Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mark Tremelling is active.

Publication


Featured researches published by Mark Tremelling.


Nature Genetics | 2008

Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease

Jeffrey C. Barrett; Sarah Hansoul; Dan L. Nicolae; Judy H. Cho; Richard H. Duerr; John D. Rioux; Steven R. Brant; Mark S. Silverberg; Kent D. Taylor; M. Michael Barmada; Alain Bitton; Themistocles Dassopoulos; Lisa W. Datta; Todd Green; Anne M. Griffiths; Emily O. Kistner; Miguel Regueiro; Jerome I. Rotter; L. Philip Schumm; A. Hillary Steinhart; Stephan R. Targan; Ramnik J. Xavier; Cécile Libioulle; Cynthia Sandor; Mark Lathrop; Jacques Belaiche; Olivier Dewit; Ivo Gut; Simon Heath; Debby Laukens

Several risk factors for Crohns disease have been identified in recent genome-wide association studies. To advance gene discovery further, we combined data from three studies on Crohns disease (a total of 3,230 cases and 4,829 controls) and carried out replication in 3,664 independent cases with a mixture of population-based and family-based controls. The results strongly confirm 11 previously reported loci and provide genome-wide significant evidence for 21 additional loci, including the regions containing STAT3, JAK2, ICOSLG, CDKAL1 and ITLN1. The expanded molecular understanding of the basis of this disease offers promise for informed therapeutic development.


Nature Genetics | 2007

Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn's disease susceptibility.

Miles Parkes; Jeffrey C. Barrett; Natalie J. Prescott; Mark Tremelling; Carl A. Anderson; Sheila Fisher; Roland G. Roberts; Elaine R. Nimmo; Fraser Cummings; Dianne Soars; Hazel E. Drummond; Charlie W. Lees; Saud A Khawaja; Richard Bagnall; D. A. Burke; Ce Todhunter; Tariq Ahmad; Clive M. Onnie; Wendy L. McArdle; David P. Strachan; Graeme Bethel; Claire Bryan; Cathryn M. Lewis; Panos Deloukas; Alastair Forbes; Jeremy Sanderson; Derek P. Jewell; Jack Satsangi; John C. Mansfield; Lon R. Cardon

A genome-wide association scan in individuals with Crohns disease by the Wellcome Trust Case Control Consortium detected strong association at four novel loci. We tested 37 SNPs from these and other loci for association in an independent case-control sample. We obtained replication for the autophagy-inducing IRGM gene on chromosome 5q33.1 (replication P = 6.6 × 10−4, combined P = 2.1 × 10−10) and for nine other loci, including NKX2-3, PTPN2 and gene deserts on chromosomes 1q and 5p13.


Nature Genetics | 2008

Genetic determinants of ulcerative colitis include the ECM1 locus and five loci implicated in Crohn's disease

Sheila Fisher; Mark Tremelling; Carl A. Anderson; Rhian Gwilliam; Suzannah Bumpstead; Natalie J. Prescott; Elaine R. Nimmo; Dunecan Massey; Carlo Berzuini; Christopher M. Johnson; Jeffrey C. Barrett; Fraser Cummings; Hazel E. Drummond; Charlie W. Lees; Clive M. Onnie; Catherine Hanson; Katarzyna Blaszczyk; Michael Inouye; Philip Ewels; Radhi Ravindrarajah; Andrew Keniry; Sarah Hunt; Martyn J. Carter; Nicholas J. Watkins; Willem H. Ouwehand; Cathryn M. Lewis; L R Cardon; Alan J. Lobo; Alastair Forbes; Jeremy Sanderson

We report results of a nonsynonymous SNP scan for ulcerative colitis and identify a previously unknown susceptibility locus at ECM1. We also show that several risk loci are common to ulcerative colitis and Crohns disease (IL23R, IL12B, HLA, NKX2-3 and MST1), whereas autophagy genes ATG16L1 and IRGM, along with NOD2 (also known as CARD15), are specific for Crohns disease. These data provide the first detailed illustration of the genetic relationship between these common inflammatory bowel diseases.


Gastroenterology | 2009

Investigation of Crohn's Disease Risk Loci in Ulcerative Colitis Further Defines Their Molecular Relationship

Carl A. Anderson; Dunecan Massey; Jeffrey C. Barrett; Natalie J. Prescott; Mark Tremelling; Sheila Fisher; Rhian Gwilliam; Jemima Jacob; Elaine R. Nimmo; Hazel E. Drummond; Charlie W. Lees; Clive M. Onnie; Catherine Hanson; Katarzyna Blaszczyk; Radhi Ravindrarajah; Sarah Hunt; Dhiraj Varma; Naomi Hammond; Gregory Lewis; Heather Attlesey; Nicholas A. Watkins; Willem H. Ouwehand; David P. Strachan; Wendy L. McArdle; Cathryn M. Lewis; Alan J. Lobo; Jeremy Sanderson; Derek P. Jewell; Panos Deloukas; John C. Mansfield

BACKGROUND & AIMS Identifying shared and disease-specific susceptibility loci for Crohns disease (CD) and ulcerative colitis (UC) would help define the biologic relationship between the inflammatory bowel diseases. More than 30 CD susceptibility loci have been identified. These represent important candidate susceptibility loci for UC. Loci discovered by the index genome scans in CD have previously been tested for association with UC, but those identified in the recent meta-analysis await such investigation. Furthermore, the recently identified UC locus at ECM1 requires formal testing for association with CD. METHODS We analyzed 45 single nucleotide polymorphisms, tagging 29 of the loci recently associated with CD in 2527 UC cases and 4070 population controls. We also genotyped the UC-associated ECM1 variant rs11205387 in 1560 CD patients and 3028 controls. RESULTS Nine regions showed association with UC at a threshold corrected for the 29 loci tested (P < .0017). The strongest association (P = 4.13 x 10(-8); odds ratio = 1.27) was identified with a 170-kilobase region on chromosome 1q32 that contains 3 genes. We also found association with JAK2 and replicated a recently reported association with STAT3, further implicating the role of this signaling pathway in inflammatory bowel disease. Additional novel UC susceptibility genes were LYRM4 and CDKAL1. Twenty of the loci were not associated with UC, and several appear to be specific to CD. ECM1 variation was not associated with CD. CONCLUSIONS Collectively, these data help define the genetic relationship between CD and UC and characterize common, as well as disease-specific mechanisms of pathogenesis.


Gut | 2006

Evidence for association of OCTN genes and IBD5 with ulcerative colitis

Sarah Waller; Mark Tremelling; Fran Bredin; Lisa Godfrey; Joanna M. M. Howson; Miles Parkes

Background and aims: Genetic association between Crohn’s disease (CD) and OCTN1 (SLC22A4) C1672T/OCTN2 (SLC22A5) G−207C variants in IBD5 has recently been reported. These genes encode solute carriers and the association was suggested to be distinct from the background IBD5 risk haplotype. There have been conflicting reports of the association between markers in the IBD5 region and ulcerative colitis (UC) and interaction (epistasis) between this locus and CARD15. Our aim was to ascertain the contribution of OCTN variants to UC and CD in a large independent UK dataset, to seek genetic evidence that the OCTN association is distinct from the IBD5 risk haplotype and to identify interactions between the IBD5 and CARD15 loci. Methods: A total of 1104 unrelated Caucasian subjects with inflammatory bowel disease (IBD) (496 CD, 512 UC, 96 indeterminate) and 750 ethnically matched controls were genotyped for three single nucleotide polymorphisms (SNPs) in the CD associated genes (OCTN1+1672, OCTN2−207, and IGR2230), and two flanking IBD5 tagging SNPs, IGR2096 and IGR3096. Data were analysed by logistic regression methods within STATA. Results: OCTN variants were as strongly associated with UC and IBD overall as they were with CD (p = 0.0001; OR 1.3 (95% confidence interval 1.1–1.5)). OCTN variants were in tight linkage disequilibrium with the extended IBD5 risk haplotype D′ 0.79 and 0.88, and r2 = 0.62 and 0.72 for IGR2096 and 3096, respectively. There was no deviation from a multiplicative model of interaction between CARD15 and IBD5 on the penetrance scale. Conclusions: The OCTN variants were associated with susceptibility to IBD overall. The effect was equally strong in UC and CD. Although OCTN variants may account for the increased risk of IBD associated with IBD5, a role for other candidate genes within this extended haplotype was not excluded. There was no statistical evidence of interaction between CARD15 and either OCTN or IBD5 variants in susceptibility to IBD.


Nature Genetics | 2017

Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease

Katrina M de Lange; Loukas Moutsianas; James C. Lee; Christopher A. Lamb; Yang Luo; Nicholas A. Kennedy; Luke Jostins; Daniel L. Rice; Javier Gutierrez-Achury; Sun-Gou Ji; Graham A. Heap; Elaine R. Nimmo; Cathryn Edwards; Paul Henderson; Craig Mowat; Jeremy Sanderson; Jack Satsangi; Alison Simmons; David C. Wilson; Mark Tremelling; Ailsa Hart; Christopher G. Mathew; William G. Newman; Miles Parkes; Charlie W. Lees; Holm H. Uhlig; Christopher J. Hawkey; Natalie J. Prescott; Tariq Ahmad; John C. Mansfield

Genetic association studies have identified 215 risk loci for inflammatory bowel disease, thereby uncovering fundamental aspects of its molecular biology. We performed a genome-wide association study of 25,305 individuals and conducted a meta-analysis with published summary statistics, yielding a total sample size of 59,957 subjects. We identified 25 new susceptibility loci, 3 of which contain integrin genes that encode proteins in pathways that have been identified as important therapeutic targets in inflammatory bowel disease. The associated variants are correlated with expression changes in response to immune stimulus at two of these genes (ITGA4 and ITGB8) and at previously implicated loci (ITGAL and ICAM1). In all four cases, the expression-increasing allele also increases disease risk. We also identified likely causal missense variants in a gene implicated in primary immune deficiency, PLCG2, and a negative regulator of inflammation, SLAMF8. Our results demonstrate that new associations at common variants continue to identify genes relevant to therapeutic target identification and prioritization.


Nature Genetics | 2014

HLA-DQA1-HLA-DRB1 variants confer susceptibility to pancreatitis induced by thiopurine immunosuppressants

Graham A. Heap; Michael N. Weedon; C Bewshea; Abhey Singh; Mian Chen; Jack B. Satchwel; Julian P. Vivian; Kenji So; P Dubois; Jane M. Andrews; Vito Annese; Peter A. Bampton; Martin Barnardo; Sally Bell; Andy Cole; Susan J. Connor; Tom J. Creed; Fraser Cummings; Mauro D'Amato; Tawfique K. Daneshmend; Richard N. Fedorak; Timothy H. Florin; Daniel R. Gaya; Emma Greig; Jonas Halfvarson; Alisa Hart; Peter M. Irving; Gareth Jones; Amir Karban; Ian C. Lawrance

Pancreatitis occurs in approximately 4% of patients treated with the thiopurines azathioprine or mercaptopurine. Its development is unpredictable and almost always leads to drug withdrawal. We identified patients with inflammatory bowel disease (IBD) who had developed pancreatitis within 3 months of starting these drugs from 168 sites around the world. After detailed case adjudication, we performed a genome-wide association study on 172 cases and 2,035 controls with IBD. We identified strong evidence of association within the class II HLA region, with the most significant association identified at rs2647087 (odds ratio 2.59, 95% confidence interval 2.07–3.26, P = 2 × 10−16). We replicated these findings in an independent set of 78 cases and 472 controls with IBD matched for drug exposure. Fine mapping of the HLA region identified association with the HLA-DQA1*02:01–HLA-DRB1*07:01 haplotype. Patients heterozygous at rs2647087 have a 9% risk of developing pancreatitis after administration of a thiopurine, whereas homozygotes have a 17% risk.


Inflammatory Bowel Diseases | 2008

Contribution of TNFSF15 gene variants to Crohn's disease susceptibility confirmed in UK population

Mark Tremelling; Carlo Berzuini; Dunecan Massey; Francesca Bredin; Catherine Waddams Price; Claire Dawson; Sheila A. Bingham; Miles Parkes

Background: Identification of Crohns disease (CD)‐associated genetic variants is key to understanding pathogenic pathways underlying disease susceptibility. Recent reports of an association between TNFSF15 variants and CD have been modestly replicated in European populations, suggesting heterogeneity at this locus with stronger CD association in Japanese than European populations. Methods: We investigated the association between variants in TNFSF15 and CD in 756 CD patients and 636 controls. Disease subphenotype associations were also investigated. Results: TNFSF15 single nucleotide polymorphism (SNP) variants were associated with CD in our panel with peak odds ratio (OR) 1.2 (95% confidence interval [CI] 1.01–1.41) P = 0.033. The presence of a risk haplotype was replicated for the first time in a European population (frequency 67% in cases and 61% in controls) OR = 1.44 (95% CI 1.23–1.68) P = 0.00012. This result mirrors the UK panel in the index study (Yamazaki et al [2005] Hum Mol Genet 14:3499–3506) but is less significant than that reported in Japanese populations. There was no evidence of association with any individual CD subphenotype. Conclusions: Variants in TNFSF15 contribute to overall CD susceptibility in European populations, although to a lesser extent than that seen in the Japanese. Further studies to define the precise disease‐causing variants as well as targeted functional studies are now required in human CD as TNFSF15 is a potential target for biological therapies.


PLOS Medicine | 2008

Analysis of germline GLI1 variation implicates hedgehog signalling in the regulation of intestinal inflammatory pathways.

Charlie W. Lees; William J. Zacharias; Mark Tremelling; Colin L. Noble; Elaine R. Nimmo; Albert Tenesa; Jennine Cornelius; Leif Törkvist; John Y. Kao; Susan M. Farrington; Hazel E. Drummond; Gwo Tzer Ho; D. R. Ian Arnott; Henry D. Appelman; Lauri Diehl; Harry Campbell; Malcolm G. Dunlop; Miles Parkes; E. M. Sarah Howie; Deborah L. Gumucio; Jack Satsangi

Background Ulcerative colitis (UC) and Crohns disease (CD) are polygenic chronic inflammatory bowel diseases (IBD) of high prevalence that are associated with considerable morbidity. The hedgehog (HH) signalling pathway, which includes the transcription factor glioma-associated oncogene homolog 1 (GLI1), plays vital roles in gastrointestinal tract development, homeostasis, and malignancy. We identified a germline variation in GLI1 (within the IBD2 linkage region, 12q13) in patients with IBD. Since this IBD-associated variant encodes a GLI1 protein with reduced function and our expression studies demonstrated down-regulation of the HH response in IBD, we tested whether mice with reduced Gli1 activity demonstrate increased susceptibility to chemically induced colitis. Methods and Findings Using a gene-wide haplotype-tagging approach, germline GLI1 variation was examined in three independent populations of IBD patients and healthy controls from Northern Europe (Scotland, England, and Sweden) totalling over 5,000 individuals. On log-likelihood analysis, GLI1 was associated with IBD, predominantly UC, in Scotland and England (p < 0.0001). A nonsynonymous SNP (rs2228226C→G), in exon 12 of GLI1 (Q1100E) was strongly implicated, with pooled odds ratio of 1.194 (confidence interval = 1.09–1.31, p = 0.0002). GLI1 variants were tested in vitro for transcriptional activity in luciferase assays. Q1100E falls within a conserved motif near the C terminus of GLI1; the variant GLI protein exhibited reduced transactivation function in vitro. In complementary expression studies, we noted the colonic HH response, including GLI1, patched (PTCH), and hedgehog-interacting protein (HHIP), to be down-regulated in patients with UC. Finally, Gli1+/lacZ mice were tested for susceptibility to dextran sodium sulphate (DSS)-induced colitis. Clinical response, histology, and expression of inflammatory cytokines and chemokines were recorded. Gli1+/lacZ mice rapidly developed severe intestinal inflammation, with considerable morbidity and mortality compared with wild type. Local myeloid cells were shown to be direct targets of HH signals and cytokine expression studies revealed robust up-regulation of IL-12, IL-17, and IL-23 in this model. Conclusions HH signalling through GLI1 is required for appropriate modulation of the intestinal response to acute inflammatory challenge. Reduced GLI1 function predisposes to a heightened myeloid response to inflammatory stimuli, potentially leading to IBD.


Inflammatory Bowel Diseases | 2006

Genetic variants in TNF‐α but not DLG5 are associated with inflammatory bowel disease in a large United Kingdom cohort

Mark Tremelling; Sarah Waller; Francesca Bredin; Simon M. Greenfield; Miles Parkes

Background: Genetic variants in DLG5, which encodes a scaffolding protein on chromosome 10q23, and tumor necrosis factor (TNF)‐&agr;, encoding a proinflammatory cytokine on chromosome 6p, have recently been reported to be associated with inflammatory bowel disease (IBD). We studied these variants to seek evidence of association with IBD in a large independent dataset. Methods: We genotyped 1104 unrelated white IBD subjects‐496 with Crohns disease, 512 with ulcerative colitis, and 96 with indeterminate colitis from the Cambridge/Eastern (UK) panel‐and 760 healthy control subjects for DLG5_113G/A, DLG5_4136C/A, TNF‐857C/T, and TNF‐1031T/C polymorphisms. Known Crohns disease‐predisposing variants in CARD15/NOD2 were also genotyped to permit analysis for reported epistatic interactions. Results: TNF‐857 was shown to be associated with IBD overall (P = 0.0079). A formal interaction test showed that TNF‐857 is associated equally with ulcerative colitis and Crohns disease. Neither of the DLG5 alleles, however, was associated with IBD (P = 0.32 and 0.35). Subgroup analysis also failed to show evidence of association between either DLG5 allele or genotype frequencies and ulcerative colitis or Crohns disease. Stratification of TNF‐&agr; and DLG5 cases by CARD15 genotype made no significant difference in the strength of associations. Conclusions: We have confirmed an association between the TNF‐857 promoter polymorphism and IBD in a large independent UK dataset but were unable to replicate an association at the previously reported loci within DLG5. This may reflect heterogeneity between the populations, a smaller effect size than originally predicted, or possibly a false‐positive result in the original study. Further fine mapping studies of the TNF promoter region and studies assessing functional consequences of TNF promoter polymorphisms are now required in IBD.

Collaboration


Dive into the Mark Tremelling's collaboration.

Top Co-Authors

Avatar

Miles Parkes

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy Sanderson

Guy's and St Thomas' NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony M. Marinaki

Guy's and St Thomas' NHS Foundation Trust

View shared research outputs
Researchain Logo
Decentralizing Knowledge