Genetics of the innate immune response in inflammatory bowel disease†
Corresponding Author
Johan Van Limbergen MD, MRCPCH
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK EH91LF
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UKSearch for more papers by this authorRichard K. Russell MRCPH
Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK EH91LF
Search for more papers by this authorElaine R. Nimmo BSc, MSc, PhD
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorGwo-Tzer Ho MRCP
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorIan D. Arnott MD
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorDavid C. Wilson MD, FRCPCH
Department of Child Life and Health, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK EH91LF
Search for more papers by this authorJack Satsangi DPhil, FRCPCH
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorCorresponding Author
Johan Van Limbergen MD, MRCPCH
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK EH91LF
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UKSearch for more papers by this authorRichard K. Russell MRCPH
Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK EH91LF
Search for more papers by this authorElaine R. Nimmo BSc, MSc, PhD
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorGwo-Tzer Ho MRCP
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorIan D. Arnott MD
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorDavid C. Wilson MD, FRCPCH
Department of Child Life and Health, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK EH91LF
Search for more papers by this authorJack Satsangi DPhil, FRCPCH
Gastrointestinal Unit, Molecular Medicine Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
Search for more papers by this authorThe authors declare they have no competing interests.
Abstract
The discovery of nucleotide-binding oligomerization domain 2/caspase recruitment domain-containing protein 15 (NOD2/CARD15) as the first susceptibility gene in Crohn's disease (CD) has shifted the focus of research into the pathogenesis of inflammatory bowel disease (IBD) firmly to the innate immune response and the integrity of the epithelial barrier. The subsequent implication in IBD of variant alleles of OCTN, DLG5, MDR1, and TLRs has provided further support for a new, more complex model of innate immunity function in the gastrointestinal tract. In this review, we examine the recent advances in our understanding of the influence of genetics of the innate immune response on IBD. We will focus on germline variation of genes encoding pathogen-recognition receptors, proteins involved in epithelial homeostasis and secreted antimicrobial proteins.
(Inflamm Bowel Dis 2007)
REFERENCES
- 1 Shirazi T, Longman RJ, Corfield AP, et al. Mucins and inflammatory bowel disease. Postgrad Med J. 2000; 76: 473–478.
- 2 Van der Sluis M, De Koning BA, et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology. 2006; 131: 117–129.
- 3 Einerhand AW, Renes IB, Makkink MK, et al. Role of mucins in inflammatory bowel disease: important lessons from experimental models. Eur J Gastroenterol Hepatol. 2002; 14: 757–765.
- 4 Duraisamy S, Ramasamy S, Kharbanda S, et al. Distinct evolution of the human carcinoma-associated transmembrane mucins, MUC1, MUC4 AND MUC16. Gene. 2006; 373: 28–34.
- 5 Porchet N, Aubert JP. MUC genes: mucin or not mucin? That is the question. Med Sci. 2004; 569–574.
- 6 Pigny P, Guyonnet-Duperat V, Hill AS, et al. Human mucin genes assigned to 11p15.5: identification and organization of a cluster of genes. Genomics. 1996; 38: 340–352.
- 7 van Heel DA, Fisher SA, Kirby A, et al. Inflammatory bowel disease susceptibility loci defined by genome scan meta-analysis of 1952 affected relative pairs. Hum Mol Genet. 2004; 13: 763–770.
- 8 Satsangi J, Parkes M, Louis E, et al. Two stage genome-wide search in inflammatory bowel disease provides evidence for susceptibility loci on chromosomes 3, 7 and 12. Nat Genet. 1996; 14: 199–202.
- 9 Koscinski I, Viville S, Porchet N, et al. MUC4 gene polymorphism and expression in women with implantation failure. Hum Reprod. 2006;e-pub 28-6/2006.
- 10 Van Klinken BJ, Dekker J, Buller HA, et al. Biosynthesis of mucins (MUC2–6) along the longitudinal axis of the human gastrointestinal tract. Am J Physiol. 1997; 273: G296–G302.
- 11 Chang SK, Dohrman AF, Basbaum CB, et al. Localization of mucin (MUC2 and MUC3) messenger RNA and peptide expression in human normal intestine and colon cancer. Gastroenterology. 1994; 107: 28–36.
- 12 Velcich A, Yang W, Heyer J, et al. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 2002; 295: 1726–1729.
- 13 Yang W, Velcich A, Lozonschi I, et al. Inactivation of p21WAF1/cip1 enhances intestinal tumor formation in Muc2−/− mice. Am J Pathol. 2005; 166: 1239–1246.
- 14 Van Klinken BJ, Dekker J, van Gool SA, et al. MUC5B is the prominent mucin in human gallbladder and is also expressed in a subset of colonic goblet cells. Am J Physiol. 1998; 274: G871–G878.
- 15 Ho SB, Roberton AM, Shekels LL, et al. Expression cloning of gastric mucin complementary DNA and localization of mucin gene expression. Gastroenterology. 1995; 109: 735–747.
- 16 Toribara NW, Roberton AM, Ho SB, et al. Human gastric mucin. Identification of a unique species by expression cloning. J Biol Chem. 1993; 268: 5879–5885.
- 17 Gendler SJ, Spicer AP. Epithelial mucin genes. Annu Rev Physiol. 1995; 57: 607–634.
- 18 Pullan RD, Thomas GAO, Rhodes M, et al. Thickness of adherent mucus gel on colonic mucosa in humans and its relevance to colitis. Gut. 1994; 35: 353–359.
- 19 Dwarakanath AD, Campbell BJ, Tsai HH, et al. Faecal mucinase activity assessed in inflammatory bowel disease using 14C threonine labelled mucin substrate. Gut. 1995; 37: 58–62.
- 20 Tsai HH, Dwarakanath AD, Hart CA, et al. Increased faecal mucin sulphatase activity in ulcerative colitis: a potential target for treatment. Gut. 1995; 36: 570–576.
- 21 Rhodes JM. Unifying hypothesis for inflammatory bowel disease and associated colon cancer: sticking the pieces together with sugar. Lancet. 1996; 347: 40–44.
- 22 Bodger K, Halfvarson J, Dodson AR, et al. Altered colonic glycoprotein expression in unaffected monozygotic twins of inflammatory bowel disease patients. Gut. 2006; 55: 973–977.
- 23 Tysk C, Riedesel H, Lindberg E, et al. Colonic glycoproteins in monozygotic twins with inflammatory bowel disease. Gastroenterology. 1991; 100: 419–423.
- 24 Van S, I, Pigny P, Perrais M, et al. Transcriptional regulation of the 11p15 mucin genes. Towards new biological tools in human therapy, in inflammatory diseases and cancer? Front Biosci. 2001; 6: D1216–D1234.
- 25 Campbell BJ, Rowe GE, Leiper K, et al. Increasing the intra-Golgi pH of cultured LS174T goblet-differentiated cells mimics the decreased mucin sulfation and increased Thomsen-Friedenreich antigen (Gal beta1−3GalNac alpha−) expression seen in colon cancer. Glycobiology. 2001; 11: 385–393.
- 26 Gouyer V, Wiede A, Buisine MP, et al. Specific secretion of gel-forming mucins and TFF peptides in HT-29 cells of mucin-secreting phenotype. Biochim Biophys Acta. 2001; 1539: 71–84.
- 27 Enss ML, Cornberg M, Wagner S, et al. Proinflammatory cytokines trigger MUC gene expression and mucin release in the intestinal cancer cell line LS180. Inflamm Res. 2000; 49: 162–169.
- 28 Gaudier E, Jarry A, Blottiere HM, et al. Butyrate specifically modulates MUC gene expression in intestinal epithelial goblet cells deprived of glucose. Am J Physiol Gastrointest Liver Physiol. 2004; 287: G1168–G1174.
- 29 Mack DR, Ahrne S, Hyde L, et al. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut. 2003; 52: 827–833.
- 30 Hoebler C, Gaudier E, de Coppet P, et al. MUC genes are differently expressed during onset and maintenance of inflammation in dextran sodium sulfate-treated mice. Dig Dis Sci. 2006; 51: 381–389.
- 31 Wright NA, Pike C, Elia G. Induction of a novel epidermal growth factor-secreting cell lineage by mucosal ulceration in human gastrointestinal stem cells. Nature. 1990; 343: 82–85.
- 32 Schwerbrock NM, Makkink MK, Van der Sluis M, et al. Interleukin 10-deficient mice exhibit defective colonic Muc2 synthesis before and after induction of colitis by commensal bacteria. Inflamm Bowel Dis. 2004; 10: 811–823.
- 33 Faure M, Mettraux C, Moennoz D, et al. Specific amino acids increase mucin synthesis and microbiota in dextran sulfate sodium-treated rats. J Nutr. 2006; 136: 1558–1564.
- 34 Kleessen B, Blaut M. Modulation of gut mucosal biofilms. Br J Nutr. 2005; 93: S35–S40.
- 35 Smirnov A, Perez R, Amit-Romach E, et al. Mucin dynamics and microbial populations in chicken small intestine are changed by dietary probiotic and antibiotic growth promoter supplementation. J Nutr. 2005; 135: 187–192.
- 36 Ho SB, Dvorak LA, Moor RE, et al. Cysteine-rich domains of Muc3 intestinal mucin promote cell migration, inhibit apoptosis, and accelerate wound healing. Gastroenterology. 2006; 131: 1501–1517.
- 37 Tytgat KM, Van der Wal JW, Einerhand AW, et al. Quantitative analysis of MUC2 synthesis in ulcerative colitis. Biochem Biophys Res Commun. 1996; 224: 397–405.
- 38 Tytgat KM, Opdam FJ, Einerhand AW, et al. MUC2 is the prominent colonic mucin expressed in ulcerative colitis. Gut. 1996; 38: 554–563.
- 39 Van Klinken BJ, Van der Wal JW, Einerhand AW, et al. Sulphation and secretion of the predominant secretory human colonic mucin MUC2 in ulcerative colitis. Gut. 1999; 44: 387–393.
- 40 Myerscough N, Warren B, Gough M, et al. Expression of mucin genes in ulcerative colitis. Biochem Soc Trans. 1995; 23: 536S.
- 41 Buisine MP, Desreumaux P, Debailleul V, et al. Abnormalities in mucin gene expression in Crohn's disease. Inflamm Bowel Dis. 1999; 5: 24–32.
- 42 Buisine MP, Desreumaux P, Leteurtre E, et al. Mucin gene expression in intestinal epithelial cells in Crohn's disease. Gut. 2001; 49: 544–551.
- 43 Longman RJ, Douthwaite J, Sylvester PA, et al. Coordinated localisation of mucins and trefoil peptides in the ulcer associated cell lineage and the gastrointestinal mucosa. Gut. 2000; 47: 792–800.
- 44 Shaoul R, Okada Y, Cutz E, et al. Colonic expression of MUC2, MUC5AC, and TFF1 in inflammatory bowel disease in children. J Pediatr Gastroenterol Nutr. 2004; 38: 488–493.
- 45 Kyo K, Parkes M, Takei Y, et al. Association of ulcerative colitis with rare VNTR alleles of the human intestinal mucin gene, MUC3. Hum Mol Genet. 1999; 8: 307–311.
- 46 Kyo K, Muto T, Nagawa H, et al. Associations of distinct variants of the intestinal mucin gene MUC3A with ulcerative colitis and Crohn's disease. J Hum Genet. 2001; 46: 5–20.
- 47 Schreiber S. Slipping the barrier: how variants in CARD15 could alter permeability of the intestinal wall and population health. Gut. 2006; 55: 308–309.
- 48 Hugot JP, Chamaillard M, Zouali H, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature. 2001; 411: 599–603.
- 49 Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001; 411: 603–606.
- 50 Hampe J, Cuthbert A, Croucher PJ, et al. Association between insertion mutation in NOD2 gene and Crohn's disease in German and British populations. Lancet. 2001; 357: 1925–1928.
- 51 Rosenstiel P, Fantini M, Brautigam K, et al. TNF-alpha and IFN-gamma regulate the expression of the NOD2 (CARD15) gene in human intestinal epithelial cells. Gastroenterology. 2003; 124: 1001–1009.
- 52 Hisamatsu T, Suzuki M, Podolsky DK. Interferon-gamma augments CARD4/NOD1 gene and protein expression through interferon regulatory factor-1 in intestinal epithelial cells. J Biol Chem. 2003; 278: 32962–32968.
- 53 Hisamatsu T, Suzuki M, Reinecker HC, et al. CARD15/NOD2 functions as an antibacterial factor in human intestinal epithelial cells. Gastroenterology. 2003; 124: 993–1000.
- 54 Abreu MT, Fukata M, Arditi M. TLR Signaling in the gut in health and disease. J Immunol. 2005; 174: 4453–4460.
- 55 Gitter AH, Bendfeldt K, Schmitz H, et al. Epithelial barrier defects in HT-29/B6 colonic cell monolayers induced by tumor necrosis factor-alpha. Ann NY Acad Sci. 2000; 915: 193–203.
- 56 Ye D, Ma I, Ma TY. Molecular mechanism of tumor necrosis factor-alpha modulation of intestinal epithelial tight junction barrier. Am J Physiol Gastrointest Liver Physiol. 2006; 290: G496–G504.
- 57 Gitter AH, Bendfeldt K, Schulzke JD, et al. Leaks in the epithelial barrier caused by spontaneous and TNF-α-induced single-cell apoptosis. FASEB J. 2000; 14: 1749–1753.
- 58 Bruewer M, Luegering A, Kucharzik T, et al. Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol. 2003; 171: 6164–6172.
- 59 Laukoetter MG, Bruewer M, Nusrat A. Regulation of the intestinal epithelial barrier by the apical junctional complex. Curr Opin Gastroenterol. 2006; 22: 85–89.
- 60 Soderholm JD, Streutker C, Yang PC, et al. Increased epithelial uptake of protein antigens in the ileum of Crohn's disease mediated by tumour necrosis factor alpha. Gut. 2004; 53: 1817–1824.
- 61 Heller F, Florian P, Bojarski C, et al. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology. 2005; 129: 550–564.
- 62 Zeissig S, Bojarski C, Buergel N, et al. Downregulation of epithelial apoptosis and barrier repair in active Crohn's disease by tumour necrosis factor alpha antibody treatment. Gut. 2004; 53: 1295–1302.
- 63 Madsen KL, Malfair D, Gray D, et al. Interleukin-10 gene-deficient mice develop a primary intestinal permeability defect in response to enteric microflora. Inflamm Bowel Dis. 1999; 5: 262–270.
- 64 Kuhn R, Lohler J, Rennick D, et al. Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993; 75: 263–274.
- 65 Panwala CM, Jones JC, Viney JL. A novel model of inflammatory bowel disease: mice deficient for the multiple drug resistance gene, mdr1a, spontaneously develop colitis. J Immunol. 1998; 161: 5733–5744.
- 66 Hermiston ML, Gordon JI. Inflammatory bowel disease and adenomas in mice expressing a dominant negative N-cadherin. Science. 1995; 270: 1203–1207.
- 67 Nancey S, Holvoet S, Graber I, et al. CD8+ cytotoxic T cells induce relapsing colitis in normal mice. Gastroenterology. 2006; 131: 485–496.
- 68 Westendorf AM, Fleissner D, Deppenmeier S, et al. Autoimmune-mediated intestinal inflammation-impact and regulation of antigen-specific CD8+ T cells. Gastroenterology. 2006; 131: 510–524.
- 69 DeMeo MT, Mutlu EA, Keshavarzian A, et al. Intestinal permeation and gastrointestinal disease. J Clin Gastroenterol. 2002; 34: 385–396.
- 70 May GR, Sutherland LR, Meddings JB. Is small intestinal permeability really increased in relatives of patients with Crohn's disease? Gastroenterology. 1993; 104: 1627–1632.
- 71 Katz KD, Hollander D, Vadheim CM, et al. Intestinal permeability in patients with Crohn's disease and their healthy relatives. Gastroenterology. 1989; 97: 927–931.
- 72 Teahon K, Smethurst P, Levi AJ, et al. Intestinal permeability in patients with Crohn's disease and their first degree relatives. Gut. 1992; 33: 320–323.
- 73 Peeters M, Geypens B, Claus D, et al. Clustering of increased small intestinal permeability in families with Crohn's disease. Gastroenterology. 1997; 113: 802–807.
- 74 Soderholm JD, Peterson KH, Olaison G, et al. Epithelial permeability to proteins in the noninflamed ileum of Crohn's disease? Gastroenterology. 1999; 117: 65–72.
- 75 Soderholm JD, Olaison G, Lindberg E, et al. Different intestinal permeability patterns in relatives and spouses of patients with Crohn's disease: an inherited defect in mucosal defence? Gut. 1999; 44: 96–100.
- 76 Gitter AH, Wullstein F, Fromm M, et al. Epithelial barrier defects in ulcerative colitis: characterization and quantification by electrophysiological imaging. Gastroenterology. 2001; 121: 1320–1328.
- 77 Thjodleifsson B, Sigthorsson G, Cariglia N, et al. Subclinical intestinal inflammation: an inherited abnormality in Crohn's disease relatives? Gastroenterology. 2003; 124: 1728–1737.
- 78 Buhner S, Buning C, Genschel J, et al. Genetic basis for increased intestinal permeability in families with Crohn's disease: role of CARD15 3020insC mutation? Gut. 2006; 55: 342–347.
- 79 Begue B, Dumant C, Bambou JC, et al. Microbial induction of CARD15 expression in intestinal epithelial cells via toll-like receptor 5 triggers an antibacterial response loop. J Cell Physiol. 2006; 241–252.
- 80 Annese V, Valvano MR, Palmieri O, et al. Multidrug resistance 1 gene in inflammatory bowel disease: a meta-analysis. World J Gastroenterol. 2006; 12: 3636–3644.
- 81 Schwab M, Schaeffeler E, Marx C, et al. Association between the C3435T MDR1 gene polymorphism and susceptibility for ulcerative colitis. Gastroenterology. 2003; 124: 26–33.
- 82 Glas J, Torok HP, Schiemann U, et al. MDR1 gene polymorphism in ulcerative colitis. Gastroenterology. 2004; 126(1): 367.
- 83 Ho GT, Nimmo ER, Tenesa A, et al. Allelic variations of the multidrug resistance gene determine susceptibility and disease behavior in ulcerative colitis. Gastroenterology. 2005; 128: 288–296.
- 84 Croucher PJ, Mascheretti S, Foelsch UR, et al. Lack of association between the C3435T MDR1 gene polymorphism and inflammatory bowel disease in two independent Northern European populations. Gastroenterology. 2003; 125: 1919–1920.
- 85 Brant SR, Panhuysen CI, Nicolae D, et al. MDR1 Ala893 polymorphism is associated with inflammatory bowel disease. Am J Hum Genet. 2003; 73: 1282–1292.
- 86 Potočnik U, Ferkolj I, Glavac D, et al. Polymorphisms in multidrug resistance 1 (MDR1) gene are associated with refractory Crohn disease and ulcerative colitis. Genes Immun. 2004; 5: 530–539.
- 87 Palmieri O, Latiano A, Valvano R, et al. Multidrug resistance 1 gene polymorphisms are not associated with inflammatory bowel disease and response to therapy in Italian patients. Aliment Pharmacol Ther. 2005; 22: 1129–1138.
- 88 Urcelay E, Mendoza JL, Martin MC, et al. MDR1 gene: susceptibility in Spanish Crohn's disease and ulcerative colitis patients. Inflamm Bowel Dis. 2006; 12: 33–37.
- 89 Oostenbrug LE, Dijkstra G, Nolte IM, et al. Absence of association between the multidrug resistance (MDR1) gene and inflammatory bowel disease. Scand J Gastroenterol. 2006; 41: 1174–1182.
- 90 Onnie CM, Fisher SA, Pattni R, et al. Associations of allelic variants of the multidrug resistance gene (ABCB1 or MDR1) and inflammatory bowel disease and their effects on disease behavior: a case-control and meta-analysis study. Inflamm Bowel Dis. 2006; 12: 263–271.
- 91 Ho GT, Soranzo N, Nimmo ER, et al. ABCB1/MDR1 gene determines susceptibility and phenotype in ulcerative colitis: discrimination of critical variants using a gene-wide haplotype tagging approach. Hum Mol Genet. 2006; 15: 797–805.
- 92 Langmann T, Schmitz G. Loss of detoxification in inflammatory bowel disease. Nat Clin Pract Gastroenterol Hepatol. 2006; 3: 358–359.
- 93 Langmann T, Moehle C, Mauerer R, et al. Loss of detoxification in inflammatory bowel disease: dysregulation of pregnane × receptor target genes. Gastroenterology. 2004; 127: 26–40.
- 94 Staudinger JL, Goodwin B, Jones SA, et al. The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity. Proc Natl Acad Sci USA. 2001; 98: 3369–3374.
- 95 Chrencik JE, Orans J, Moore LB, et al. Structural disorder in the complex of human pregnane × receptor and the macrolide antibiotic rifampicin. Mol Endocrinol. 2005; 1125–1134.
- 96 Schuetz EG, Strom S, Yasuda K, et al. Disrupted bile acid homeostasis reveals an unexpected interaction among nuclear hormone receptors, transporters, and cytochrome P450. J Biol Chem. 2001; 276: 39411–39418.
- 97 Willson TM, Kliewer SA. PXR, CAR and drug metabolism. Nat Rev Drug Disc. 2002; 1: 259–266.
- 98 Kliewer SA, Goodwin B, Willson TM. The nuclear pregnane × receptor: a key regulator of xenobiotic metabolism. Endocr Rev. 2002; 23: 687–702.
- 99 Dring MM, Goulding CA, Trimble VI, et al. The pregnane × receptor locus is associated with susceptibility to inflammatory bowel disease. Gastroenterology. 2006; 130: 341–348.
- 100 Ho GT, Soranzo N, Tate SK, et al. Lack of association of the pregnane × receptor (PXR/NR1I2) gene with inflammatory bowel disease: parallel allelic association study and gene wide haplotype analysis. Gut. 2006; 55: 1676–1677.
- 101 Stoll M, Corneliussen B, Costello CM, et al. Genetic variation in DLG5 is associated with inflammatory bowel disease. Nat Genet. 2004; 36: 476–480.
- 102 Hampe J, Schreiber S, Shaw SH, et al. A genomewide analysis provides evidence for novel linkages in inflammatory bowel disease in a large European cohort. Am J Hum Genet. 1999; 64: 808–816.
- 103 Vermeire S. Dlg5 and octn. Inflamm Bowel Dis. 2004; 10: 888–890.
- 104 Shah G, Brugada R, Gonzalez O, et al. The cloning, genomic organization and tissue expression profile of the human DLG5 gene. BMC Genom. 3(1):6,. 2002; 3: 6.
- 105 Noble CL, Nimmo ER, Drummond H, et al. DLG5 variants do not influence susceptibility to inflammatory bowel disease in the Scottish population. Gut. 2005; 54: 1416–1420.
- 106 Russell RK, Drummond HE, Nimmo ER, et al. Socio-economic status critically influences penetrance of the DLG5 113A variant in early-onset IBD: a novel gene-environmental interaction. Gastroenterology. 2006; 130: A53.
- 107 Tenesa A, Noble C, Satsangi J, et al. Association of DLG5 and inflammatory bowel disease across populations. Eur J Hum Genet. 2006; 14: 259–260.
- 108 Torok HP, Glas J, Tonenchi L, et al. Polymorphisms in the DLG5 and OCTN cation transporter genes in Crohn's disease. Gut. 2005; 54: 1421–1427.
- 109 Lakatos PL, Fischer S, Claes K, et al. DLG5 R30Q is not associated with IBD in Hungarian IBD patients but predicts clinical response to steroids in Crohn's disease. Inflamm Bowel Dis. 2006; 12: 362–368.
- 110 Ferraris A, Torres B, Knafelz D, et al. Relationship between CARD15, SLC22A4/5, and DLG5 polymorphisms and early-onset inflammatory bowel diseases: an Italian multicentric study. Inflamm Bowel Dis. 2006; 12: 355–361.
- 111 Tremelling M, Waller S, Bredin F, et al. Genetic variants in TNF-alpha but not DLG5 are associated with inflammatory bowel disease in a large United Kingdom cohort. Inflamm Bowel Dis. 2006; 12: 178–184.
- 112 Buning C, Geerdts L, Fiedler T, et al. DLG5 variants in inflammatory bowel disease. Am J Gastroenterol. 2006; 101: 786–792.
- 113 Medici V, Mascheretti S, Croucher PJ, et al. Extreme heterogeneity in CARD15 and DLG5 Crohn disease-associated polymorphisms between German and Norwegian populations. Eur J Hum Genet. 2006; 14: 459–468.
- 114 Newman WG, Gu X, Wintle RF, et al. DLG5 variants contribute to Crohn disease risk in a Canadian population. Hum Mutat. 2006; 27: 353–358.
- 115 Friedrichs F, Brescianini S, Annese V, et al. Evidence of transmission ratio distortion of DLG5 R30Q variant in general and implication of an association with Crohn disease in men. Hum Genet. 2006; 119: 305–311.
- 116 Gazouli M, Mantzaris G, Archimandritis AJ, et al. Single nucleotide polymorphisms of OCTN1, OCTN2, and DLG5 genes in Greek patients with Crohn's disease. World J Gastroenterol. 2005; 11: 7525–7230.
- 117 Vermeire S, Pierik M, Hlavaty T, et al. Association of organic cation transporter risk haplotype with perianal penetrating Crohn's disease but not with susceptibility to IBD. Gastroenterology. 2005; 129: 1845–1853.
- 118 Daly MJ, Pearce AV, Farwell L, et al. Association of DLG5 R30Q variant with inflammatory bowel disease. Eur J Hum Genet. 2005; 13: 835–839.
- 119 Yamazaki K, Takazoe M, Tanaka T, et al. Association analysis of SLC22A4, SLC22A5 and DLG5 in Japanese patients with Crohn disease. J Hum Genet. 2004; 49: 664–668.
- 120 Rioux JD, Silverberg MS, Daly MJ, et al. Genomewide search in Canadian families with inflammatory bowel disease reveals two novel susceptibility loci. Am J Hum Genet. 2000; 66: 1863–1870.
- 121 Rioux JD, Daly MJ, Silverberg MS, et al. Genetic variation in the 5q31 cytokine gene cluster confers susceptibility to Crohn disease. Nat Genet. 2001; 29: 223–228.
- 122 Reinhard C, Rioux JD. Role of the IBD5 susceptibility locus in the inflammatory bowel diseases. Inflamm Bowel Dis. 2006; 12: 227–238.
- 123 Loots GG, Locksley RM, Blankespoor CM, et al. Identification of a coordinate regulator of interleukins 4, 13, and 5 by cross-species sequence comparisons. Science. 2000; 288: 136–140.
- 124 Noble CL, Nimmo ER, Drummond H, et al. The contribution of OCTN1/2 variants within the IBD5 locus to disease susceptibility and severity in Crohn's disease. Gastroenterology. 2005; 129: 1854–1864.
- 125 Giallourakis C, Stoll M, Miller K, et al. 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.
- 126 Mirza MM, Fisher SA, King K, et al. Genetic evidence for interaction of the 5q31 cytokine locus and the CARD15 gene in Crohn disease. Am J Hum Genet. 2003; 72: 1018–1022.
- 127 Negoro K, McGovern DP, Kinouchi Y, et al. Analysis of the IBD5 locus and potential gene–gene interactions in Crohn's disease. Gut. 2003; 52: 541–546.
- 128 Armuzzi A, Ahmad T, Ling KL, et al. Genotype–phenotype analysis of the Crohn's disease susceptibility haplotype on chromosome 5q31. Gut. 2003; 52: 1133–1139.
- 129 Urcelay E, Mendoza JL, Martinez A, et al. IBD5 polymorphisms in inflammatory bowel disease: association with response to infliximab. World J Gastroenterol. 2005; 11: 1187–1192.
- 130 Newman B, Gu X, Wintle R, et al. A risk haplotype in the Solute Carrier Family 22A4/22A5 gene cluster influences phenotypic expression of Crohn's disease. Gastroenterology. 2005; 128: 260–269.
- 131 Russell RK, Drummond HE, Nimmo ER, et al. Analysis of the influence of OCTN1/2 variants within the IBD5 locus on disease susceptibility and growth indices in early onset inflammatory bowel disease. Gut. 2006; 55: 1114–1123.
- 132 Babusukumar U, Wang T, McGuire E, et al. Contribution of OCTN variants within the IBD5 locus to pediatric onset Crohn's disease. Am J Gastroenterol. 2006; 101: 1354–1361.
- 133 McGovern DP, van Heel DA, Negoro K, et al. Further evidence of IBD5/CARD15 (NOD2) epistasis in the susceptibility to ulcerative colitis. Am J Hum Genet. 2003; 73: 1465–1466.
- 134 Peltekova VD, Wintle RF, Rubin LA, et al. Functional variants of OCTN cation transporter genes are associated with Crohn disease. Nat Genet. 2004; 36: 471–475.
- 135 Fisher SA, Hampe J, Onnie CM, et al. Direct or indirect association in a complex disease: the role of SLC22A4 and SLC22A5 functional variants in Crohn disease. Hum Mutat. 2006; 27: 778–785.
- 136 Van Bodegraven AA, Curley CR, Hunt KA, et al. Genetic variation in myosin IXB is associated with ulcerative colitis. Gastroenterology. 2006. E-pub ahead of print 12/9/06.
- 137 Monsuur AJ, de Bakker PI, Alizadeh BZ, et al. Myosin IXB variant increases the risk of celiac disease and points toward a primary intestinal barrier defect. Nat Genet. 2005; 37: 1341–1344.
- 138 Amundsen SS, Monsuur AJ, Wapenaar MC, et al. Association analysis of MYO9B gene polymorphisms with celiac disease in a Swedish/Norwegian cohort. Hum Immunol. 2006; 67: 341–345.
- 139 Hunt KA, Monsuur AJ, McArdle WL, et al. Lack of association of MYO9B genetic variants with coeliac disease in a British cohort. Gut. 2006; 55: 969–972.
- 140 Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006; 124: 783–801.
- 141 Seong SY, Matzinger P. Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses. Nat Rev Immunol. 2004; 4: 469–478.
- 142 Meylan E, Tschopp J, Karin M. Intracellular pattern recognition receptors in the host response. Nature. 2006; 442: 39–44.
- 143 Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003; 21: 335–376.
- 144
Sugiyama T,
Wright SD.
Soluble CD14 mediates efflux of phospholipids from cells.
J Immunol.
2001;
166:
826–831.
10.4049/jimmunol.166.2.826 Google Scholar
- 145 Pierik M, Joossens S, Van Steen K, et al. Toll-like receptor-1, -2, and -6 polymorphisms influence disease extension in inflammatory bowel diseases. Inflamm Bowel Dis. 2006; 12: 1–8.
- 146 Rector A, Lemey P, Laffut W, et al. Mannan-binding lectin (MBL) gene polymorphisms in ulcerative colitis and Crohn's disease. Genes Immun. 2001; 2: 323–328.
- 147 Seibold F, Konrad A, Flogerzi B, et al. Genetic variants of the mannan-binding lectin are associated with immune reactivity to mannans in Crohn's disease. Gastroenterology. 2004; 127: 1076–1084.
- 148 Joossens S, Pierik M, Rector A, et al. Mannan binding lectin (MBL) gene polymorphisms are not associated with anti-Saccharomyces cerevisiae (ASCA) in patients with Crohn's disease. Gut. 2006; 55: 746.
- 149 Creagh EM, O'Neill LAJ. TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol. 2006; 27: 352–357.
- 150 Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003; 425: 516–521.
- 151 Drenth JP, van der Meer JW. The inflammasome—a linebacker of innate defense. N Engl J Med. 2006; 355: 730–732.
- 152 Martinon F, Petrilli V, Mayor A, et al. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006; 440: 237–241.
- 153 Ting JP, Kastner DL, Hoffman HM. CATERPILLERs, pyrin and hereditary immunological disorders. Nat Rev Immunol. 2006; 6: 183–195.
- 154 Anderson KV, Bokla L, Nusslein-Volhard C. Establishment of dorsal-ventral polarity in the Drosophila embryo: the induction of polarity by the Toll gene product. Cell. 1985; 42: 791–798.
- 155 Medzhitov R, Preston-Hurlburt P, Janeway CA Jr. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997; 388: 394–397.
- 156 West AP, Koblansky AA, Ghosh S. Recognition and signaling by toll-like receptors. Annu Rev Cell Del Biol. 2006; 22: 409–437.
- 157 Poltorak A, He X, Smirnova I, et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science. 1998; 282: 2085–2088.
- 158 Hoshino K, Takeuchi O, Kawai T, et al. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the LPS gene product. J Immunol. 1999; 162: 3749–3752.
- 159 Mitchell JA, Fitzgerald KA, Coyle A, et al. TOLLing away in Brazil. Nat Immunol. 2006; 7: 675–679.
- 160 Bowie A, O'Neill LA. The interleukin-1 receptor/Toll-like receptor superfamily: signal generators for pro-inflammatory interleukins and microbial products. J Leuk Biol. 2000; 67: 508–514.
- 161 Stover AG, Da Silva CJ, Evans JT, et al. Structure–activity relationship of synthetic toll-like receptor 4 agonists. J Biol Chem. 2004; 279: 4440–4449.
- 162 Hristova K, Selsted ME, White SH. Critical role of lipid composition in membrane permeabilization by rabbit neutrophil defensins. J Biol Chem. 1997; 272: 24224–24233.
- 163 Koka RM, Huang E, Lieske JC. Adhesion of uric acid crystals to the surface of renal epithelial cells. Am J Physiol Renal Physiol. 2000; 278: F989–F998.
- 164 Ozinsky A, Underhill DM, Fontenot JD, et al. The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between toll-like receptors. Proc Natl Acad Sci USA. 2000; 97: 13766–13771.
- 165 Takeuchi O, Kawai T, Muhlradt PF, et al. Discrimination of bacterial lipoproteins by Toll-like receptor 6. Int Immunol. 2001; 13: 933–940.
- 166 Takeuchi O, Sato S, Horiuchi T, et al. Cutting edge: role of Toll-like receptor 1 in mediating immune response to microbial lipoproteins. J Immunol. 2002; 169: 10–14.
- 167 Travassos LH, Girardin SE, Philpott DJ, et al. Toll-like receptor 2-dependent bacterial sensing does not occur via peptidoglycan recognition. EMBO Rep. 2004; 5: 1000–1006.
- 168 Dziarski R, Gupta D. Staphylococcus aureus peptidoglycan is a toll-like receptor 2 activator: a reevaluation. Infect Immun. 2005; 73: 5212–5216.
- 169 Werts C, Tapping RI, Mathison JC, et al. Leptospiral lipopolysaccharide activates cells through a TLR2-dependent mechanism. Nat Immunol. 2001; 2: 346–352.
- 170 Asai Y, Hashimoto M, Fletcher HM, et al. Lipopolysaccharide preparation extracted from Porphyromonas gingivalis lipoprotein-deficient mutant shows a marked decrease in toll-like receptor 2-mediated signaling. Infect Immun. 2005; 73: 2157–2163.
- 171 Choe J, Kelker MS, Wilson IA. Crystal structure of human toll-like receptor 3 (TLR3) ectodomain. Science. 2005; 309: 581–585.
- 172 Bell JK, Askins J, Hall PR, et al. The dsRNA binding site of human Toll-like receptor 3. Proc Natl Acad Sci USA. 2006; 103: 8792–8797.
- 173 Cario E. Bacterial interactions with cells of the intestinal mucosa: Toll-like receptors and NOD2. Gut. 2005; 54: 1182–1193.
- 174 Liew FY, Xu D, Brint EK, O'Neill LA. Negative regulation of toll-like receptor-mediated immune responses. Nat Rev Immunol. 2005; 5: 446–458.
- 175 Gazouli M, Mantzaris G, Kotsinas A, et al. Association between polymorphisms in the Toll-like receptor 4, CD14, and CARD15/NOD2 and inflammatory bowel disease in the Greek population. World J Gastroenterol. 2005; 11: 681–685.
- 176 Klein W, Tromm A, Griga T, et al. A polymorphism in the CD14 gene is associated with Crohn disease. Scand J Gastroenterol. 2002; 37: 189–191.
- 177 Obana N, Takahashi S, Kinouchi Y, et al. Ulcerative colitis is associated with a promoter polymorphism of lipopolysaccharide receptor gene, CD14. Scand J Gastroenterol. 2002; 37: 699–704.
- 178 Peters KE, O'Callaghan NJ, Cavanaugh JA. Lack of association of the CD14 promoter polymorphism—159C/T with Caucasian inflammatory bowel disease. Scand J Gastroenterol. 2005; 40: 194–197.
- 179 Arnott IDR, Nimmo ER, Drummond HE, et al. 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.
- 180 Hayashi F, Smith KD, Ozinsky A, et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature. 2001; 410: 1099–1103.
- 181 Smith KD, Andersen-Nissen E, Hayashi F, et al. Toll-like receptor 5 recognizes a conserved site on flagellin required for protofilament formation and bacterial motility. Nat Immunol. 2003; 4: 1247–1253.
- 182 Andersen-Nissen E, Smith KD, Strobe KL, et al. Evasion of Toll-like receptor 5 by flagellated bacteria. Proc Natl Acad Sci USA. 2005; 102: 9247–9252.
- 183 Hemmi H, Takeuchi O, Kawai T, et al. A Toll-like receptor recognizes bacterial DNA. Nature. 2000; 408: 740–745.
- 184 Barton GM, Kagan JC, Medzhitov R. Intracellular localization of Toll-like receptor 9 prevents recognition of self DNA but facilitates access to viral DNA. Nat Immunol. 2006; 7: 49–56.
- 185 Fusunyan R, Nanthakumar N, Baldeon M, et al. Evidence for an innate immune response in the immature human intestine: Toll-like receptors on fetal enterocytes. Pediatr Res. 2001; 49: 589–593.
- 186 Hausmann M, Kiessling S, Mestermann S, et al. Toll-like receptors 2 and 4 are up-regulated during intestinal inflammation. Gastroenterology. 2002; 122: 1987–2000.
- 187 Abreu MT, Vora P, Faure E, et al. Decreased expression of Toll-like receptor-4 and MD-2 correlates with intestinal epithelial cell protection against dysregulated proinflammatory gene expression in response to bacterial lipopolysaccharide. J Immunol. 2001; 167: 1609–1616.
- 188 Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression of Toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun. 2000; 68: 7010–7017.
- 189 Gewirtz AT, Navas TA, Lyons S, et al. Cutting edge: bacterial flagellin activates basolaterally expressed TLR5 to induce epithelial proinflammatory gene expression. J Immunol. 2001; 167: 1882–1885.
- 190 Otte JM, Cario E, Podolsky DK. Mechanisms of cross hyporesponsiveness to toll-like receptor bacterial ligands in intestinal epithelial cells. Gastroenterology. 2004; 126: 1054–1070.
- 191 Bambou JC, Giraud A, Menard S, et al. In vitro and ex vivo activation of the TLR5 signaling pathway in intestinal epithelial cells by a commensal Escherichia coli strain. J Biol Chem. 2004; 279: 42984–42992.
- 192 Sanders CJ, Yu Y, Moore DA III, et al. Humoral immune response to flagellin requires T cells and activation of innate immunity. J Immunol. 2006; 177: 2810–2818.
- 193 Seya T, Oshiumi H, Sasai M, et al. TICAM-1 and TICAM-2: toll-like receptor adapters that participate in induction of type 1 interferons. International J Biochem Cell Biol. 2005; 37: 524–529.
- 194 Yamamoto M, Sato S, Hemmi H, et al. Essential role for TIRAP in activation of the signalling cascade shared by TLR2 and TLR4. Nature. 2002; 420(6913): 324–329.
- 195 Yamamoto M, Sato S, Mori K, et al. Cutting edge: a novel Toll/IL-1 receptor domain-containing adapter that preferentially activates the IFN-beta promoter in the Toll-like receptor signaling. J Immunol. 2002; 169: 6668–6672.
- 196 Carty M, Goodbody R, Schroder M, et al. The human adaptor SARM negatively regulates adaptor protein TRIF-dependent Toll-like receptor signaling. Nat Immunol. 2006;advanced online publication.
- 197 Takaoka A, Yanai H, Kondo S, et al. Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature. 2005; 434: 243–249.
- 198 Honda K, Ohba Y, Yanai H, et al. Spatiotemporal regulation of MyD88-IRF-7 signalling for robust type-I interferon induction. Nature. 2005; 434: 1035–1040.
- 199 Yamamoto M, Yamazaki S, Uematsu S, et al. Regulation of Toll/IL-1-receptor-mediated gene expression by the inducible nuclear protein IkappaBzeta. Nature. 2004; 430: 218–222.
- 200 Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004; 118: 229–241.
- 201 Strober W. Epithelial cells pay a Toll for protection. Nat Med. 2004; 10: 898–900.
- 202 Fort MM, Mozaffarian A, Stover AG, et al. A Synthetic TLR4 antagonist has anti-inflammatory effects in two murine models of inflammatory bowel disease. J Immunol. 2005; 174: 6416–6423.
- 203 Iwami Ki, Matsuguchi T, Masuda A, et al. Cutting edge: naturally occurring soluble form of mouse Toll-like receptor 4 inhibits lipopolysaccharide signaling. J Immunol. 2000; 165: 6682–6686.
- 204 LeBouder E, Rey-Nores JE, Rushmere NK, et al. Soluble forms of Toll-like receptor (TLR)2 capable of modulating TLR2 signaling are present in human plasma and breast milk. J Immunol. 2003; 171: 6680–6689.
- 205 Wald D, Qin J, Zhao Z, et al. SIGIRR, a negative regulator of Toll-like receptor-interleukin 1 receptor signaling. Nat Immunol. 2003; 4: 920–927.
- 206 Garlanda C, Riva F, Polentarutti N, et al. Intestinal inflammation in mice deficient in Tir8, an inhibitory member of the IL-1 receptor family. Proc Natl Acad Sci USA. 2004; 101: 3522–3526.
- 207 Qin J, Qian Y, Yao J, et al. SIGIRR inhibits interleukin-1 receptor- and toll-like receptor 4-mediated signaling through different mechanisms. J Biol Chem. 2005; 280: 25233–25241.
- 208 Adib-Conquy MP, Adrie CM, Fitting CB, et al. Up-regulation of MyD88s and SIGIRR, molecules inhibiting Toll-like receptor signaling, in monocytes from septic patients. Crit Care Med. 2006; 34: 2377–2385.
- 209 Thomassen E, Renshaw BR, Sims JE. Identification and characterization of SIGIRR, a molecule representing a novel subtype of the IL-1R superfamily. Cytokine. 1999; 11: 389–399.
- 210 Alexander WS, Hilton DJ. The role of suppressors of cytokine signaling (SOCS) proteins in regulation of the immune response. Annu Rev Immunol. 2004; 22: 503–529.
- 211 Kinjyo I, Hanada T, Inagaki-Ohara K, et al. SOCS1/JAB is a negative regulator of LPS-induced macrophage activation. Immunity. 2002; 17: 583–591.
- 212 Nakagawa R, Naka T, Tsutsui H, et al. SOCS-1 participates in negative regulation of LPS responses. Immunity. 2002; 17: 677–687.
- 213 Chinen T, Kobayashi T, Ogata H, et al. Suppressor of cytokine signaling-1 regulates inflammatory bowel disease in which both IFNgamma and IL-4 are involved. Gastroenterology. 2006; 130: 373–388.
- 214 Inagaki-Ohara K, Sasaki A, Matsuzaki G, et al. Suppressor of cytokine signalling 1 in lymphocytes regulates the development of intestinal inflammation in mice. Gut. 2006; 55: 212–219.
- 215 Hugot JP, Laurent-Puig P, Gower-Rousseau C, et al. Mapping of a susceptibility locus for Crohn's disease on chromosome 16. Nature. 1996; 379: 821–823.
- 216 Cho JH, Nicolae DL, Gold LH, 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.
- 217 Melmed G, Thomas LS, Lee N, et al. Human intestinal epithelial cells are broadly unresponsive to Toll-like receptor 2-dependent bacterial ligands: implications for host–microbial interactions in the gut. J Immunol. 2003; 170: 1406–1415.
- 218 Burns K, Clatworthy J, Martin L, et al. Tollip, a new component of the IL-1RI pathway, links IRAK to the IL-1 receptor. Nat Cell Biol. 2000; 2: 346–351.
- 219 Bulut Y, Faure E, Thomas L, et al. Cooperation of Toll-like receptor 2 and 6 for cellular activation by soluble tuberculosis factor and Borrelia burgdorferi outer surface protein A lipoprotein: role of Toll-interacting protein and IL-1 receptor signaling molecules in Toll-like receptor 2 signaling. J Immunol. 2001; 167: 987–994.
- 220 Zhang G, Ghosh S. Negative regulation of toll-like receptor-mediated signaling by Tollip. J Biol Chem. 2002; 277: 7059–7065.
- 221 Li T, Hu J, Li L. Characterization of Tollip protein upon Lipopolysaccharide challenge. Mol Immunol. 2004; 41: 85–92.
- 222 Williams CN, Kocher K, Lander ES, et al. 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.
- 223 Paavola-Sakki P, Ollikainen V, Helio T, et al. Genome-wide search in Finnish families with inflammatory bowel disease provides evidence for novel susceptibility loci. Eur J Hum Genet. 2003; 11: 112–120.
- 224 Watanabe T, Kitani A, Murray PJ, et al. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol. 2004; 5: 800–808.
- 225 Maeda S, Hsu LC, Liu H, et al. Nod2 mutation in Crohn's disease potentiates NF-kappaB activity and IL-1beta processing. Science. 2005; 307: 734–738.
- 226 Kobayashi KS, Chamaillard M, Ogura Y, et al. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005; 307: 731–734.
- 227 Netea MG, Ferwerda G, de Jong DJ, et al. Nucleotide-binding oligomerization domain-2 modulates specific TLR pathways for the induction of cytokine release. J Immunol. 2005; 174: 6518–6523.
- 228 McCartney-Francis N, Jin W, Wahl SM. Aberrant Toll receptor expression and endotoxin hypersensitivity in mice lacking a functional TGF-beta 1 signaling pathway. J Immunol. 2004; 172: 3814–3821.
- 229 Naik S, Kelly EJ, Meijer L, et al. Absence of Toll-like receptor 4 explains endotoxin hyporesponsiveness in human intestinal epithelium. J Pediatr Gastroenterol Nutr. 2001; 32: 449–453.
- 230 Gaya DR, Russell RK, Nimmo ER, et al. New genes in inflammatory bowel disease: lessons for complex diseases? Lancet. 2006; 367: 1271–1284.
- 231 Arbour NC, Lorenz E, Schutte BC, et al. TLR4 mutations are associated with endotoxin hyporesponsiveness in humans. Nat Genet. 2000; 25: 187–191.
- 232 Franchimont D, Vermeire S, El Housni H, et al. Deficient host–bacteria interactions in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn's disease and ulcerative colitis. Gut. 2004; 53: 987–992.
- 233 Brand S, Staudinger T, Schnitzler F, et al. The role of Toll-like receptor 4 Asp299Gly and Thr399Ile polymorphisms and CARD15/NOD2 mutations in the susceptibility and phenotype of Crohn's disease. Inflamm Bowel Dis. 2005; 11: 645–652.
- 234 Braat H, Stokkers P, Hommes T, et al. Consequence of functional Nod2 and Tlr4 mutations on gene transcription in Crohn's disease patients. J Mol Med. 2005; 83: 601–609.
- 235 Ouburg S, Mallant-Hent R, Crusius JB, et al. The toll-like receptor 4 (TLR4) Asp299Gly polymorphism is associated with colonic localisation of Crohn's disease without a major role for the Saccharomyces cerevisiae mannan-LBP-CD14–TLR4 pathway. Gut. 2005; 54: 439–440.
- 236 Török HP, Glas J, Tonenchi L, et al. Crohn's disease is associated with a toll-like receptor-9 polymorphism. Gastroenterology. 2004; 127: 365–366.
- 237 Lakatos PL, Lakatos L, Szalay F, et al. Toll-like receptor 4 and NOD2/CARD15 mutations in Hungarian patients with Crohn's disease: phenotype–genotype correlations. World J Gastroenterol. 2005; 11: 1489–1495.
- 238 Oostenbrug LE, Drenth JPH, De Jong DJ, et al. Association between toll-like receptor 4 and inflammatory bowel disease. Inflamm Bowel Dis. 2005; 11: 567–575.
- 239 Rotter JI, Taylor KD, Yang HY, et al. TLR5 polymorphisms are associated with OmpC and CBir1 expression and with severity of Crohn's disease in Ashkenazi Jews. Gastroenterology. 2006; 130: A53.
- 240 Gewirtz AT, Vijay-Kumar M, Brant SR, et al. Dominant-negative TLR5 polymorphism reduces adaptive immune response to flagellin and negatively associates with Crohn's disease. Am J Physiol Gastrointest Liver Physiol. 2006; 290: G1157–G1163.
- 241 Hawn TR, Verbon A, Lettinga KD, et al. A common dominant TLR5 stop codon polymorphism abolishes flagellin signaling and is associated with susceptibility to Legionnaires' disease. J Exp Med. 2003; 198: 1563–1572.
- 242 Lammers KM, Ouburg S, Morre SA, et al. Combined carriership of TLR9–1237C and CD14–260T alleles enhances the risk of developing chronic relapsing pouchitis. World J Gastroenterol. 2005; 11: 7323–7329.
- 243 Harton JA, Ting JP. Class II transactivator: mastering the art of major histocompatibility complex expression. Mol Cell Biol. 2000; 6185–6194.
- 244 Inohara N, Nunez G. The NOD: a signaling module that regulates apoptosis and host defense against pathogens. Oncogene. 2001; 6473–6481.
- 245 Steimle V, Otten LA, Zufferey M, et al. Complementation cloning of an MHC class II transactivator mutated in hereditary MHC class II deficiency (or bare lymphocyte syndrome). Cell. 1993; 75: 135–146.
- 246 Martinon F, Tschopp J. NLRs join TLRs as innate sensors of pathogens. Trends Immunol. 2005; 26: 447–454.
- 247 Strober W, Murray PJ, Kitani A, Watanabe T. Signalling pathways and molecular interactions of NOD1 and NOD2. Nat Rev Immunol. 2006; 6: 9–20.
- 248 Swanberg M, Lidman O, Padyukov L, et al. MHC2TA is associated with differential MHC molecule expression and susceptibility to rheumatoid arthritis, multiple sclerosis and myocardial infarction. Nat Genet. 2005; 37: 486–494.
- 249 Patarroyo JC, Stuve O, Piskurich JF, et al. Single nucleotide polymorphisms in MHC2TA, the gene encoding the MHC class II transactivator (CIITA). Genes Immun. 2002; 3: 34–37.
- 250 Hoffman HM, Mueller JL, Broide DH, et al. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle–Wells syndrome. Nat Genet. 2001; 29: 301–305.
- 251 Goldbach-Mansky R, Dailey NJ, Canna SW, et al. Neonatal-onset multisystem inflammatory disease responsive to interleukin-1beta inhibition. N Engl J Med. 2006; 355: 581–592.
- 252 Int FMF Consortium. Ancient missense mutations in a new member of the RoRet gene family are likely to cause familial Mediterranean fever. Cell. 1997; 90: 797–807.
- 253 French FMF Consortium. A candidate gene for familial Mediterranean fever. Nat Genet. 1997; 17: 25–31.
- 254 Villani AC, Lemire M, Louis E, et al. The familial Mediterranean fever (FMF) gene (MEFV) as a disease susceptibility gene in inflammatory bowel disease (IBD). Gastroenterology. 2006; 130: A53.
- 255 Karban A, Dagan E, Eliakim R, et al. Prevalence and significance of mutations in the familial Mediterranean fever gene in patients with Crohn's disease. Genes Immun. 2004; 6: 134–139.
- 256 Fidder H, Chowers Y, Ackerman Z, et al. The familial Mediterranean fever (MEVF) gene as a modifier of Crohn's disease. Am J Gastroenterol. 2005; 100: 338–343.
- 257 Inohara N, Nunez G. NODs: intracellular proteins involved in inflammation and apoptosis. Nat Rev Immunol. 2003; 3: 371–382.
- 258 Hysi P, Kabesch M, Moffatt MF, et al. NOD1 variation, immunoglobulin E and asthma. Hum Mol Genet. 2005; 14: 935–941.
- 259 Lala S, Ogura Y, Osborne C, et al. Crohn's disease and the NOD2 gene: a role for paneth cells. Gastroenterology. 2003; 125: 47–57.
- 260 Ogura Y, Lala S, Xin W, et al. Expression of NOD2 in Paneth cells: a possible link to Crohn's ileitis. Gut. 2003; 52: 1591–1597.
- 261 Abraham C, Cho JH. Functional consequences of NOD2 (CARD15) mutations. Inflamm Bowel Dis. 2006; 12: 641–650.
- 262 Berrebi D, Maudinas R, Hugot JP, et al. Card15 gene overexpression in mononuclear and epithelial cells of the inflamed Crohn's disease colon. Gut. 2003; 52: 840–846.
- 263 Girardin SE, Jehanno M, Mengin-Lecreulx D, et al. Identification of the critical residues involved in peptidoglycan detection by Nod1. J Biol Chem. 2005; 280: 38648–38656.
- 264 Girardin SE, Boneca IG, Carneiro LAM, et al. Nod1 detects a unique muropeptide from gram-negative bacterial peptidoglycan. Science. 2003; 300: 1584–1587.
- 265 Chamaillard M, Hashimoto M, Horie Y, et al. An essential role for NOD1 in host recognition of bacterial peptidoglycan containing diaminopimelic acid. Nat Immunol. 2003; 4: 702–707.
- 266 Uehara A, Fujimoto Y, Kawasaki A, et al. Meso-diaminopimelic acid and meso-lanthionine, amino acids specific to bacterial peptidoglycans, activate human epithelial cells through NOD1. J Immunol. 2006; 177: 1796–1804.
- 267 Inohara N, Ogura Y, Fontalba A, et al. Host recognition of bacterial muramyl dipeptide mediated through NOD2sulfimplications for Crohn's disease. J Biol Chem. 2003; 278: 5509–5512.
- 268 Girardin SE, Boneca IG, Viala J, et al. Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J Biol Chem. 2003; 278: 8869–8872.
- 269 Inohara N, Ogura Y, Chen FF, et al. Human Nod1 confers responsiveness to bacterial lipopolysaccharides. J Biol Chem. 2001; 276: 2551–2554.
- 270 Girardin SE, Tournebize R, Mavris M, et al. CARD4/Nod1 mediates NF-kappaB and JNK activation by invasive Shigella flexneri. EMBO Rep. 2001; 2: 736–742.
- 271 Franchi L, Amer A, Body-Malapel M, et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1beta in salmonella-infected macrophages. Nat Immunol. 2006; 7: 576–582.
- 272 Miao EA, Alpuche-Aranda CM, Dors M, et al. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1beta via Ipaf. Nat Immunol. 2006; 7: 569–575.
- 273 Ren T, Zamboni DS, Roy CR, et al. Flagellin-deficient Legionella mutants evade caspase-1- and Naip5-mediated macrophage immunity. PLoS Pathogens. 2006; 2: e18.
- 274 Molofsky AB, Byrne BG, Whitfield NN, et al. Cytosolic recognition of flagellin by mouse macrophages restricts Legionella pneumophila infection. J Exp Med. 2006; 1093–1104.
- 275 Krantz DD, Zidovetzki R, Kagan BL, et al. Amphipathic beta structure of a leucine-rich repeat peptide. J Biol Chem. 1991; 266: 16801–16807.
- 276 Viala J, Chaput C, Boneca IG, et al. Nod1 responds to peptidoglycan delivered by the Helicobacter pylori cag pathogenicity island. Nat Immunol. 2004; 5: 1166–1174.
- 277 Vavricka SR, Musch MW, Chang JE, et al. hPepT1 transports muramyl dipeptide, activating NF-kappaB and stimulating IL-8 secretion in human colonic Caco2/bbe cells. Gastroenterology. 2004; 127: 1401–1409.
- 278 Barnich N, Aguirre JE, Reinecker HC, et al. Membrane recruitment of NOD2 in intestinal epithelial cells is essential for nuclear factor-{kappa}B activation in muramyl dipeptide recognition. J Cell Biol. 2005; 170: 21–26.
- 279 Inohara N, Koseki T, Lin J, et al. An induced proximity model for NF-kappa B activation in the Nod1/RICK and RIP signaling pathways. J Biol Chem. 2000; 275: 27823–27831.
- 280 Tanabe T, Chamaillard M, Ogura Y, et al. Regulatory regions and critical residues of NOD2 involved in muramyl dipeptide recognition. EMBO J. 2004; 23: 1587–1597.
- 281 Inohara N, Chamaillard M, McDonald C, et al. NOD-LRR proteins: role in host–microbial interactions and inflammatory disease. Annu Rev Biochem. 2005; 74: 355–383.
- 282 Pauleau AL, Murray PJ. Role of nod2 in the response of macrophages to toll-like receptor agonists. Mol Cell Biol. 2003; 23: 7531–7539.
- 283 Barnich N, Hisamatsu T, Aguirre JE, et al. GRIM-19 interacts with nucleotide oligomerization domain 2 and serves as downstream effector of anti-bacterial function in intestinal epithelial cells. J Biol Chem. 2005; 280: 19021–19026.
- 284 McDonald C, Chen FF, Ollendorff V, et al. A role for Erbin in the regulation of Nod2-dependent NF-kappaB signaling. J Biol Chem. 2005; 280: 40301–40309.
- 285 Kufer TA, Kremmer E, Banks DJ, et al. Role for erbin in bacterial activation of Nod2. Infect Immun. 2006; 74: 3115–3124.
- 286 Chen CM, Gong Y, Zhang M, et al. Reciprocal cross-talk between Nod2 and TAK1 signaling pathways. J Biol Chem. 2004; 279: 25876–25882.
- 287 Sato S, Sanjo H, Tsujimura T, et al. TAK1 is indispensable for development of T cells and prevention of colitis by the generation of regulatory T cells. Int Immunol. 2006;e-pub ahead of print.
- 288 Martinon F, Tschopp J. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell. 2004; 117: 561–574.
- 289 Mariathasan S, Newton K, Monack DM, et al. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature. 2004; 430: 213–218.
- 290 Damiano JS, Oliveira V, Welsh K, et al. Heterotypic interactions among NACHT domains: implications for regulation of innate immune responses. Biochem J. 2004; 381: 213–219.
- 291 Stehlik C, Hayashi H, Pio F, et al. CARD6 is a modulator of NF-kappa B activation by Nod1- and Cardiak-mediated pathways. J Biol Chem. 2003; 278: 31941–31949.
- 292 Martinon F, Agostini L, Meylan E, et al. Identification of bacterial muramyl dipeptide as activator of the NALP3/cryopyrin inflammasome. Curr Biol. 2004; 14: 1929–1934.
- 293 McGovern DPB, Hysi P, Ahmad T, et al. Association between a complex insertion/deletion polymorphism in NOD1 (CARD4) and susceptibility to inflammatory bowel disease. Hum Mol Genet. 2005; 14: 1245–1250.
- 294 Zouali H, Lesage S, Merlin F, et al. CARD4/NOD1 is not involved in inflammatory bowel disease. Gut. 2003; 52: 71–74.
- 295 Ozen SC, Dagli U, Kilic MY, et al. Nod2/card15, nod1/card4, and icam-1 gene polymorphisms in Turkish patients with inflammatory bowel disease. J Gastroenterol. 2006; 41: 304–310.
- 296 Van Limbergen J, Lees CW, Nimmo ER, et al. Association of a complex insertion/deletion polymorphism of NOD1/CARD4 with susceptibility to inflammatory bowel disease in the Scottish population. Gastroenterology. 2006; 130: A64.
- 297 Tremelling M, Hancock L, Bredin F, et al. Complex insertion/deletion polymorphism in NOD1 (CARD4) is not associated with inflammatory bowel disease susceptibility in east anglia panel. Inflamm Bowel Dis. 2006; 12: 967–971.
- 298 Franke A, Ruether A, Wedemeyer N, et al. No association between the functional CARD4 insertion/deletion polymorphism and inflammatory bowel diseases in the German population. Gut. 2006; 55: 1679–1680.
- 299 van Heel DA, Hunt KA, King K, et al. Detection of muramyl dipeptide-sensing pathway defects in patients with Crohn's disease. Inflamm Bowel Dis. 2006; 12: 598–605.
- 300 Bairead E, Harmon DL, Curtis AM, et al. Association of NOD2 with Crohn's disease in a homogenous Irish population. Eur J Hum Genet. 2003; 11: 237–244.
- 301 Lesage S, Zouali H, Cezard JP, et al. CARD15/NOD2 mutational analysis and genotype–phenotype correlation in 612 patients with inflammatory bowel disease. Am J Hum Genet. 2002; 70: 845–857.
- 302 Ahmad T, Armuzzi A, Bunce M, et al. The molecular classification of the clinical manifestations of Crohn's disease. Gastroenterology. 2002; 122: 854–856.
- 303 Brant SR, Picco MF, Achkar JP, et al. 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.
- 304 Holler E, Rogler G, Brenmoehl J, et al. Prognostic significance of NOD2/CARD15 variants in HLA-identical sibling hematopoietic stem cell transplantation: effect on long-term outcome is confirmed in 2 independent cohorts and may be modulated by the type of gastrointestinal decontamination. Blood. 2006; 107: 4189–4193.
- 305 Miceli-Richard C, Lesage S, Rybojad M, et al. CARD15 mutations in Blau syndrome. Nat Genet. 2001; 29: 19–20.
- 306 Kanazawa N, Okafuji I, Kambe N, et al. Early-onset sarcoidosis and CARD15 mutations with constitutive nuclear factor-kappaB activation: common genetic etiology with Blau syndrome. Blood. 2005; 105: 1195–1197.
- 307 Economou M, Trikalinos TA, Loizou KT, et al. Differential effects of NOD2 variants on Crohn's disease risk and phenotype in diverse populations: a metaanalysis. Am J Gastroenterol. 2004; 99: 2393–2404.
- 308 Oostenbrug LE, Nolte IM, Oosterom E, et al. CARD15 in inflammatory bowel disease and Crohn's disease phenotypes: an association study and pooled analysis. Dig Liver Dis. 2006; in press.
- 309 Duerr RH, Taylor KD, Brant SR, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;e-pub 26/10/2006:1135245.
- 310 Törkvist L, Noble CL, Lördal M, et al. Contribution of CARD15 variants in determining susceptibility to Crohn's disease in Sweden. Scand J Gastroenterol. 2006; 41: 700–705.
- 311 Inoue N, Tamura K, Kinouchi Y, et al. Lack of common NOD2 variants in Japanese patients with Crohn's disease. Gastroenterology. 2002; 123: 86–91.
- 312 Yamazaki K, Takazoe M, Tanaka T, et al. Absence of mutation in the NOD2/CARD15 gene among 483 Japanese patients with Crohn's disease. J Hum Genet. 2002; 47: 469–472.
- 313 van Heel DA, Ghosh S, Butler M, et al. Muramyl dipeptide and toll-like receptor sensitivity in NOD2-associated Crohn's disease. Lancet. 2005; 365: 1794–1796.
- 314 Li J, Moran T, Swanson E, et al. Regulation of IL-8 and IL-1beta expression in Crohn's disease associated NOD2/CARD15 mutations. Hum Mol Genet. 2004; 13: 1715–1725.
- 315 van Heel DA, Ghosh S, Butler M, et al. Synergistic enhancement of Toll-like receptor responses by NOD1 activation. Eur J Immunol. 2005; 35: 2471–2476.
- 316 van Heel DA, Ghosh S, Hunt KA, et al. Synergy between TLR9 and NOD2 innate immune responses is lost in genetic Crohn's disease. Gut. 2005; 54: 1553–1557.
- 317 Tada H, Aiba S, Shibata K, et al. Synergistic effect of Nod1 and Nod2 agonists with toll-like receptor agonists on human dendritic cells to generate interleukin-12 and T helper type 1 cells. Infect Immun. 2005; 73: 7967–7976.
- 318 Fritz JH, Girardin SE, Fitting C, et al. Synergistic stimulation of human monocytes and dendritic cells by Toll-like receptor 4 and NOD1- and NOD2-activating agonists. Eur J Immunol. 2005; 35: 2459–2470.
- 319 Yang S, Tamai R, Akashi S, et al. Synergistic effect of muramyldipeptide with lipopolysaccharide or lipoteichoic acid to induce inflammatory cytokines in human monocytic cells in culture. Infect Immun. 2001; 69: 2045–2053.
- 320 Wolfert MA, Murray TF, Boons GJ, et al. The origin of the synergistic effect of muramyl dipeptide with endotoxin and peptidoglycan. J Biol Chem. 2002; 277: 39179–39186.
- 321 Uehara A, Yang S, Fujimoto Y, et al. Muramyldipeptide and diaminopimelic acid-containing desmuramylpeptides in combination with chemically synthesized Toll-like receptor agonists synergistically induced production of interleukin-8 in a NOD2- and NOD1-dependent manner, respectively, in human monocytic cells in culture. Cell Microbiol. 2005; 7: 53–61.
- 322 Netea MG, Ferwerda G, De Jong DJ, et al. The frameshift mutation in Nod2 results in unresponsiveness not only to Nod2- but also Nod1-activating peptidoglycan agonists. J Biol Chem. 2005; 280: 35859–35867.
- 323 van Heel DA, Hunt KA, Ghosh S, et al. Normal responses to specific NOD1-activating peptidoglycan agonists in the presence of the NOD2 frameshift and other mutations in Crohn's disease. Eur J Immunol. 2006; 36: 1629–1635.
- 324 MacPherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol. 2004; 4: 478–485.
- 325 Eckmann L. Defence molecules in intestinal innate immunity against bacterial infections. Curr Opin Gastroenterol. 2005; 21: 147–151.
- 326 Cash HL, Whitham CV, Behrendt CL, et al. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science. 2006; 313: 1126–1130.
- 327 Strober W. Immunology. Unraveling gut inflammation. Science. 2006; 313: 1052–1054.
- 328 Ganz T, Selsted ME, Szklarek D, et al. Defensins. Natural peptide antibiotics of human neutrophils. J Clin Invest. 1985; 76: 1427–1435.
- 329 Selsted ME, Harwig SS, Ganz T, et al. Primary structures of three human neutrophil defensins. J Clin Invest. 1985; 76: 1436–1439.
- 330 Ganz T. Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol. 2003; 3: 710–720.
- 331 Pazgier M, Hoover DM, Yang D, et al. Human beta-defensins. Cell Mol Life Sci. 2006; 63: 1294–1313.
- 332 Selsted ME, Ouellette AJ. Mammalian defensins in the antimicrobial immune response. Nat Immunol. 2005; 6: 551–557.
- 333 Lynn DJ, Lloyd AT, Fares MA, et al. Evidence of positively selected sites in mammalian alpha-defensins. Mol Biol Evol. 2004; 21: 819–827.
- 334 Semple CA, Gautier P, Taylor K, et al. The changing of the guard: molecular diversity and rapid evolution of beta-defensins. Mol Divers. 2006;epub 13/09/2006.
- 335 Ghosh D, Porter E, Shen B, et al. Paneth cell trypsin is the processing enzyme for human defensin-5. Nat Immunol. 2002; 3: 583–590.
- 336 Rumio C, Besusso D, Palazzo M, et al. Degranulation of Paneth cells via Toll-like receptor 9. Am J Pathol. 2004; 165: 373–381.
- 337 Wilson CL, Ouellette AJ, Satchell DP, et al. Regulation of intestinal alpha-defensin activation by the metalloproteinase matrilysin in innate host defense. Science. 1999; 286: 113–117.
- 338 Salzman NH, Ghosh D, Huttner KM, et al. Protection against enteric salmonellosis in transgenic mice expressing a human intestinal defensin. Nature. 2003; 422: 522–526.
- 339 Wehkamp J, Salzman NH, Porter E, et al. Reduced Paneth cell alpha-defensins in ileal Crohn's disease. Proc Natl Acad Sci USA. 2005; 102: 18129–18134.
- 340 Wehkamp J, Harder J, Weichenthal M, et al. NOD2 (CARD15) mutations in Crohn's disease are associated with diminished mucosal alpha-defensin expression. Gut. 2004; 53: 1658–1664.
- 341 O'Neil DA, Porter EM, Elewaut D, et al. Expression and regulation of the human beta-defensins hBD-1 and hBD-2 in intestinal epithelium. J Immunol. 1999; 163: 6718–6724.
- 342 Wehkamp J, Harder J, Weichenthal M, et al. Inducible and constitutive beta-defensins are differentially expressed in Crohn's disease and ulcerative colitis. Inflamm Bowel Dis. 2003; 9: 215–223.
- 343 Wehkamp J, Fellermann K, Herrlinger KR, et al. 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: 745–652.
- 344 Fahlgren A, Hammarstrom S, Danielsson A, et al. beta-Defensin-3 and -4 in intestinal epithelial cells display increased mRNA expression in ulcerative colitis. Clin Exp Immunol. 2004; 137: 379–385.
- 345
Birchler T,
Seibl R,
Buchner K, et al.
Human Toll-like receptor 2 mediates induction of the antimicrobial peptide human beta-defensin 2 in response to bacterial lipoprotein.
Eur J Immunol.
2001;
31:
3131–3137.
10.1002/1521-4141(200111)31:11<3131::AID-IMMU3131>3.0.CO;2-G CAS PubMed Web of Science® Google Scholar
- 346 Vora P, Youdim A, Thomas LS, et al. Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. J Immunol. 2004; 173: 5398–5405.
- 347 Salzman NH, Chou MM, de Jong H, et al. Enteric salmonella infection inhibits Paneth cell antimicrobial peptide expression. Infect Immun. 2003; 71: 1109–1115.
- 348 Zaalouk TK, Bajaj-Elliott M, George JT, et al. Differential regulation of beta-defensin gene expression during Cryptosporidium parvum infection. Infect Immun. 2004; 72: 2772–2779.
- 349 Voss E, Wehkamp J, Wehkamp K, et al. NOD2/CARD15 mediates induction of the antimicrobial peptide human beta-defensin-2. J Biol Chem. 2006; 281: 2005–2011.
- 350 Fellermann K, Stange DE, Schaeffeler E, et al. A chromosome 8 gene-cluster polymorphism with low human Beta-defensin 2 gene copy number predisposes to crohn disease of the colon. Am J Hum Genet. 2006; 79: 439–448.