen M, Fodstad H, Paavola-Sakki P, Turunen U, Färkkilä M, Krusius T, Kontula K. CARD15/NOD2 gene variants are associated with familially occurring and complicated forms of Crohn's disease. Gut. 2003;52:558-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 206]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
31.  Ideström M, Rubio C, Granath F, Finkel Y, Hugot JP. CARD15 mutations are rare in Swedish pediatric Crohn disease. J Pediatr Gastroenterol Nutr. 2005;40:456-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
32.  Halfvarson J, Bresso F, D'Amato M, Järnerot G, Pettersson S, Tysk C. CARD15/NOD2 polymorphisms do not explain concordance of Crohn's disease in Swedish monozygotic twins. Dig Liver Dis. 2005;37:768-772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
33.  Vind I, Vieira A, Hougs L, Tavares L, Riis L, Andersen PS, Locht H, Freitas J, Monteiro E, Christensen IJ. NOD2/CARD15 gene polymorphisms in Crohn's disease: a genotype-phenotype analysis in Danish and Portuguese patients and controls. Digestion. 2005;72:156-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
34.  Rioux JD, Daly MJ, Silverberg MS, Lindblad K, Steinhart H, Cohen Z, Delmonte T, Kocher K, Miller K, Guschwan S. Genetic variation in the 5q31 cytokine gene cluster confers susceptibility to Crohn disease. Nat Genet. 2001;29:223-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 566]  [Cited by in F6Publishing: 597]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
35.  Peltekova VD, Wintle RF, Rubin LA, Amos CI, Huang Q, Gu X, Newman B, Van Oene M, Cescon D, Greenberg G. Functional variants of OCTN cation transporter genes are associated with Crohn disease. Nat Genet. 2004;36:471-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 572]  [Cited by in F6Publishing: 551]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
36.  Urban TJ, Giacomini KM, Risch N. Haplotype structure and ethnic-specific allele frequencies at the OCTN locus: implications for the genetics of Crohn's disease. Inflamm Bowel Dis. 2005;11:78-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
37.  Giallourakis C, Stoll M, Miller K, Hampe J, Lander ES, Daly MJ, Schreiber S, Rioux JD. IBD5 is a general risk factor for inflammatory bowel disease: replication of association with Crohn disease and identification of a novel association with ulcerative colitis. Am J Hum Genet. 2003;73:205-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 128]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
38.  Negoro K, McGovern DP, Kinouchi Y, Takahashi S, Lench NJ, Shimosegawa T, Carey A, Cardon LR, Jewell DP, van Heel DA. Analysis of the IBD5 locus and potential gene-gene interactions in Crohn's disease. Gut. 2003;52:541-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 87]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
39.  Stoll M, Corneliussen B, Costello CM, Waetzig GH, Mellgard B, Koch WA, Rosenstiel P, Albrecht M, Croucher PJ, Seegert D. Genetic variation in DLG5 is associated with inflammatory bowel disease. Nat Genet. 2004;36:476-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 367]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
40.  Yamazaki K, Takazoe M, Tanaka T, Ichimori T, Saito S, Iida A, Onouchi Y, Hata A, Nakamura Y. Association analysis of SLC22A4, SLC22A5 and DLG5 in Japanese patients with Crohn disease. J Hum Genet. 2004;49:664-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 82]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
41.  Ahmad T, Armuzzi A, Bunce M, Mulcahy-Hawes K, Marshall SE, Orchard TR, Crawshaw J, Large O, de Silva A, Cook JT. The molecular classification of the clinical manifestations of Crohn's disease. Gastroenterology. 2002;122:854-866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 426]  [Cited by in F6Publishing: 446]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
42.  Fernandez L, Mendoza JL, Martinez A, Urcelay E, Fernandez-Arquero M, Garcia-Paredes J, Peña AS, Diaz-Rubio M, de la Concha EG. IBD1 and IBD3 determine location of Crohn's disease in the Spanish population. Inflamm Bowel Dis. 2004;10:715-722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 25]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
43.  Orchard TR, Wordsworth BP, Jewell DP. Peripheral arthropathies in inflammatory bowel disease: their articular distribution and natural history. Gut. 1998;42:387-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 370]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
44.  Orchard TR, Thiyagaraja S, Welsh KI, Wordsworth BP, Hill Gaston JS, Jewell DP. Clinical phenotype is related to HLA genotype in the peripheral arthropathies of inflammatory bowel disease. Gastroenterology. 2000;118:274-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 189]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
45.  van Heel DA, Fisher SA, Kirby A, Daly MJ, Rioux JD, Lewis CM. Inflammatory bowel disease susceptibility loci defined by genome scan meta-analysis of 1952 affected relative pairs. Hum Mol Genet. 2004;13:763-770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 189]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
46.  Török HP, Glas J, Tonenchi L, Lohse P, Müller-Myhsok B, Limbersky O, Neugebauer C, Schnitzler F, Seiderer J, Tillack C. Polymorphisms in the DLG5 and OCTN cation transporter genes in Crohn's disease. Gut. 2005;54:1421-1427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 118]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
47.  Newman B, Siminovitch KA. Recent advances in the genetics of inflammatory bowel disease. Curr Opin Gastroenterol. 2005;21:401-407.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Ho GT, Nimmo ER, Tenesa A, Fennell J, Drummond H, Mowat C, Arnott ID, Satsangi J. Allelic variations of the multidrug resistance gene determine susceptibility and disease behavior in ulcerative colitis. Gastroenterology. 2005;128:288-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 141]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
49.  Wilk JN, Bilsborough J, Viney JL. The mdr1a-/- mouse model of spontaneous colitis: a relevant and appropriate animal model to study inflammatory bowel disease. Immunol Res. 2005;31:151-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 54]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
50.  van Heel DA, Ghosh S, Butler M, Hunt K, Foxwell BM, Mengin-Lecreulx D, Playford RJ. Synergistic enhancement of Toll-like receptor responses by NOD1 activation. Eur J Immunol. 2005;35:2471-2476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 122]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
51.  van Heel DA, Ghosh S, Hunt KA, Mathew CG, Forbes A, Jewell DP, Playford RJ. Synergy between TLR9 and NOD2 innate immune responses is lost in genetic Crohn's disease. Gut. 2005;54:1553-1557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 99]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
52.  Netea MG, Ferwerda G, de Jong DJ, Jansen T, Jacobs L, Kramer M, Naber TH, Drenth JP, Girardin SE, Kullberg BJ. Nucleotide-binding oligomerization domain-2 modulates specific TLR pathways for the induction of cytokine release. J Immunol. 2005;174:6518-6523.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005;102:10604-10609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2436]  [Cited by in F6Publishing: 2134]  [Article Influence: 112.3]  [Reference Citation Analysis (0)]