6451
|
Increasing incidence of paediatric inflammatory bowel disease in northern Stockholm County, 2002-2007. J Pediatr Gastroenterol Nutr 2013; 57:29-34. [PMID: 23459320 DOI: 10.1097/mpg.0b013e31828f21b4] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES A sharp increase in paediatric (younger than 16 years) inflammatory bowel disease (IBD) incidence was observed in northern Stockholm County, Sweden, in 1990-2001. The increasing incidence was primarily explained by a rising incidence of Crohn disease (CD). Here, we present an update on the trends in incidence of paediatric IBD, 2002-2007. METHOD Medical records of all children diagnosed as having suspected IBD in northern Stockholm County, 2002-2007, were scrutinised using defined diagnostic criteria. Disease extension, localisation, and behaviour at diagnosis were classified within the framework of the Paris classification. RESULT A total of 133 children were diagnosed as having IBD 2002-2007 corresponding to a sex- and age-standardised incidence (per 10 person-years) for paediatric IBD of 12.8 (95% CI 10.8-15.2). The standardised incidence was 9.2 (95% CI 7.5-11.2) for CD and 2.8 (95% CI 1.9-4.0) for ulcerative colitis (UC). A significant increasing incidence of UC (P < 0.05) was observed during the study period. No temporal trend was observed for the incidence of CD. CONCLUSIONS The incidence rate of paediatric IBD in northern Stockholm was significantly higher in 2002-2007 than that observed in our earlier study covering 1990-2001. The former sharp increase in incidence of paediatric CD seems, however, to have levelled out, although at a higher rate than reported from most other regions in the world. Although CD was still predominant, the observed increase in incidence of UC during the study period is notable.
Collapse
|
6452
|
Nys K, Agostinis P, Vermeire S. Autophagy: a new target or an old strategy for the treatment of Crohn's disease? Nat Rev Gastroenterol Hepatol 2013; 10:395-401. [PMID: 23591407 DOI: 10.1038/nrgastro.2013.66] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past 5 years much progress has been made in understanding the molecular basis of Crohn's disease, a multifactorial chronic inflammatory disease of the gastrointestinal tract. Data suggest that hampered autophagy--the major lysosomal pathway for recycling of cytoplasmic material--might contribute to an increased susceptibility to Crohn's disease. Consequently, intense investigations have started to evaluate the potential value of autophagy as a therapeutic target and as a highly needed diagnostic tool. Interestingly, as well as the promising introduction of direct autophagic modulators, several drugs already used in the treatment of Crohn's disease might exert at least part of their effect through the regulation of autophagy. However, whether this phenomenon contributes to or rather counteracts their therapeutic use, remains to be determined and might prove to be highly compound-specific. Here we review the complex and emerging role of autophagy modulation in the battle against Crohn's disease. Moreover, we discuss the potential benefits and deleterious effects of autophagic regulation by both new and clinically used drugs.
Collapse
Affiliation(s)
- Kris Nys
- Translational Research in Gastrointestinal Disorders, Department of Clinical and Experimental Medicine, Faculty of Medicine, Catholic University of Leuven, Herestraat 49, Box 701, 3000 Leuven, Belgium
| | | | | |
Collapse
|
6453
|
Abstract
PURPOSE OF REVIEW Stem cell therapy has emerged as a promising therapeutic strategy for inflammatory bowel diseases (IBDs). Currently, stem cell therapy is not part of the standard of care and is usually only performed as a part of clinical trials. In this review, clinical results, proposed underlying mechanisms, and future research directions will be discussed. RECENT FINDINGS Administration of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) has been evaluated for IBD treatment over the past years. MSC therapy is being explored as a treatment option for fistulizing Crohn's disease and for luminal Crohn's disease. It is shown to be well tolerated, but results on efficacy are inconsistent. HSC transplantation seems to be very effective, but serious adverse events are common. Therefore, future research should focus on improving efficacy of MSC therapy, and on improvement of safety of HSC therapy. SUMMARY Both MSC and HSC therapy offer clinical potential, but currently are not routinely used for IBD treatment. MSC therapy seems well tolerated but results on efficacy are conflicting. HSC transplantation is shown to be effective but safety concerns remain. Nonetheless, for severe refractory IBD cases, stem cell therapy could well become the next-generation treatment option.
Collapse
|
6454
|
Abstract
Phosphoinositides (PIs) make up only a small fraction of cellular phospholipids, yet they control almost all aspects of a cell's life and death. These lipids gained tremendous research interest as plasma membrane signaling molecules when discovered in the 1970s and 1980s. Research in the last 15 years has added a wide range of biological processes regulated by PIs, turning these lipids into one of the most universal signaling entities in eukaryotic cells. PIs control organelle biology by regulating vesicular trafficking, but they also modulate lipid distribution and metabolism via their close relationship with lipid transfer proteins. PIs regulate ion channels, pumps, and transporters and control both endocytic and exocytic processes. The nuclear phosphoinositides have grown from being an epiphenomenon to a research area of its own. As expected from such pleiotropic regulators, derangements of phosphoinositide metabolism are responsible for a number of human diseases ranging from rare genetic disorders to the most common ones such as cancer, obesity, and diabetes. Moreover, it is increasingly evident that a number of infectious agents hijack the PI regulatory systems of host cells for their intracellular movements, replication, and assembly. As a result, PI converting enzymes began to be noticed by pharmaceutical companies as potential therapeutic targets. This review is an attempt to give an overview of this enormous research field focusing on major developments in diverse areas of basic science linked to cellular physiology and disease.
Collapse
Affiliation(s)
- Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
6455
|
Abstract
PURPOSE OF REVIEW The occurrence of inflammatory bowel disease (IBD) is increasing worldwide, yet the reasons remain unknown. New therapeutic approaches have been introduced in medical IBD therapy, but their impact on the natural history of IBD remains uncertain. This review will summarize the recent findings in the epidemiology of IBD. RECENT FINDINGS The incidence of IBD in western Europe is twice as high as in eastern Europe, whereas the highest IBD incidence in the world is found in the Faroe Islands. Early intervention with immunosuppressant and biological agents seems to have reduced the colectomy rates for ulcerative colitis, whereas the impact on Crohn's disease has yet to be determined. Mortality in Crohn's disease has not changed despite improvements in medical and surgical management. Specialized care in IBD centres, treatments to target and obtaining mucosal healing, early intervention at relapse and avoiding Clostridium difficile super infection might reduce the mortality rate in the future. The risk of colorectal cancer in Crohn's disease seems to be equivalent to the risk in ulcerative colitis. Patients with small bowel Crohn's disease are at increased risk of small bowel adenocarcinoma. SUMMARY The natural disease course of IBD seems to change along with the new 'treat to target' goal of achieving intestinal mucosal healing. Future population-based studies of unselected IBD cohorts should be considered the gold standard for studies investigating these issues.
Collapse
|
6456
|
Abstract
PURPOSE OF REVIEW To use insights from evolutionary biology to assess the current evidence for the causes, treatment, and prevention of inflammatory bowel disease (IBD). RECENT FINDINGS When analyzed in the context of evolutionary adaptation, recent assessments of genetic, microbial, and environmental associations with IBD implicate infectious causation. SUMMARY An evolutionary perspective provides insight into the causes of IBD, interpretation of its manifestations, and assessment of interventions. The evidence implicating infectious causation suggests that future studies of IBD would benefit from increased focus on infectious causes and interventions that prevent or inhibit them.
Collapse
|
6457
|
Harrison OJ, Powrie FM. Regulatory T cells and immune tolerance in the intestine. Cold Spring Harb Perspect Biol 2013; 5:5/7/a018341. [PMID: 23818502 DOI: 10.1101/cshperspect.a018341] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A fundamental role of the mammalian immune system is to eradicate pathogens while minimizing immunopathology. Instigating and maintaining immunological tolerance within the intestine represents a unique challenge to the mucosal immune system. Regulatory T cells are critical for continued immune tolerance in the intestine through active control of innate and adaptive immune responses. Dynamic adaptation of regulatory T-cell populations to the intestinal tissue microenvironment is key in this process. Here, we discuss specialization of regulatory T-cell responses in the intestine, and how a breakdown in these processes can lead to chronic intestinal inflammation.
Collapse
Affiliation(s)
- Oliver J Harrison
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | | |
Collapse
|
6458
|
Abstract
PURPOSE OF REVIEW Epigenetic studies are transforming our understanding of a variety of complex pathological conditions including cancer, autoimmune, and inflammatory diseases. A selection of the major recent advances in this area will be reviewed, focusing on the important emerging themes that are relevant to these diseases including inflammatory bowel disease (IBD). RECENT FINDINGS The main current themes that will be addressed on the role of epigenetics in disease pathogenesis include current understanding of the nature and function of histone modifications and DNA methylation; the connection between epigenetics and metabolic pathways; new studies on the mechanism of heritability of epigenetic changes; the role of stochastic noise and the expanding research on chromatin readers and their potential as selective therapeutic targets. The recent contribution of epigenetic modifications in defining the molecular basis of IBD and how such changes may act as fine-tuners of gene expression in these intestinal disorders are also discussed. SUMMARY Published evidence over the last 12-18 months indicates that targeting epigenetic factors can be efficacious in cancer and inflammatory disease. All the indications are that future research will continue to reveal new epigenetic targets and mechanisms that will advance the prospects for selective epigenetic therapy for IBD and other complex diseases.
Collapse
|
6459
|
Jun JC, Cominelli F, Abbott DW. RIP2 activity in inflammatory disease and implications for novel therapeutics. J Leukoc Biol 2013; 94:927-32. [PMID: 23794710 DOI: 10.1189/jlb.0213109] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The role of NOD2 and RIP2 in inflammatory disease has been paradoxical. Whereas loss-of-function NOD2 polymorphisms cause CD, a granulomatous disease of the gastrointestinal tract, gain-of-function mutations cause EOS-a granulomatous disease primarily affecting the skin, joints, and eyes. Thus, gain-of-function mutations and loss-of-function polymorphisms cause granulomatous inflammatory disease, only in different anatomic locations. The situation is complicated further by the fact that WT NOD2 and WT RIP2 activity has been implicated in diseases such as asthma, inflammatory arthritis and MS. This article reviews the role that the NOD2:RIP2 complex plays in inflammatory disease, with an emphasis on the inhibition of this signaling pathway as a novel pharmaceutical target in inflammatory disease.
Collapse
Affiliation(s)
- Janice C Jun
- 1.Case Western Reserve University School of Medicine, Wolstein Research Bldg., 2103 Cornell Rd., Room 6532, Cleveland, OH 44122, USA.
| | | | | |
Collapse
|
6460
|
Rietveld CA, Medland SE, Derringer J, Yang J, Esko T, Martin NW, Westra HJ, Shakhbazov K, Abdellaoui A, Agrawal A, Albrecht E, Alizadeh BZ, Amin N, Barnard J, Baumeister SE, Benke KS, Bielak LF, Boatman JA, Boyle PA, Davies G, de Leeuw C, Eklund N, Evans DS, Ferhmann R, Fischer K, Gieger C, Gjessing HK, Hägg S, Harris JR, Hayward C, Holzapfel C, Ibrahim-Verbaas CA, Ingelsson E, Jacobsson B, Joshi PK, Jugessur A, Kaakinen M, Kanoni S, Karjalainen J, Kolcic I, Kristiansson K, Kutalik Z, Lahti J, Lee SH, Lin P, Lind PA, Liu Y, Lohman K, Loitfelder M, McMahon G, Vidal PM, Meirelles O, Milani L, Myhre R, Nuotio ML, Oldmeadow CJ, Petrovic KE, Peyrot WJ, Polašek O, Quaye L, Reinmaa E, Rice JP, Rizzi TS, Schmidt H, Schmidt R, Smith AV, Smith JA, Tanaka T, Terracciano A, van der Loos MJ, Vitart V, Völzke H, Wellmann J, Yu L, Zhao W, Allik J, Attia JR, Bandinelli S, Bastardot F, Beauchamp J, Bennett DA, Berger K, Bierut LJ, Boomsma DI, Bültmann U, Campbell H, Chabris CF, Cherkas L, Chung MK, Cucca F, de Andrade M, De Jager PL, De Neve JE, Deary IJ, Dedoussis GV, Deloukas P, Dimitriou M, Eiriksdottir G, Elderson MF, Eriksson JG, et alRietveld CA, Medland SE, Derringer J, Yang J, Esko T, Martin NW, Westra HJ, Shakhbazov K, Abdellaoui A, Agrawal A, Albrecht E, Alizadeh BZ, Amin N, Barnard J, Baumeister SE, Benke KS, Bielak LF, Boatman JA, Boyle PA, Davies G, de Leeuw C, Eklund N, Evans DS, Ferhmann R, Fischer K, Gieger C, Gjessing HK, Hägg S, Harris JR, Hayward C, Holzapfel C, Ibrahim-Verbaas CA, Ingelsson E, Jacobsson B, Joshi PK, Jugessur A, Kaakinen M, Kanoni S, Karjalainen J, Kolcic I, Kristiansson K, Kutalik Z, Lahti J, Lee SH, Lin P, Lind PA, Liu Y, Lohman K, Loitfelder M, McMahon G, Vidal PM, Meirelles O, Milani L, Myhre R, Nuotio ML, Oldmeadow CJ, Petrovic KE, Peyrot WJ, Polašek O, Quaye L, Reinmaa E, Rice JP, Rizzi TS, Schmidt H, Schmidt R, Smith AV, Smith JA, Tanaka T, Terracciano A, van der Loos MJ, Vitart V, Völzke H, Wellmann J, Yu L, Zhao W, Allik J, Attia JR, Bandinelli S, Bastardot F, Beauchamp J, Bennett DA, Berger K, Bierut LJ, Boomsma DI, Bültmann U, Campbell H, Chabris CF, Cherkas L, Chung MK, Cucca F, de Andrade M, De Jager PL, De Neve JE, Deary IJ, Dedoussis GV, Deloukas P, Dimitriou M, Eiriksdottir G, Elderson MF, Eriksson JG, Evans DM, Faul JD, Ferrucci L, Garcia ME, Grönberg H, Gudnason V, Hall P, Harris JM, Harris TB, Hastie ND, Heath AC, Hernandez DG, Hoffmann W, Hofman A, Holle R, Holliday EG, Hottenga JJ, Iacono WG, Illig T, Järvelin MR, Kähönen M, Kaprio J, Kirkpatrick RM, Kowgier M, Latvala A, Launer LJ, Lawlor DA, Lehtimäki T, Li J, Lichtenstein P, Lichtner P, Liewald DC, Madden PA, Magnusson PKE, Mäkinen TE, Masala M, McGue M, Metspalu A, Mielck A, Miller MB, Montgomery GW, Mukherjee S, Nyholt DR, Oostra BA, Palmer LJ, Palotie A, Penninx B, Perola M, Peyser PA, Preisig M, Räikkönen K, Raitakari OT, Realo A, Ring SM, Ripatti S, Rivadeneira F, Rudan I, Rustichini A, Salomaa V, Sarin AP, Schlessinger D, Scott RJ, Snieder H, Pourcain BS, Starr JM, Sul JH, Surakka I, Svento R, Teumer A, Tiemeier H, Rooij FJA, Van Wagoner DR, Vartiainen E, Viikari J, Vollenweider P, Vonk JM, Waeber G, Weir DR, Wichmann HE, Widen E, Willemsen G, Wilson JF, Wright AF, Conley D, Davey-Smith G, Franke L, Groenen PJF, Hofman A, Johannesson M, Kardia SL, Krueger RF, Laibson D, Martin NG, Meyer MN, Posthuma D, Thurik AR, Timpson NJ, Uitterlinden AG, van Duijn CM, Visscher PM, Benjamin DJ, Cesarini D, Koellinger PD. GWAS of 126,559 individuals identifies genetic variants associated with educational attainment. Science 2013; 340:1467-71. [PMID: 23722424 PMCID: PMC3751588 DOI: 10.1126/science.1235488] [Show More Authors] [Citation(s) in RCA: 503] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A genome-wide association study (GWAS) of educational attainment was conducted in a discovery sample of 101,069 individuals and a replication sample of 25,490. Three independent single-nucleotide polymorphisms (SNPs) are genome-wide significant (rs9320913, rs11584700, rs4851266), and all three replicate. Estimated effects sizes are small (coefficient of determination R(2) ≈ 0.02%), approximately 1 month of schooling per allele. A linear polygenic score from all measured SNPs accounts for ≈2% of the variance in both educational attainment and cognitive function. Genes in the region of the loci have previously been associated with health, cognitive, and central nervous system phenotypes, and bioinformatics analyses suggest the involvement of the anterior caudate nucleus. These findings provide promising candidate SNPs for follow-up work, and our effect size estimates can anchor power analyses in social-science genetics.
Collapse
Affiliation(s)
- Cornelius A. Rietveld
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, 3000 DR Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Sarah E. Medland
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Jaime Derringer
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80309–0447, USA
| | - Jian Yang
- University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland 4102, Australia
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Nicolas W. Martin
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
- School of Psychology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Harm-Jan Westra
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Konstantin Shakhbazov
- University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland 4102, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Abdel Abdellaoui
- Department of Biological Psychology, VU University Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Behrooz Z. Alizadeh
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, the Netherlands
| | - John Barnard
- Heart and Vascular and Lerner Research Institutes, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | - Kelly S. Benke
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109–2029, USA
| | - Jeffrey A. Boatman
- Division of Biostatistics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Patricia A. Boyle
- Rush University Medical Center, Rush Alzheimer’s Disease Center, Chicago, IL 60612, USA
| | - Gail Davies
- Centre for Cognitive Aging and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, Scotland, UK
| | - Christiaan de Leeuw
- Department of Functional Genomics, VU University Amsterdam and VU Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Niina Eklund
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Public Health Genomics Unit, Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki 00014, Finland
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA 94107–1728, USA
| | - Rudolf Ferhmann
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Håkon K. Gjessing
- Department of Genes and Environment, Division of Epidemiology, Norwegian Institute of Public Health, Nydalen, N-0403 Oslo, Norway
| | - Sara Hägg
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, 751 85 Uppsala, Sweden
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jennifer R. Harris
- Department of Genes and Environment, Division of Epidemiology, Norwegian Institute of Public Health, Nydalen, N-0403 Oslo, Norway
| | - Caroline Hayward
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Christina Holzapfel
- Else Kroener-Fresenius-Centre for Nutritional Medicine, Technische Universität München, 81675 Munich, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Carla A. Ibrahim-Verbaas
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, the Netherlands
- Department of Neurology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Erik Ingelsson
- Molecular Epidemiology, Department of Medical Sciences, Uppsala University, 751 85 Uppsala, Sweden
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, 751 23 Uppsala, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Bo Jacobsson
- Department of Genes and Environment, Division of Epidemiology, Norwegian Institute of Public Health, Nydalen, N-0403 Oslo, Norway
- Department of Obstetrics and Gynecology, Institute of Public Health, Sahlgrenska Academy, Sahgrenska University Hospital, Gothenburg, 413 45, Sweden
| | - Peter K. Joshi
- Centre for Population Health Sciences, The University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Astanand Jugessur
- Department of Genes and Environment, Division of Epidemiology, Norwegian Institute of Public Health, Nydalen, N-0403 Oslo, Norway
| | - Marika Kaakinen
- Institute of Health Sciences, University of Oulu, Oulu 90014, Finland
- Biocenter Oulu, University of Oulu, Oulu 90014, Finland
| | - Stavroula Kanoni
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Juha Karjalainen
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, 21000 Split, Croatia
| | - Kati Kristiansson
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Public Health Genomics Unit, Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki 00014, Finland
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, 1005 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Jari Lahti
- Institute of Behavioral Sciences, University of Helsinki, Helsinki 00014, Finland
| | - Sang H. Lee
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Peng Lin
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Penelope A. Lind
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Yongmei Liu
- Department of Epidemiology & Prevention, Division of Public Health Sciences, Wake Forest University Health Sciences, Winston-Salem, NC 27157–1063, USA
| | - Kurt Lohman
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest University Health Sciences, Winston-Salem, NC 27157–1063, USA
| | - Marisa Loitfelder
- Division for Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz 8036, Austria
| | - George McMahon
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
| | - Pedro Marques Vidal
- Institute of Social and Preventive Medicine, Lausanne University Hospital, 1005 Lausanne, Switzerland
| | - Osorio Meirelles
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Ronny Myhre
- Department of Genes and Environment, Division of Epidemiology, Norwegian Institute of Public Health, Nydalen, N-0403 Oslo, Norway
| | - Marja-Liisa Nuotio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Public Health Genomics Unit, Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki 00014, Finland
| | - Christopher J. Oldmeadow
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Katja E. Petrovic
- Division of General Neurology, Department of Neurology, General Hospital and Medical University of Graz, Graz 8036, Austria
| | - Wouter J. Peyrot
- Department of Psychiatry, VU University Medical Center, 1081 HL Amsterdam, The Netherlands
| | - Ozren Polašek
- Faculty of Medicine, University of Split, 21000 Split, Croatia
| | - Lydia Quaye
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Eva Reinmaa
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - John P. Rice
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thais S. Rizzi
- Department of Functional Genomics, VU University Amsterdam and VU Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Graz 8036, Austria
| | - Reinhold Schmidt
- Division for Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz 8036, Austria
| | - Albert V. Smith
- Icelandic Heart Association, Kopavogur 201, Iceland
- Department of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109–2029, USA
| | - Toshiko Tanaka
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Antonio Terracciano
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
- College of Medicine, Florida State University, Tallahassee, FL 32306–4300, USA
| | - Matthijs J.H.M. van der Loos
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, 3000 DR Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Veronique Vitart
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17489, Germany
| | - Jürgen Wellmann
- Institute of Epidemiology and Social Medicine, University of Muenster, 48129 Muenster, Germany
| | - Lei Yu
- Rush University Medical Center, Rush Alzheimer’s Disease Center, Chicago, IL 60612, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109–2029, USA
| | - Jüri Allik
- Department of Psychology, University of Tartu, Tartu 50410, Estonia
| | - John R. Attia
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | | | - François Bastardot
- Department of Internal Medicine, University Hospital, 1011 Lausanne, Switzerland
| | | | - David A. Bennett
- Rush University Medical Center, Rush Alzheimer’s Disease Center, Chicago, IL 60612, USA
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Muenster, 48129 Muenster, Germany
| | - Laura J. Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - Ute Bültmann
- Department of Health Sciences, Community & Occupational Medicine, University Medical Center Groningen, 9700 AD Groningen, The Netherlands
| | - Harry Campbell
- Centre for Population Health Sciences, The University of Edinburgh, Edinburgh EH8 9AG, UK
| | | | - Lynn Cherkas
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Mina K. Chung
- Heart and Vascular and Lerner Research Institutes, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, CNR, Monserrato, 09042, Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 SS, Italy
| | - Mariza de Andrade
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Philip L. De Jager
- Program in Translational Neuropsychiatric Genomics, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jan-Emmanuel De Neve
- School of Public Policy, University College London, London WC1H 9QU, UK
- Centre for Economic Performance, London School of Economics, London WC2A 2AE, UK
| | - Ian J. Deary
- Centre for Cognitive Aging and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, Scotland, UK
- Department of Psychology, The University of Edinburgh, Edinburgh EH8 9JZ, Scotland, UK
| | - George V. Dedoussis
- Department of Nutrition and Dietetics, Harokopio University of Athens, Athens 17671, Greece
| | - Panos Deloukas
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Maria Dimitriou
- Department of Nutrition and Dietetics, Harokopio University of Athens, Athens 17671, Greece
| | | | - Martin F. Elderson
- LifeLines Cohort Study, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Johan G. Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki 00014, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki 00280, Finland
- Folkhälsan Research Center, Helsinki 00250, Finland
- Vaasa Central Hospital, Vaasa 65130, Finland
| | - David M. Evans
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48106, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Melissa E. Garcia
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur 201, Iceland
- Department of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Juliette M. Harris
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK
| | - Tamara B. Harris
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Nicholas D. Hastie
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Andrew C. Heath
- Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110–1093, USA
| | - Dena G. Hernandez
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Wolfgang Hoffmann
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17489, Germany
| | - Adriaan Hofman
- Faculty of Behavioral and Social Sciences, University of Groningen, 9747 AD Groningen, The Netherlands
| | - Rolf Holle
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Elizabeth G. Holliday
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - William G. Iacono
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455–0344, USA
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, 30625 Hannover, Germany
| | - Marjo-Riitta Järvelin
- Institute of Health Sciences, University of Oulu, Oulu 90014, Finland
- Biocenter Oulu, University of Oulu, Oulu 90014, Finland
- Department of Epidemiology and Biostatistics, MRC-HPA Centre for Environment and Health, Imperial College London, London W2 1PG, UK
- Unit of Primary Care, Oulu University Hospital, Oulu 90220, Finland
- Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu 90101, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere 33520, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Department of Public Health, University of Helsinki, 00014 Helsinki, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, 00300 Helsinki, Finland
| | | | - Matthew Kowgier
- Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Antti Latvala
- Department of Public Health, University of Helsinki, 00014 Helsinki, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, 00300 Helsinki, Finland
| | - Lenore J. Launer
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Debbie A. Lawlor
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere 33520, Finland
| | - Jingmei Li
- Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Centre Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - David C. Liewald
- Centre for Cognitive Aging and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, Scotland, UK
| | - Pamela A. Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patrik K. E. Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Tomi E. Mäkinen
- Department of Health, Functional Capacity and Welfare, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Marco Masala
- Istituto di Ricerca Genetica e Biomedica, CNR, Monserrato, 09042, Cagliari, Italy
| | - Matt McGue
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455–0344, USA
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Andreas Mielck
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Michael B. Miller
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455–0344, USA
| | - Grant W. Montgomery
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Sutapa Mukherjee
- Western Australia Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Perth, Western Australia 6009, Australia
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Women’s College Research Institute, University of Toronto, Toronto, Ontario M5G 1N8, Canada
| | - Dale R. Nyholt
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Ben A. Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, the Netherlands
| | - Lyle J. Palmer
- Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
- Department of Medical Genetics, University of Helsinki, 00014 Helsinki, Finland
| | - Brenda Penninx
- Department of Psychiatry, VU University Medical Center, 1081 HL Amsterdam, The Netherlands
| | - Markus Perola
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Public Health Genomics Unit, Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki 00014, Finland
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109–2029, USA
| | - Martin Preisig
- Department of Internal Medicine, University Hospital, 1011 Lausanne, Switzerland
| | - Katri Räikkönen
- Institute of Behavioral Sciences, University of Helsinki, Helsinki 00014, Finland
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Anu Realo
- Department of Psychology, University of Tartu, Tartu 50410, Estonia
| | - Susan M. Ring
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Public Health Genomics Unit, Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki 00014, Finland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Igor Rudan
- Centre for Population Health Sciences, The University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Aldo Rustichini
- Department of Economics, University of Minnesota, Minneapolis, MN 55455–0462, USA
| | - Veikko Salomaa
- Chronic Disease Epidemiology Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA
| | - Rodney J. Scott
- Hunter Medical Research Institute and Faculty of Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Beate St Pourcain
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
- School of Oral and Dental Sciences, University of Bristol, Bristol BS1 2LY, UK
| | - John M. Starr
- Centre for Cognitive Aging and Cognitive Epidemiology, The University of Edinburgh, Edinburgh EH8 9JZ, Scotland, UK
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh EH8 9JZ, Scotland, UK
| | - Jae Hoon Sul
- Department of Computer Science, University of California, Los Angeles, CA 90095, USA
| | - Ida Surakka
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Public Health Genomics Unit, Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki 00014, Finland
| | - Rauli Svento
- Department of Economics, Oulu Business School, University of Oulu, Oulu 90014, Finland
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald 17487, Germany
| | | | - Henning Tiemeier
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, 3000 CB Rotterdam, The Netherlands
| | - Frank JAan Rooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - David R. Van Wagoner
- Heart and Vascular and Lerner Research Institutes, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Erkki Vartiainen
- Division of Welfare and Health Promotion, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Jorma Viikari
- Department of Medicine, Turku University Hospital, Turku 20520, Finland
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital, 1011 Lausanne, Switzerland
| | - Judith M. Vonk
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Gérard Waeber
- Department of Internal Medicine, University Hospital, 1011 Lausanne, Switzerland
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48106, USA
| | - H.-Erich Wichmann
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- Klinikum Grosshadern, 81377 Munich, Germany
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764 Neuherberg, Germany
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University Amsterdam, 1081 BT Amsterdam, The Netherlands
| | - James F. Wilson
- Centre for Population Health Sciences, The University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Alan F. Wright
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Dalton Conley
- Department of Sociology, New York University, New York, NY 10012, USA
| | - George Davey-Smith
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Patrick J. F. Groenen
- Econometric Institute, Erasmus School of Economics, Erasmus University Rotterdam, Rotterdam 3000 DR, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Magnus Johannesson
- Department of Economics, Stockholm School of Economics, Stockholm 113 83, Sweden
| | - Sharon L.R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109–2029, USA
| | - Robert F. Krueger
- Department of Psychology, University of Minnesota, Minneapolis, MN 55455–0344, USA
| | - David Laibson
- Department of Economics, Harvard University, Cambridge, MA 02138, USA
| | - Nicholas G. Martin
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
| | - Michelle N. Meyer
- Petrie-Flom Center for Health Law Policy, Biotechnology, & Bioethics, Harvard Law School, Cambridge, MA 02138, USA
- Nelson A. Rockefeller Institute of Government, State University of New York, Albany, NY 12203–1003, USA
| | - Danielle Posthuma
- Department of Functional Genomics, VU University Amsterdam and VU Medical Center, 1081 HV Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus Medical Center, 3000 CB Rotterdam, The Netherlands
- Department of Clinical Genetics, VU University Medical Centrer, 1081 BT Amsterdam, The Netherlands
| | - A. Roy Thurik
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, 3000 DR Rotterdam, The Netherlands
- Panteia, Zoetermeer 2701 AA, Netherlands
- GSCM-Montpellier Business School, Montpellier 34185, France
| | - Nicholas J. Timpson
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol BS8 2PR, UK
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, the Netherlands
- Centre for Medical Systems Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter M. Visscher
- Queensland Institute of Medical Research, 300 Herston Road, Brisbane, Queensland 4006, Australia
- University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland 4102, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - David Cesarini
- Center for Experimental Social Science, Department of Economics, New York University, New York, NY 10012, USA
- Division of Social Science, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, UAE
- Research Institute of Industrial Economics, Stockholm 102 15, Sweden
| | - Philipp D. Koellinger
- Department of Applied Economics, Erasmus School of Economics, Erasmus University Rotterdam, 3000 DR Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| |
Collapse
|
6461
|
Hermsdorff HH, Mansego ML, Campión J, Milagro FI, Zulet MA, Martínez JA. TNF-alpha promoter methylation in peripheral white blood cells: relationship with circulating TNFα, truncal fat and n-6 PUFA intake in young women. Cytokine 2013; 64:265-71. [PMID: 23796695 DOI: 10.1016/j.cyto.2013.05.028] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/29/2013] [Accepted: 05/29/2013] [Indexed: 12/27/2022]
Abstract
The aim of this article is to assess the potential relationships between TNFα gene promoter methylation in peripheral white blood cells and central adiposity (truncal fat), metabolic features and dietary fat intake. A group of 40 normal-weight young women (21 ± 3y; BMI 21.0 ± 1.7 kg/m(2)) was included in this cross-sectional study. Anthropometric, biochemical and dietary data were assessed using validated procedures. DNA from white blood cells was isolated and 5-methylcytosine levels of the CpGs sites present in TNFα gene promoter (from -170 to +359 pb) were analyzed by Sequenom EpiTyper. Those women with high truncal fat (≥52.3%) showed lower 5-methylcytosine levels (P<0.05) in the site CpG13 (at position +207) and CpG19 (+317 pb) of the TNFα gene promoter when were compared to women with lower truncal adiposity. The methylation levels of CpG13 were also correlated with circulating TNFα levels, which were higher in those women with greater truncal adiposity. In a linear regression model, truncal fat, HDL-cholesterol, insulin, plasma TNFα, and daily n-6 PUFA intake explained the methylation levels of CpG13 site +207 by 48% and the average of CpG13 and CpG19 by 43% (P<0.001). In conclusion, women with higher truncal fat showed lower methylation levels of TNFα promoter in peripheral white blood cells and higher plasma TNFα concentrations. DNA methylation levels of TNFα promoter were associated with some metabolic features and with n-6 PUFA intake, suggesting a complex nutriepigenomic network in the regulation of this recognized pro-inflammatory marker.
Collapse
Affiliation(s)
- H H Hermsdorff
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, MG 36570, Brazil
| | | | | | | | | | | |
Collapse
|
6462
|
Zulfiqar F, Hozo I, Rangarajan S, Mariuzza RA, Dziarski R, Gupta D. Genetic Association of Peptidoglycan Recognition Protein Variants with Inflammatory Bowel Disease. PLoS One 2013; 8:e67393. [PMID: 23840689 PMCID: PMC3686734 DOI: 10.1371/journal.pone.0067393] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 05/17/2013] [Indexed: 12/19/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a common disease, includes Crohn's disease (CD) and ulcerative colitis (UC), and is determined by altered gut bacterial populations and aberrant host immune response. Peptidoglycan recognition proteins (PGLYRP) are innate immunity bactericidal proteins expressed in the intestine. In mice, PGLYRPs modulate bacterial populations in the gut and sensitivity to experimentally induced UC. The role of PGLYRPs in humans with CD and/or UC has not been previously investigated. Here we tested the hypothesis that genetic variants in PGLYRP1, PGLYRP2, PGLYRP3 and PGLYRP4 genes associate with CD and/or UC and with gender and/or age of onset of disease in the patient population. We sequenced all PGLYRP exons in 372 CD patients, 77 UC patients, 265 population controls, 210 familial CD controls, and 24 familial UC controls, identified all polymorphisms in these populations, and analyzed the variants for significant association with CD and UC. We identified 16 polymorphisms in the four PGLYRP genes that significantly associated with CD, UC, and/or subgroups of patient populations. Of the 16, 5 significantly associated with both CD and UC, 6 with CD, and 5 with UC. 12 significant variants result in amino acid substitutions and based on structural modeling several of these missense variants may have structural and/or functional consequences for PGLYRP proteins. Our data demonstrate that genetic variants in PGLYRP genes associate with CD and UC and may provide a novel insight into the mechanism of pathogenesis of IBD.
Collapse
Affiliation(s)
- Fareeha Zulfiqar
- Indiana University School of Medicine–Northwest, Gary, Indiana, United States of America
| | - Iztok Hozo
- Department of Mathematics, Indiana University Northwest, Gary, Indiana, United States of America
| | - Sneha Rangarajan
- The Institute of Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, United States of America
| | - Roy A. Mariuzza
- The Institute of Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland, United States of America
| | - Roman Dziarski
- Indiana University School of Medicine–Northwest, Gary, Indiana, United States of America
| | - Dipika Gupta
- Indiana University School of Medicine–Northwest, Gary, Indiana, United States of America
- * E-mail:
| |
Collapse
|
6463
|
Bradfute SB, Castillo EF, Arko-Mensah J, Chauhan S, Jiang S, Mandell M, Deretic V. Autophagy as an immune effector against tuberculosis. Curr Opin Microbiol 2013; 16:355-65. [PMID: 23790398 DOI: 10.1016/j.mib.2013.05.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 05/08/2013] [Accepted: 05/08/2013] [Indexed: 11/15/2022]
Abstract
The now well-accepted innate immunity paradigm that autophagy acts as a cell-autonomous defense against intracellular bacteria has its key origins in studies with Mycobacterium tuberculosis, an important human pathogen and a model microorganism infecting macrophages. A number of different factors have been identified that play into the anti-mycobacterial functions of autophagy, and recent in vivo studies in the mouse model of tuberculosis have uncovered additional anti-inflammatory and tissue-sparing functions of autophagy. Complementing these observations, genome wide association studies indicate a considerable overlap between autophagy, human susceptibility to mycobacterial infections and predisposition loci for inflammatory bowel disease. Finally, recent studies show that autophagy is an important regulator and effector of IL-1 responses, and that autophagy intersects with type I interferon pathology-modulating responses.
Collapse
Affiliation(s)
- Steven B Bradfute
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | | | | | |
Collapse
|
6464
|
Koláček M, Muchová J, Dvořáková M, Paduchová Z, Žitňanová I, Čierna I, Országhová Z, Székyová D, Jajcaiová-Zedníčková N, Kovács L, Ďuračková Z. Effect of natural polyphenols (Pycnogenol) on oxidative stress markers in children suffering from Crohn's disease--a pilot study. Free Radic Res 2013; 47:624-34. [PMID: 23710677 DOI: 10.3109/10715762.2013.807508] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Crohn's disease (CD) is a nonspecific, chronic inflammatory disease of the gastrointestinal tract. It is supposed that in etiopathogenesis oxidative stress (OS) plays a role. However, its precise role in the active and non-active states of disease is not known yet. We conducted a pilot study focusing on the relationship between OS of CD in remission and the possibility to influence clinical parameters and markers of OS by polyphenolic extract, Pycnogenol® (Pyc). Compared to 15 healthy controls 15 pediatric CD patients (all were in remission according to their disease activity index - PCDAI) had reduced the activity of Cu/Zn-superoxide dismutase (SOD) and increased the oxidative damage to proteins. We found negative correlations between markers of inflammation (calprotectin, CRP) as well as between PCDAI and total antioxidant capacity (TAC). Activities of antioxidant enzymes, SOD, and glutathione peroxidase (GPX) negatively correlated with calprotectin and PCDAI. Pyc (2 mg/kg) positively influenced the parameters of OS in CD patients after 10 weeks of administration.
Collapse
Affiliation(s)
- M Koláček
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6465
|
Hunt KA, Mistry V, Bockett NA, Ahmad T, Ban M, Barker JN, Barrett JC, Blackburn H, Brand O, Burren O, Capon F, Compston A, Gough SCL, Jostins L, Kong Y, Lee JC, Lek M, MacArthur DG, Mansfield JC, Mathew CG, Mein CA, Mirza M, Nutland S, Onengut-Gumuscu S, Papouli E, Parkes M, Rich SS, Sawcer S, Satsangi J, Simmonds MJ, Trembath RC, Walker NM, Wozniak E, Todd JA, Simpson MA, Plagnol V, van Heel DA. Negligible impact of rare autoimmune-locus coding-region variants on missing heritability. Nature 2013; 498:232-5. [PMID: 23698362 PMCID: PMC3736321 DOI: 10.1038/nature12170] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/08/2013] [Indexed: 02/06/2023]
Abstract
Genome-wide association studies (GWAS) have identified common variants of modest-effect size at hundreds of loci for common autoimmune diseases; however, a substantial fraction of heritability remains unexplained, to which rare variants may contribute. To discover rare variants and test them for association with a phenotype, most studies re-sequence a small initial sample size and then genotype the discovered variants in a larger sample set. This approach fails to analyse a large fraction of the rare variants present in the entire sample set. Here we perform simultaneous amplicon-sequencing-based variant discovery and genotyping for coding exons of 25 GWAS risk genes in 41,911 UK residents of white European origin, comprising 24,892 subjects with six autoimmune disease phenotypes and 17,019 controls, and show that rare coding-region variants at known loci have a negligible role in common autoimmune disease susceptibility. These results do not support the rare-variant synthetic genome-wide-association hypothesis (in which unobserved rare causal variants lead to association detected at common tag variants). Many known autoimmune disease risk loci contain multiple, independently associated, common and low-frequency variants, and so genes at these loci are a priori stronger candidates for harbouring rare coding-region variants than other genes. Our data indicate that the missing heritability for common autoimmune diseases may not be attributable to the rare coding-region variant portion of the allelic spectrum, but perhaps, as others have proposed, may be a result of many common-variant loci of weak effect.
Collapse
Affiliation(s)
- Karen A Hunt
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6466
|
Solovieff N, Cotsapas C, Lee PH, Purcell SM, Smoller JW. Pleiotropy in complex traits: challenges and strategies. Nat Rev Genet 2013; 14:483-95. [PMID: 23752797 DOI: 10.1038/nrg3461] [Citation(s) in RCA: 745] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genome-wide association studies have identified many variants that each affects multiple traits, particularly across autoimmune diseases, cancers and neuropsychiatric disorders, suggesting that pleiotropic effects on human complex traits may be widespread. However, systematic detection of such effects is challenging and requires new methodologies and frameworks for interpreting cross-phenotype results. In this Review, we discuss the evidence for pleiotropy in contemporary genetic mapping studies, new and established analytical approaches to identifying pleiotropic effects, sources of spurious cross-phenotype effects and study design considerations. We also outline the molecular and clinical implications of such findings and discuss future directions of research.
Collapse
Affiliation(s)
- Nadia Solovieff
- Center for Human Genetics Research, Massachusetts General Hospital, 185 Cambridge Street, Boston, Massachusetts 02114, USA
| | | | | | | | | |
Collapse
|
6467
|
Large sample size, wide variant spectrum, and advanced machine-learning technique boost risk prediction for inflammatory bowel disease. Am J Hum Genet 2013; 92:1008-12. [PMID: 23731541 DOI: 10.1016/j.ajhg.2013.05.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/19/2013] [Accepted: 05/02/2013] [Indexed: 02/08/2023] Open
Abstract
We performed risk assessment for Crohn's disease (CD) and ulcerative colitis (UC), the two common forms of inflammatory bowel disease (IBD), by using data from the International IBD Genetics Consortium's Immunochip project. This data set contains ~17,000 CD cases, ~13,000 UC cases, and ~22,000 controls from 15 European countries typed on the Immunochip. This custom chip provides a more comprehensive catalog of the most promising candidate variants by picking up the remaining common variants and certain rare variants that were missed in the first generation of GWAS. Given this unprecedented large sample size and wide variant spectrum, we employed the most recent machine-learning techniques to build optimal predictive models. Our final predictive models achieved areas under the curve (AUCs) of 0.86 and 0.83 for CD and UC, respectively, in an independent evaluation. To our knowledge, this is the best prediction performance ever reported for CD and UC to date.
Collapse
|
6468
|
Abstract
The aetiology of systemic lupus erythematosus (SLE) is complex and is known to involve both genetic and environmental factors. In a small number of patients, single-gene defects can lead to the development of SLE. Such genes include those encoding early components of the complement cascade and the 3'-5' DNA exonuclease TREX1. In addition, genome-wide association studies have identified single-nucleotide polymorphisms that confer some susceptibility to SLE. In this Review, we discuss selected examples of genes whose products have distinctly altered function in SLE and contribute to the pathogenic process. Specifically, we focus on the genes encoding integrin αM (ITGAM), IgG Fc receptors, sialic acid O-acetyl esterase (SIAE), the catalytic subunit of protein phosphatase PP2A (PPP2CA) and signalling lymphocytic activation molecule (SLAM) family members. Moreover, we highlight the changes in epigenetic signatures that occur in SLE. Such epigenetic modifications, which are abundantly present and might alter gene expression in the presence or absence of susceptibility variants, should be carefully considered when deconstructing the contribution of individual genes to the complex pathogenesis of SLE.
Collapse
|
6469
|
Garn H, Neves JF, Blumberg RS, Renz H. Effect of barrier microbes on organ-based inflammation. J Allergy Clin Immunol 2013; 131:1465-78. [PMID: 23726530 PMCID: PMC4592166 DOI: 10.1016/j.jaci.2013.04.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/17/2013] [Accepted: 04/23/2013] [Indexed: 12/22/2022]
Abstract
The prevalence and incidence of chronic inflammatory disorders, including allergies and asthma, as well as inflammatory bowel disease, remain on the increase. Microbes are among the environmental factors that play an important role in shaping normal and pathologic immune responses. Several concepts have been put forward to explain the effect of microbes on the development of these conditions, including the hygiene hypothesis and the microbiota hypothesis. Recently, the dynamics of the development of (intestinal) microbial colonization, its effect on innate and adaptive immune responses (homeostasis), and the role of environmental factors, such as nutrition and others, have been extensively investigated. Furthermore, there is now increasing evidence that a qualitative and quantitative disturbance in colonization (dysbiosis) is associated with dysfunction of immune responses and development of various chronic inflammatory disorders. In this article the recent epidemiologic, clinical, and experimental evidence for this interaction is discussed.
Collapse
Affiliation(s)
- Holger Garn
- Institute of Laboratory Medicine, Philipps-Universität Marburg
| | - Joana F. Neves
- Division of Gastroenterology and Hepatology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Richard S. Blumberg
- Division of Gastroenterology and Hepatology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps-Universität Marburg
| |
Collapse
|
6470
|
Liu JZ, Hov JR, Folseraas T, Ellinghaus E, Rushbrook SM, Doncheva NT, Andreassen OA, Weersma RK, Weismüller TJ, Eksteen B, Invernizzi P, Hirschfield GM, Gotthardt DN, Pares A, Ellinghaus D, Shah T, Juran BD, Milkiewicz P, Rust C, Schramm C, Müller T, Srivastava B, Dalekos G, Nöthen MM, Herms S, Winkelmann J, Mitrovic M, Braun F, Ponsioen CY, Croucher PJP, Sterneck M, Teufel A, Mason AL, Saarela J, Leppa V, Dorfman R, Alvaro D, Floreani A, Onengut-Gumuscu S, Rich SS, Thompson WK, Schork AJ, Næss S, Thomsen I, Mayr G, König IR, Hveem K, Cleynen I, Gutierrez-Achury J, Ricaño-Ponce I, van Heel D, Björnsson E, Sandford RN, Durie PR, Melum E, Vatn MH, Silverberg MS, Duerr RH, Padyukov L, Brand S, Sans M, Annese V, Achkar JP, Boberg KM, Marschall HU, Chazouillères O, Bowlus CL, Wijmenga C, Schrumpf E, Vermeire S, Albrecht M, Rioux JD, Alexander G, Bergquist A, Cho J, Schreiber S, Manns MP, Färkkilä M, Dale AM, Chapman RW, Lazaridis KN, Franke A, Anderson CA, Karlsen TH. Dense genotyping of immune-related disease regions identifies nine new risk loci for primary sclerosing cholangitis. Nat Genet 2013; 45:670-675. [PMID: 23603763 PMCID: PMC3667736 DOI: 10.1038/ng.2616] [Citation(s) in RCA: 292] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 03/29/2013] [Indexed: 12/14/2022]
Abstract
Primary sclerosing cholangitis (PSC) is a severe liver disease of unknown etiology leading to fibrotic destruction of the bile ducts and ultimately to the need for liver transplantation. We compared 3,789 PSC cases of European ancestry to 25,079 population controls across 130,422 SNPs genotyped using the Immunochip. We identified 12 genome-wide significant associations outside the human leukocyte antigen (HLA) complex, 9 of which were new, increasing the number of known PSC risk loci to 16. Despite comorbidity with inflammatory bowel disease (IBD) in 72% of the cases, 6 of the 12 loci showed significantly stronger association with PSC than with IBD, suggesting overlapping yet distinct genetic architectures for these two diseases. We incorporated association statistics from 7 diseases clinically occurring with PSC in the analysis and found suggestive evidence for 33 additional pleiotropic PSC risk loci. Together with network analyses, these findings add to the genetic risk map of PSC and expand on the relationship between PSC and other immune-mediated diseases.
Collapse
Affiliation(s)
- Jimmy Z. Liu
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Johannes Roksund Hov
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Trine Folseraas
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eva Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Simon M. Rushbrook
- Department of Gastroenterology and Hepatology, Norfolk and Norwich, University Hospitals NHS Trust, Norwich, UK
| | | | - Ole A. Andreassen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Ulleval, Oslo, Norway
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Centre Groningen, Groningen, the Netherlands
| | - Tobias J. Weismüller
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Integrated Research and Treatment Center-Transplantation (IFB-tx), Hannover Medical School, Hannover, Germany
- Current affiliation: Department of Internal Medicine 1, University Hospital of Bonn, Bonn, Germany
| | - Bertus Eksteen
- Snyder Institute of Chronic Diseases, Department of Medicine, University of Calgary, Calgary, Canada
| | - Pietro Invernizzi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano (MI), Italy
| | - Gideon M. Hirschfield
- Division of Gastroenterology, Department of Medicine, University of Toronto, Toronto, Canada
- Centre for Liver Research, NIHR Biomedical Research Unit, Birmingham, UK
| | | | - Albert Pares
- Liver Unit, Hospital Clínic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Tejas Shah
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Brian D. Juran
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, College of Medicine, Rochester, Minnesota, USA
| | - Piotr Milkiewicz
- Liver Unit and Liver Research Laboratories, Pomeranian Medical University, Szczecin, Poland
| | - Christian Rust
- Department of Medicine 2, Grosshadern, University of Munich, Munich, Germany
| | - Christoph Schramm
- 1st Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Müller
- Department of Internal Medicine, Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Brijesh Srivastava
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Georgios Dalekos
- Department of Medicine, Medical School, University of Thessaly, Larissa, Greece
- Research Laboratory of Internal Medicine, Medical School, University of Thessaly, Larissa, Greece
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Juliane Winkelmann
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Department of Neurology, Technische Universität München, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Mitja Mitrovic
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Felix Braun
- Department of General, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Medical Centre Schleswig-Holstein, Campus Kiel, Germany
| | - Cyriel Y. Ponsioen
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, the Netherlands
| | - Peter J. P. Croucher
- Department of Environmental Science, Policy, and Management, University of California, Berkeley, United States of America
| | - Martina Sterneck
- Department of Hepatobiliary Surgery and Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Teufel
- 1st Department of Medicine, University of Mainz, Mainz, Germany
| | - Andrew L. Mason
- Division of Gastroenterology and Hepatology, University of Alberta, Edmonton, Alberta, Canada
| | - Janna Saarela
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Virpi Leppa
- Public Health Genomics Unit, Institute for Molecular Medicine Finland FIMM, University of Helsinki and National Institute for Health and Welfare, Helsinki, Finland
| | - Ruslan Dorfman
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada
| | - Domenico Alvaro
- Department of Clinical Medicine, Division of Gastroenterology, Sapienza University of Rome, Rome, Italy
| | - Annarosa Floreani
- Dept. of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, Division of Endocrinology & Metabolism, University of Virginia, Charlottesville, USA
- Department of Internal Medicine, Division of Endocrinology & Metabolism, University of Virginia, Charlottesville, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, USA
| | - Wesley K. Thompson
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Andrew J. Schork
- Graduate Program in Cognitive Science, University of California, San Diego, La Jolla, CA, USA
| | - Sigrid Næss
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingo Thomsen
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Gabriele Mayr
- Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Inke R. König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Kristian Hveem
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Isabelle Cleynen
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
- Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Javier Gutierrez-Achury
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Isis Ricaño-Ponce
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - David van Heel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Einar Björnsson
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Landspitali University Hospital, Reykjavik, Iceland
| | - Richard N. Sandford
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Peter R. Durie
- Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Espen Melum
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Morten H Vatn
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- EpiGen, Campus AHUS, Akershus University Hospital, Nordbyhagen, Norway
| | - Mark S. Silverberg
- Inflammatory Bowel Disease (IBD) Group, Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital Toronto, Ontario, Canada
| | - Richard H. Duerr
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Stephan Brand
- Department of Medicine II, University Hospital Munich-Grosshadern, Ludwig-Maximilians-University Munich, Germany
| | - Miquel Sans
- Department of Digestive Diseases, Centro Médico Teknon, Barcelona, Spain
| | - Vito Annese
- Division of Gastroenterology, Istituto di Ricovero e Cura a Carattere Scientifico-Casa Sollievodella Sofferenza Hospital, San Giovanni Rotondo, Italy
- Unit of Gastroenterology SOD2, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Jean-Paul Achkar
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kirsten Muri Boberg
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Hanns-Ulrich Marschall
- Department of Internal Medicine, Institute of Medicine, Sahlgrenska Academy and University Hospital, Gothenburg, Sweden
| | - Olivier Chazouillères
- AP-HP, Hôpital Saint Antoine, Department of Hepatology, UPMC Univ Paris 06, Paris, France
| | - Christopher L. Bowlus
- Division of Gastroenterology and Hepatology, University of California Davis, Davis, CA, USA
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Erik Schrumpf
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Severine Vermeire
- Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
- Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium
| | - Mario Albrecht
- Max Planck Institute for Informatics, Saarbrücken, Germany
- Department of Bioinformatics, Institute of Biometrics and Medical Informatics, University Medicine Greifswald, Greifswald, Germany
| | | | | | - John D. Rioux
- Université de Montréal, Research Center, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Graeme Alexander
- Department of Medicine, Division of Hepatology, University of Cambridge, Cambridge, UK
| | - Annika Bergquist
- Department of Gastroenterology and Hepatology, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Judy Cho
- Department of Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
- Department for General Internal Medicine, Christian-Albrechts-University, Kiel, Germany
- Popgen Biobank, University Hospital Schleswig-Holstein, Christian-Albrechts-University, 24105 Kiel, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
- Integrated Research and Treatment Center-Transplantation (IFB-tx), Hannover Medical School, Hannover, Germany
| | - Martti Färkkilä
- Division of Gastroenterology, Department of Medicine, Helsinki University Hospital, Finland
| | - Anders M. Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Roger W. Chapman
- Department of Hepatology, John Radcliffe University Hospitals NHS Trust, Oxford, UK
| | - Konstantinos N. Lazaridis
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, College of Medicine, Rochester, Minnesota, USA
| | | | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Carl A. Anderson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Tom H. Karlsen
- Norwegian PSC Research Center, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Division of Gastroenterology, Institute of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
6471
|
Denson LA. Linking genetic variation to phenotype: eQTL analysis of normal human ileum. Gastroenterology 2013; 144:1339-41. [PMID: 23623963 DOI: 10.1053/j.gastro.2013.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
6472
|
Hinks A, Cobb J, Marion MC, Prahalad S, Sudman M, Bowes J, Martin P, Comeau ME, Sajuthi S, Andrews R, Brown M, Chen WM, Concannon P, Deloukas P, Edkins S, Eyre S, Gaffney PM, Guthery SL, Guthridge JM, Hunt SE, James JA, Keddache M, Moser KL, Nigrovic PA, Onengut-Gumuscu S, Onslow ML, Rosé CD, Rich SS, Steel KJA, Wakeland EK, Wallace CA, Wedderburn LR, Woo P, Bohnsack JF, Haas JP, Glass DN, Langefeld CD, Thomson W, Thompson SD. Dense genotyping of immune-related disease regions identifies 14 new susceptibility loci for juvenile idiopathic arthritis. Nat Genet 2013; 45:664-9. [PMID: 23603761 PMCID: PMC3673707 DOI: 10.1038/ng.2614] [Citation(s) in RCA: 276] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 03/25/2013] [Indexed: 12/15/2022]
Abstract
We used the Immunochip array to analyze 2,816 individuals with juvenile idiopathic arthritis (JIA), comprising the most common subtypes (oligoarticular and rheumatoid factor-negative polyarticular JIA), and 13,056 controls. We confirmed association of 3 known JIA risk loci (the human leukocyte antigen (HLA) region, PTPN22 and PTPN2) and identified 14 loci reaching genome-wide significance (P < 5 × 10(-8)) for the first time. Eleven additional new regions showed suggestive evidence of association with JIA (P < 1 × 10(-6)). Dense mapping of loci along with bioinformatics analysis refined the associations to one gene in each of eight regions, highlighting crucial pathways, including the interleukin (IL)-2 pathway, in JIA disease pathogenesis. The entire Immunochip content, the HLA region and the top 27 loci (P < 1 × 10(-6)) explain an estimated 18, 13 and 6% of the risk of JIA, respectively. In summary, this is the largest collection of JIA cases investigated so far and provides new insight into the genetic basis of this childhood autoimmune disease.
Collapse
Affiliation(s)
- Anne Hinks
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6473
|
Ananthakrishnan AN. Environmental risk factors for inflammatory bowel disease. Gastroenterol Hepatol (N Y) 2013; 9:367-374. [PMID: 23935543 PMCID: PMC3736793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are chronic immunologically mediated diseases that often have a relapsing-remitting course in young persons. Genetic-risk polymorphisms explain less than one third of the heritability of disease. Epidemiologic and laboratory data suggest that environmental factors play a significant role in influencing the risk and natural history of disease. Smoking is the most widely and consistently described risk factor. It, however, increases the risk of CD while conferring protection against UC. The gut microbiome is a key component in the development of inflammatory bowel disease (IBD). Several external factors potentially exert an effect by influencing the composition of the gut microbiome or disrupting the intestinal barrier. These external influences include the use of antibiotics or nonsteroidal anti-inflammatory drugs and the presence of enteric infections. Data on diet have been inconsistent, but high fiber intake, particularly of soluble fiber, appears to protect against CD, whereas protein intake may increase disease risk. Vitamin D may also play an important protective role, particularly in patients with CD. Neurobehavioral factors, such as stress and depression, also influence the risk of IBD. Systematic and rigorous studies of environmental exposures in the management of IBD are needed. In particular, studies of whether environmental factors can be modified to reduce the likelihood of relapse or improve patient outcomes would be valuable.
Collapse
Affiliation(s)
- Ashwin N Ananthakrishnan
- Dr. Ananthakrishnan is an Assistant Professor of Medicine in the Division of Gastroenterology at the Massachusetts General Hospital and Harvard Medical School in Boston, Massachusetts
| |
Collapse
|
6474
|
Belkaid Y, Bouladoux N, Hand TW. Effector and memory T cell responses to commensal bacteria. Trends Immunol 2013; 34:299-306. [PMID: 23643444 PMCID: PMC3733441 DOI: 10.1016/j.it.2013.03.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 02/08/2023]
Abstract
Barrier surfaces are home to a vast population of commensal organisms that together encode millions of proteins; each of them possessing several potential foreign antigens. Regulation of immune responses to this enormous antigenic load represents a tremendous challenge for the immune system. Tissues exposed to commensals have developed elaborate systems of regulation including specialized populations of resident lymphocytes that maintain barrier function and limit potential responses to commensal antigens. However, in settings of infection and inflammation these regulatory mechanisms are compromised and specific effector responses against commensal bacteria can develop. This review discusses the circumstances controlling the fate of commensal specific T cells and how dysregulation of these responses could lead to severe pathological outcomes.
Collapse
Affiliation(s)
- Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, NIH, Bethesda 20892, USA.
| | | | | |
Collapse
|
6475
|
HIRSCHFIELD GIDEONM, CHAPMAN ROGERW, KARLSEN TOMH, LAMMERT FRANK, LAZARIDIS KONSTANTINOSN, MASON ANDREWL. The genetics of complex cholestatic disorders. Gastroenterology 2013; 144:1357-74. [PMID: 23583734 PMCID: PMC3705954 DOI: 10.1053/j.gastro.2013.03.053] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/24/2013] [Accepted: 03/27/2013] [Indexed: 02/07/2023]
Abstract
Cholestatic liver diseases are caused by a range of hepatobiliary insults and involve complex interactions among environmental and genetic factors. Little is known about the pathogenic mechanisms of specific cholestatic diseases, which has limited our ability to manage patients with these disorders. However, recent genome-wide studies have provided insight into the pathogenesis of gallstones, primary biliary cirrhosis, and primary sclerosing cholangitis. A lithogenic variant in the gene that encodes the hepatobiliary transporter ABCG8 has been identified as a risk factor for gallstone disease; this variant has been associated with altered cholesterol excretion and metabolism. Other variants of genes encoding transporters that affect the composition of bile have been associated with cholestasis, namely ABCB11, which encodes the bile salt export pump, and ABCB4, which encodes hepatocanalicular phosphatidylcholine floppase. In contrast, studies have associated primary biliary cirrhosis and primary sclerosing cholangitis with genes encoding major histocompatibility complex proteins and identified loci associated with microbial sensing and immune regulatory pathways outside this region, such as genes encoding IL12, STAT4, IRF5, IL2 and its receptor (IL2R), CD28, and CD80. These discoveries have raised interest in the development of reagents that target these gene products. We review recent findings from genetic studies of patients with cholestatic liver disease. Future characterization of genetic variants in animal models, stratification of risk alleles by clinical course, and identification of interacting environmental factors will increase our understanding of these complex cholestatic diseases.
Collapse
Affiliation(s)
- GIDEON M. HIRSCHFIELD
- Centre for Liver Research, National Institute for Health Research Biomedical Research Unit, University of Birmingham, Birmingham, England
| | - ROGER W. CHAPMAN
- Department of Gastroenterology, John Radcliffe Hospital, Oxford, England
| | - TOM H. KARLSEN
- Research Institute for Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - FRANK LAMMERT
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
| | - KONSTANTINOS N. LAZARIDIS
- Center for Basic Research in Digestive Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - ANDREW L. MASON
- Centre of Excellence in Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6476
|
Russell RK, Protheroe A, Roughton M, Croft NM, Murphy MS, Spray C, Rodrigues AF, Wilson DC, Puntis J, Cosgrove M, Tamok A, Rao P, Down C, Arnott IDR, Mitton SG. Contemporary outcomes for ulcerative colitis inpatients admitted to pediatric hospitals in the United Kingdom. Inflamm Bowel Dis 2013; 19:1434-40. [PMID: 23624885 DOI: 10.1097/mib.0b013e31828133d6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pediatric ulcerative colitis (UC) care is variable with a lack of appropriate guidelines to guide practice until recently. METHODS UC inpatients <17 years old admitted to 23 U.K. pediatric hospitals had clinical details collected between September 2010 and 2011. Comparative data for 248 patients were available from a previous audit in 2008. RESULTS One hundred and seventy-six patients (98 males) of median age 13 years (interquartile range, 10-13) were analyzed; 23 were elective surgical admissions, 47 new diagnoses, and 106 needed acute medical care for established UC. Median length of stay was 6 days (interquartile range, 3-10) with no deaths. Eighty-eight of 126 patients (70%) with active disease had standard stool cultures performed (3 [2%] were positive), and 57 (45%) had Clostridium difficile toxin tested (none positive). Twenty-five of 66 (38%) emergency admissions had an abdominal x-ray on admission, and 13 of 66 patients (20%) had a Pediatric Ulcerative Colitis Activity Index score. There were 3 cases of toxic megacolon and 2 thromboses. Eighty-one of 116 patients (71%) responded to steroids. Nineteen patients who did not respond adequately to steroids received rescue therapy (7 infliximab, 11 ciclosporin, and 1 both) with overall response rate of 90%; 7 patients needed surgery acutely, 5 without previous rescue therapy. Compared with the 2008 data, stool culture rates improved significantly (86 of 121 [71%] versus 76 of 147 [52%], P = 0.001) as did heparinization rates (15 of 150 [10%] versus 5 of 215 [2%], P = 0.002) and rescue therapy usage (17 of 33 [52%] versus 10 of 38 [26%], P = 0.03). CONCLUSIONS There were signs of improving UC care with significantly increased rates of stool culture and rescue therapy. The majority of sites, however, did not use Pediatric Ulcerative Colitis Activity Index scores.
Collapse
Affiliation(s)
- Richard K Russell
- Department of Paediatric Gastroenterology, Yorkhill Hospital, Glasgow, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6477
|
Kabakchiev B, Silverberg MS. Expression quantitative trait loci analysis identifies associations between genotype and gene expression in human intestine. Gastroenterology 2013; 144:1488-96, 1496.e1-3. [PMID: 23474282 PMCID: PMC3775712 DOI: 10.1053/j.gastro.2013.03.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/30/2013] [Accepted: 03/01/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND & AIMS Genome-wide association studies have greatly increased our understanding of intestinal disease. However, little is known about how genetic variations result in phenotypic changes. Some polymorphisms have been shown to modulate quantifiable phenotypic traits; these are called quantitative trait loci. Quantitative trait loci that affect levels of gene expression are called expression quantitative trait loci (eQTL), which can provide insight into the biological relevance of data from genome-wide association studies. We performed a comprehensive eQTL scan of intestinal tissue. METHODS Total RNA was extracted from ileal biopsy specimens and genomic DNA was obtained from whole-blood samples from the same cohort of individuals. Cis- and trans-eQTL analyses were performed using a custom software pipeline for samples from 173 subjects. The analyses determined the expression levels of 19,047 unique autosomal genes listed in the US National Center for Biotechnology Information database and more than 580,000 variants from the Single Nucleotide Polymorphism database. RESULTS The presence of more than 15,000 cis- and trans-eQTL was detected with statistical significance. eQTL associated with the same expression trait were in high linkage disequilibrium. Comparative analysis with previous eQTL studies showed that 30% to 40% of genes identified as eQTL in monocytes, liver tissue, lymphoblastoid cell lines, T cells, and fibroblasts are also eQTL in ileal tissue. Conversely, most of the significant eQTL have not been previously identified and could be tissue specific. These are involved in many cell functions, including division and antigen processing and presentation. Our analysis confirmed that previously published cis-eQTL are single nucleotide polymorphisms associated with inflammatory bowel disease: rs2298428/UBE2L3, rs1050152/SLC22A4, and SLC22A5. We identified many new associations between inflammatory bowel disease susceptibility loci and gene expression. CONCLUSIONS eQTL analysis of intestinal tissue supports findings that some eQTL remain stable across cell types, whereas others are specific to the sampled location. Our findings confirm and expand the number of known genotypes associated with expression and could help elucidate mechanisms of intestinal disease.
Collapse
Affiliation(s)
- Boyko Kabakchiev
- Zane Cohen Centre for Digestive Diseases, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
6478
|
Qin X. Have genome-wide association studies or knockout mice more reflected the true nature of inflammatory bowel disease? J Crohns Colitis 2013; 7:419-420. [PMID: 23403038 DOI: 10.1016/j.crohns.2013.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 01/20/2013] [Indexed: 02/08/2023]
Affiliation(s)
- Xiaofa Qin
- Department of Surgery, UMDNJ-New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
6479
|
Vazquez P, Garrido JM, Juste RA. Specific antibody and interferon-gamma responses associated with immunopathological forms of bovine paratuberculosis in slaughtered Friesian cattle. PLoS One 2013; 8:e64568. [PMID: 23724062 PMCID: PMC3665815 DOI: 10.1371/journal.pone.0064568] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/16/2013] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium avium subsp. paratuberculosis (MAP) infection causes a chronic granulomatous inflammatory regional enteritis in ruminants. Cell-mediated immune responses are assumed to be protective and therefore, to be associated with its more delimited lesion types, while humoral responses are mainly associated with diffuse histopathological lesions. However, this duality of immune responses has been recently questioned. The aim of this study was to assess the relationship between both types of immunological responses and the type and extension of intestinal lesions and the presence of MAP in bovine tissues. Standard histopathological examinations, two microbiological procedures (culture and real time PCR (rtPCR)), as well as MAP specific antibody and interferon gamma (IFN-γ) release assays (IGRA) were performed on tissues and blood of 333 slaughtered Holstein-Friesian animals. Paratuberculous lesions were observed in 176 (52.9%) of the animals and overall MAP detection rates were estimated at 13.5% and 28.5% for tissue culture and rtPCR, respectively. Unlike the relatively constant non-specific IFN-γ release, both the antibody levels and the specific IFN-γ release significantly increased with tissue damage. Delimited immunopathological forms, which accounted for 93.2% of all forms, were mostly related to positive testing in the IGRA (38.4%) whereas diffuse ones (6.8%) were associated with antibody seropositivity (91.7%). However, since the frequency of positive immune responses in both tests increased as the lesions severity increased, polarization of Th1/Th2 responses was less prominent than expected. MAP was detected in the majority of ELISA-positive animals (culture+: 90%, rtPCR+: 85%) but the bacteria was only confirmed in the 36.1% of IGRA-positive animals by any of the two microbiological tests. In terms of diagnosis, the antibody test was a good indicator of advanced tissue damage (diffuse forms), but the IGRA did not associate well with more delimited forms or with MAP detection.
Collapse
Affiliation(s)
- Patricia Vazquez
- Department of Animal Health, NEIKER-Tecnalia, Derio, Bizkaia, Spain
| | | | - Ramon A. Juste
- Department of Animal Health, NEIKER-Tecnalia, Derio, Bizkaia, Spain
- * E-mail:
| |
Collapse
|
6480
|
Rao A, Standing JF, Naik S, Savage MO, Sanderson IR. Mathematical modelling to restore circulating IGF-1 concentrations in children with Crohn's disease-induced growth failure: a pharmacokinetic study. BMJ Open 2013; 3:bmjopen-2013-002737. [PMID: 23793696 PMCID: PMC3664353 DOI: 10.1136/bmjopen-2013-002737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES Children with Crohn's disease grow poorly, and inflammation depresses the response of insulin-like growth factor-1 (IGF-1) to growth hormone. Correcting the inflammation normalises growth velocity; however, removing inflammation cannot be achieved in all children. Our lack of understanding of IGF-1 kinetics has hampered its use, particularly as high IGF-1 concentrations over long periods may predispose to colon cancer. We hypothesised that mathematical modelling of IGF-1 would define dosing regimes that return IGF-1 concentrations into the normal range, without reaching values that risk cancer. DESIGN Pharmacokinetic intervention study. SETTING Tertiary paediatric gastroenterology unit. PARTICIPANTS 8 children (M:F; 4:4) entered the study. All completed: 5 South Asian British; 2 White British; 1 African British. INCLUSION CRITERIA Children over 10 years with active Crohn's disease (C reactive protein >10 mg/l or erythrocyte sedimentation rate >25 mm/h) and height velocity <-2 SD score. EXCLUSION CRITERIA closed epiphyses; corticosteroids within 3 months; neoplasia or known hypersensitivity to recombinant human IGF-1 (rhIGF-1). INTERVENTIONS Subcutaneous rhIGF-1 (120 μg/kg) per dose over two admissions: the first as a single dose and the second as twice daily doses over 5 days. PRIMARY OUTCOME Significant increase in circulating IGF-1. SECONDARY OUTCOMES Incidence of side effects of IGF-1. A mathematical model of circulating IGF-1 (Ac) was developed to include parameters of endogenous synthesis (Ksyn); exogenous uptake (Ka) from the subcutaneous dose (As): and IGF-1 clearance: where dAc/dt=Ksyn - Kout×Ac+Ka×As. RESULTS Subcutaneous IGF-1 increased concentrations, which were maintained on twice daily doses. In covariate analysis, disease activity reduced Ksyn (p<0.001). Optimal dosing was derived from least squares regression fitted to a dataset of 384 Crohn's patients, with model parameters assigned by simulation. CONCLUSIONS By using age, weight and disease activity scaling in IGF-1 dosing, over 95% of children will have normalised IGF-1 concentrations below +2.5 SDs of the normal population mean, a level not associated with cancer risk.
Collapse
Affiliation(s)
- A Rao
- Centre for Digestive Diseases, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, London, England, UK
| | | | | | | | | |
Collapse
|
6481
|
Abstract
Inflammatory bowel diseases [IBD; Crohn's disease (CD), ulcerative colitis (UC)] are chronic immunologically mediated diseases that are due to a dysregulated immune response to intestinal flora in a genetically susceptible host. Despite advances in genetics, the likelihood of occurrence of disease remains incompletely explained and there appears to be a strong role for the environment in mediating risk of disease. Smoking remains the most widely studied and replicated risk factor, contributing to increased risk and severity of CD while conferring protection against UC. Recent data has suggested novel risk factors. Lower plasma vitamin D is associated with an increased risk of Crohn's disease, and vitamin D supplementation may prevent relapse of disease. Several medications including oral contraceptives, post-menopausal hormone replacement, aspirin, NSAIDs, and antibiotics may increase risk of CD or UC with the mechanisms of effect remaining inadequately defined. There is continuing evidence that depression and psychosocial stress may play a role in the pathogenesis of both CD and UC, while at the same time also increasing risk for disease flares. There is also a growing understanding of the role of diet on IBD, in particular through its effect on the microbiome. Animal protein intake and n-6 fatty acids may increase risk of UC while n-3 fatty acids and dietary fiber may confer protection. The effect of diet on established disease remains poorly studied. There is need for routine measurement of a spectrum of environmental exposures in prospective studies to further our understanding.
Collapse
|
6482
|
Hendriks WJAJ, Pulido R. Protein tyrosine phosphatase variants in human hereditary disorders and disease susceptibilities. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1673-96. [PMID: 23707412 DOI: 10.1016/j.bbadis.2013.05.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/14/2013] [Accepted: 05/16/2013] [Indexed: 12/18/2022]
Abstract
Reversible tyrosine phosphorylation of proteins is a key regulatory mechanism to steer normal development and physiological functioning of multicellular organisms. Phosphotyrosine dephosphorylation is exerted by members of the super-family of protein tyrosine phosphatase (PTP) enzymes and many play such essential roles that a wide variety of hereditary disorders and disease susceptibilities in man are caused by PTP alleles. More than two decades of PTP research has resulted in a collection of PTP genetic variants with corresponding consequences at the molecular, cellular and physiological level. Here we present a comprehensive overview of these PTP gene variants that have been linked to disease states in man. Although the findings have direct bearing for disease diagnostics and for research on disease etiology, more work is necessary to translate this into therapies that alleviate the burden of these hereditary disorders and disease susceptibilities in man.
Collapse
Affiliation(s)
- Wiljan J A J Hendriks
- Department of Cell Biology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | |
Collapse
|
6483
|
|
6484
|
Somma V, Ababneh H, Ababneh A, Gatti S, Romagnoli V, Bendia E, Conrad K, Bogdanos DP, Roggenbuck D, Ciarrocchi G. The Novel Crohn's Disease Marker Anti-GP2 Antibody Is Associated with Ileocolonic Location of Disease. Gastroenterol Res Pract 2013; 2013:683824. [PMID: 23762038 PMCID: PMC3671301 DOI: 10.1155/2013/683824] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 03/19/2013] [Indexed: 12/19/2022] Open
Abstract
Crohn's disease (CD) is an inflammatory bowel disease (IBD) that can affect the whole gastrointestinal tract. The ileocolonic variant of CD, an inflammation of both the ileum and the large intestine, accounts for up to 50% of the cases with CD, whereas Crohn's ileitis affecting the ileum is diagnosed in about 30%. Crohn's colitis, which is confined to the large intestine and accounts for the remaining 20%, is difficult to distinguish from the large bowel inflammation seen in patients with ulcerative colitis (UC). The pathogenesis of CD is not yet completely understood. Autoimmunity is one factor that can partake in the triggering or modulation of inflammatory processes in IBD. The major zymogen-granule membrane glycoprotein 2 (GP2) has been recently identified as a major autoantigenic target in CD. Interestingly, GP2 is mainly expressed in the pancreas and has also been demonstrated to be a membrane-anchored receptor of microfold cells in the follicle-associated epithelium. Remarkably, GP2 is overexpressed at the site of CD inflammation in contrast to the one in UC. By utilizing novel enzyme-linked immunosorbent assays for the detection of GP2-specific IgA and IgG, the loss of tolerance to GP2 has been associated with a specific clinical phenotype in CD, in particular with the ileocolonic location of the disease.
Collapse
Affiliation(s)
| | - Hani Ababneh
- Immunology Department, King Hussein Medical Center, Amman 11855, Jordan
| | | | - Simona Gatti
- Department of Pediatrics, Polytechnic University of Marche, 60123 Ancona, Italy
| | - Vittorio Romagnoli
- Department of Pediatrics, Polytechnic University of Marche, 60123 Ancona, Italy
| | - Emanuele Bendia
- Department of Gastroenterology, “Ospedali Riuniti” University Hospital, 60020 Ancona, Italy
| | - Karsten Conrad
- Institute of Immunology, Technical University, 01307 Dresden, Germany
| | - Dimitrios P. Bogdanos
- Division of Transplantation Immunology and Mucosal Biology, Institute of Liver Studies, King's College London School of Medicine at King's College Hospital, Denmark Hill Campus, London SE5 9RJ, UK
| | - Dirk Roggenbuck
- R/D, Medipan GmbH, 15827 Dahlewitz/Berlin, Germany
- Faculty of Natural Sciences, Lausitz University of Applied Sciences, 01968 Senftenberg, Germany
| | - Gino Ciarrocchi
- Central Analytical Laboratory, “Ospedali Riuniti” University Hospital, 60020 Ancona, Italy
| |
Collapse
|
6485
|
Bargagna-Mohan P, Deokule SP, Thompson K, Wizeman J, Srinivasan C, Vooturi S, Kompella UB, Mohan R. Withaferin A effectively targets soluble vimentin in the glaucoma filtration surgical model of fibrosis. PLoS One 2013; 8:e63881. [PMID: 23667686 PMCID: PMC3648549 DOI: 10.1371/journal.pone.0063881] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/09/2013] [Indexed: 01/23/2023] Open
Abstract
Withaferin A (WFA) is a natural product that binds to soluble forms of the type III intermediate filament (IF) vimentin. Currently, it is unknown under what pathophysiological contexts vimentin is druggable, as cytoskeltal vimentin-IFs are abundantly expressed. To investigate druggability of vimentin, we exploited rabbit Tenon's capsule fibroblast (RbTCF) cell cultures and the rabbit glaucoma filtration surgical (GFS) model of fibrosis. WFA potently caused G₀/G₁ cell cycle inhibition (IC₅₀ 25 nM) in RbTCFs, downregulating ubiquitin E3 ligase skp2 and inducing p27(Kip1) expression. Transforming growth factor (TGF)-ß-induced myofibroblast transformation caused development of cell spheroids with numerous elongated invadopodia, which WFA blocked potently by downregulating soluble vimentin and α-smooth muscle actin (SMA) expression. In the pilot proof-of-concept study using the GFS model, subconjunctival injections of a low WFA dose reduced skp2 expression in Tenon's capsule and increased p27(Kip1) expression without significant alteration to vimentin-IFs. This treatment maintains significant nanomolar WFA concentrations in anterior segment tissues that correspond to WFA's cell cycle targeting activity. A ten-fold higher WFA dose caused potent downregulation of soluble vimentin and skp2 expression, but as found in cell cultures, no further increase in p27(Kip1) expression was observed. Instead, this high WFA dose potently induced vimentin-IF disruption and downregulated α-SMA expression that mimicked WFA activity in TGF-ß-treated RbTCFs that blocked cell contractile activity at submicromolar concentrations. These findings illuminate that localized WFA injection to ocular tissues exerts pharmacological control over the skp2-p27(Kip1) pathway by targeting of soluble vimentin in a model of surgical fibrosis.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Sunil P. Deokule
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kyle Thompson
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - John Wizeman
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Cidambi Srinivasan
- Statistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sunil Vooturi
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Uday B. Kompella
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Royce Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
6486
|
Rhodes G, Henrys P, Thomson BC, Pickup RW. Mycobacterium avium subspecies paratuberculosis is widely distributed in British soils and waters: implications for animal and human health. Environ Microbiol 2013; 15:2761-74. [PMID: 23648004 DOI: 10.1111/1462-2920.12137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 03/24/2013] [Indexed: 12/25/2022]
Abstract
In the first comprehensive geographical survey of distribution in Great Britain, Mycobacterium avium ssp. paratuberculosis (MAP) was detected in 115 of 1092 (10.5%) soil cores, in the range of 5 × 10(2) to 3 × 10(6) MAP cell equivalents (CE) g(-1) wet weight soil with the majority of the positive PCR reactions (n = 75; 65%) occurring around the limit of detection (500-5000 CE g(-1) wet weight soil). The distribution of MAP significantly increased from North to South and was significantly correlated with increasing cattle numbers over the same longitudinal axis. Similarly MAP occurrence significantly increased towards easterly latitudes although none of the parameters measured were associated. Comparisons of land use indicated that MAP was widely distributed in both farming and non-farming areas. Soil core samples taken from the rivers Wyre and Douglas catchments (Lancashire, UK) and river Tywi (South Wales) were negative for MAP. However, river monitoring showed a consistent presence of MAPs throughout those catchments over a 6-month period. We concluded that MAP is widely distributed within and outside the confines of the farming environment; its geographical distribution is wider than originally anticipated and; monitoring rivers describes the MAP status of catchment better than individual soil samples.
Collapse
Affiliation(s)
- Glenn Rhodes
- Centre for Ecology and Hydrology, Lancaster Environment Centre, Lancaster, LA1 4AP, UK
| | | | | | | |
Collapse
|
6487
|
Verdier J, Deroche L, Allez M, Loy C, Biet F, Bodier CC, Bay S, Ganneau C, Matysiak-Budnik T, Reyrat JM, Heyman M, Cerf-Bensussan N, Ruemmele FM, Ménard S. Specific IgG response against Mycobacterium avium paratuberculosis in children and adults with Crohn's disease. PLoS One 2013; 8:e62780. [PMID: 23658774 PMCID: PMC3642204 DOI: 10.1371/journal.pone.0062780] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 03/25/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIMS Presence of serum antibodies against Mycobacterium avium paratuberculosis (MAP) in Crohn's Disease (CD) as a disease characteristic remains controversial. In the present work, we assessed antibody reactivity of serum and intestinal fluid against four distinct MAP-antigens, including the recently identified MAP-specific lipopentapeptide (L5P). METHODS Immunoglobulin concentrations and specificity against 3 non MAP-specific antigens: glycosyl-transferase-d (GSD), purified protein derivative from MAP (Johnin-PPD), heparin binding haemagglutinin (MAP-HBHA) and one MAP-specific antigen: synthetic L5P were determined by ELISA in gut lavage fluids from adult controls or patients with CD, and in sera of children or adult controls or patients with CD, ulcerative colitis or celiac disease. RESULTS Total IgA and IgG concentrations were increased in sera of children with CD but were decreased in sera of adults with CD, thereof specificity against MAP antigens was assessed by normalizing immunoglobulin concentrations between samples. In CD patients, IgG reactivity was increased against the four MAP antigens, including L5P in gut lavage fluids but it was only increased against L5P in sera. By contrast, anti-L5P IgG were not increased in patients with ulcerative colitis or celiac disease. CONCLUSIONS A significant increase in anti-L5P IgG is observed in sera of children and adults with CD but not in patients with other intestinal inflammatory diseases. Anti-L5P antibodies may serve as serological marker for CD.
Collapse
Affiliation(s)
- Julien Verdier
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
| | - Louis Deroche
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, APHP, Paris, France
| | - Caroline Loy
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
| | - Franck Biet
- UMR ISP 1282, Infectiologie et Santé Publique,INRA centre de Tours, Nouzilly, France
| | - Christelle C. Bodier
- UMR ISP 1282, Infectiologie et Santé Publique,INRA centre de Tours, Nouzilly, France
| | - Sylvie Bay
- Institut Pasteur, Unité de Chimie des Biomolécules, Département de Biologie Structurale et Chimie, Paris, France
- CNRS UMR 3523, Paris, France
| | - Christelle Ganneau
- Institut Pasteur, Unité de Chimie des Biomolécules, Département de Biologie Structurale et Chimie, Paris, France
- CNRS UMR 3523, Paris, France
| | | | - Jean Marc Reyrat
- Université Paris Descartes, Sorbonne Paris Cité, France
- INSERM-UMR 570, unité de Pathogénie des Infections Systémiques, Groupe Avenir, Paris, France
| | - Martine Heyman
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
| | - Nadine Cerf-Bensussan
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
| | - Frank M. Ruemmele
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
- APHP, Hôpital Necker-Enfants Malades, Service de Gastroenterology, Paris, France
| | - Sandrine Ménard
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR989, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, France
- Neuro-Gastroenterology and Nutrition Unit, Toxalim Research Center, Institut National de la Recherche Agronomique, Toulouse, France
- * E-mail:
| |
Collapse
|
6488
|
Haplotype-tagging analysis of common variants of the IL23R gene demonstrates gene-wide extent of association with IBD. Inflamm Bowel Dis 2013; 19:E79-80. [PMID: 23535247 DOI: 10.1097/mib.0b013e318280e790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
6489
|
Cavanaugh JA. Genetics of inflammatory bowel disease: the state of play. J Gastroenterol Hepatol 2013; 28:759-60. [PMID: 23614340 DOI: 10.1111/jgh.12120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2012] [Indexed: 12/09/2022]
|
6490
|
Stevens C, Henderson P, Nimmo ER, Soares DC, Dogan B, Simpson KW, Barrett JC, Wilson DC, Satsangi J. The intermediate filament protein, vimentin, is a regulator of NOD2 activity. Gut 2013; 62:695-707. [PMID: 22684479 PMCID: PMC4225453 DOI: 10.1136/gutjnl-2011-301775] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Mutations in the nucleotide-binding oligomerisation domain-containing protein 2 (NOD2) gene remain the strongest genetic determinants for Crohn's disease (CD). Having previously identified vimentin as a novel NOD2-interacting protein, the authors aimed to investigate the regulatory effects of vimentin on NOD2 function and the association of variants in Vim with CD susceptibility. DESIGN Coimmunoprecipitation, fluorescent microscopy and fractionation were used to confirm the interaction between NOD2 and vimentin. HEK293 cells stably expressing wild-type NOD2 or a NOD2 frameshift variant (L1007fs) and SW480 colonic epithelial cells were used alongside the vimentin inhibitor, withaferin A (WFA), to assess effects on NOD2 function using the nuclear factor-kappaB (NF-κB) reporter gene, green fluorescent protein-LC3-based autophagy, and bacterial gentamicin protection assays. International genome-wide association meta-analysis data were used to test for associations of single-nucleotide polymorphisms in Vim with CD susceptibility. RESULTS The leucine-rich repeat domain of NOD2 contained the elements required for vimentin binding; CD-associated polymorphisms disrupted this interaction. NOD2 and vimentin colocalised at the cell plasma membrane, and cytosolic mislocalisation of the L1007fs and R702W variants correlated with an inability to interact with vimentin. Use of WFA demonstrated that vimentin was required for NOD2-dependent NF-κB activation and muramyl dipeptide-induced autophagy induction, and that NOD2 and vimentin regulated the invasion and survival properties of a CD-associated adherent-invasive Escherichia coli strain. Genetic analysis revealed an association signal across the haplotype block containing Vim. CONCLUSION Vimentin is an important regulator of NOD2 function and a potential novel therapeutic target in the treatment of CD. In addition, Vim is a candidate susceptibility gene for CD, supporting the functional data.
Collapse
Affiliation(s)
- Craig Stevens
- Centre for Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
6491
|
|
6492
|
Roberts CL, Rushworth SL, Richman E, Rhodes JM. Hypothesis: Increased consumption of emulsifiers as an explanation for the rising incidence of Crohn's disease. J Crohns Colitis 2013; 7:338-41. [PMID: 23360575 DOI: 10.1016/j.crohns.2013.01.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 01/03/2013] [Indexed: 02/08/2023]
Abstract
Crohn's disease (CD) incidence has increased over the past fifty years but the explanation is unclear. CD can be brought into remission by liquid enteral feeding, but the mechanism for this response is unknown. We suggest that consumption of emulsifiers in processed foods may promote CD by increasing bacterial translocation. This is supported by evidence that (i) geographical variation in CD correlates with emulsifier consumption as does the increasing incidence of CD in Japan; (ii) although CD incidence also correlates with fat consumption, the response to enteral feeding is not affected by the fat content of the feed and (iii) very small concentrations of the emulsifier polysorbate 80 enhance bacterial translocation across intestinal epithelia. Undigested emulsifiers may increase bacterial translocation, particularly in the small intestine where the mucus layer is discontinuous. The hypothesis should be testable by trials of enteral feeding with/without emulsifiers.
Collapse
Affiliation(s)
- Carol L Roberts
- Department of Gastroenterology, University of Liverpool, Duncan Building, Daulby Street, L69 3GA, United Kingdom
| | | | | | | |
Collapse
|
6493
|
Latiano A, Palmieri O, Pastorelli L, Vecchi M, Pizarro TT, Bossa F, Merla G, Augello B, Latiano T, Corritore G, Settesoldi A, Valvano MR, D'Incà R, Stronati L, Annese V, Andriulli A. Associations between genetic polymorphisms in IL-33, IL1R1 and risk for inflammatory bowel disease. PLoS One 2013; 8:e62144. [PMID: 23634226 PMCID: PMC3636262 DOI: 10.1371/journal.pone.0062144] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/18/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Recent evidence suggests that the IL-33/IL1RL1 axis plays a critical role in several autoimmune and inflammatory disorders; however, its mechanistic role in inflammatory bowel disease (IBD) has not been clearly defined. We investigated the contribution of IL-33 and IL1RL1 polymorphisms to IBD risk, and possible correlations with phenotype in an Italian cohort of adult and pediatric patients. METHODS We evaluated the association of six SNPs in IL-33 and IL1RL1 genes, in 805 Crohn's disease (CD), 816 ulcerative colitis (UC), and 752 controls, using Taqman. IL-33 and IL1RL1 mRNA expression was also analyzed. RESULTS Significant allele and genotype associations with IL-33 rs3939286 were found in CD (P = 0.004; P = 0.035) and UC patients (P = 0.002; P = 0.038). After stratifying the cohort for age at diagnosis, the differences remained significant only in the IBD adult-onset. Significant associations were also obtained in CD patients with two IL1RL1 polymorphisms (rs13015714 and rs2058660, P<0.015). By combining homo- and heterozygous carriers of the rs13015714 risk allele, differences were still significant for both CD adult- and pediatric-onset. Upon genotype-phenotype evaluation, an increased frequency of extensive colitis in adult UC (P = 0.019) and in steroid-responsive pediatric patients (P = 0.024) carrying the IL-33 rs3939286 risk genotype, was observed. mRNA expression of IL-33 and IL1RL1 in inflamed IBD biopsy samples was significantly increased. CONCLUSIONS Common IL-33 and IL1RL1 polymorphisms contribute to the risk of IBD in an Italian cohort of adult and pediatric patients, with some influence on sub-phenotypes.
Collapse
Affiliation(s)
- Anna Latiano
- Division of Gastroenterology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6494
|
Extended haplotype association study in Crohn's disease identifies a novel, Ashkenazi Jewish-specific missense mutation in the NF-κB pathway gene, HEATR3. Genes Immun 2013; 14:310-6. [PMID: 23615072 PMCID: PMC3785105 DOI: 10.1038/gene.2013.19] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/12/2013] [Accepted: 03/21/2013] [Indexed: 12/19/2022]
Abstract
The Ashkenazi Jewish population has a several-fold higher prevalence of Crohn’s disease compared to non-Jewish European ancestry populations and has a unique genetic history. Haplotype association is critical to Crohn’s disease etiology in this population, most notably at NOD2, in which three causal, uncommon, and conditionally independent NOD2 variants reside on a shared background haplotype. We present an analysis of extended haplotypes which showed significantly greater association to Crohn’s disease in the Ashkenazi Jewish population compared to a non-Jewish population (145 haplotypes and no haplotypes with P-value < 10−3, respectively). Two haplotype regions, one each on chromosomes 16 and 21, conferred increased disease risk within established Crohn’s disease loci. We performed exome sequencing of 55 Ashkenazi Jewish individuals and follow-up genotyping focused on variants in these two regions. We observed Ashkenazi Jewish-specific nominal association at R755C in TRPM2 on chromosome 21. Within the chromosome 16 region, R642S of HEATR3 and rs9922362 of BRD7 showed genome-wide significance. Expression studies of HEATR3 demonstrated a positive role in NOD2-mediated NF-κB signaling. The BRD7 signal showed conditional dependence with only the downstream rare Crohn’s disease-causal variants in NOD2, but not with the background haplotype; this elaborates NOD2 as a key illustration of synthetic association.
Collapse
|
6495
|
Peloquin JM, Nguyen DD. The microbiota and inflammatory bowel disease: insights from animal models. Anaerobe 2013; 24:102-6. [PMID: 23603043 DOI: 10.1016/j.anaerobe.2013.04.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 04/05/2013] [Accepted: 04/06/2013] [Indexed: 12/17/2022]
Abstract
Inflammatory bowel disease (IBD) is thought to result from a dysregulated immune response to intestinal microbial flora in individuals with genetic predisposition(s). Genome-wide association studies (GWAS) in human IBD have identified more than 150 associated loci, some of which are key players in innate immunity and bacterial handling, reflecting the importance of the microbiota in disease pathogenesis. In fact, the presence of a microbial flora is not only crucial to the development of a normal murine immune system but also critical for the development of disease in the majority of animal models of IBD. Although animal models do not perfectly recapitulate human IBD, they have led to the discovery of important concepts in IBD pathogenesis, such as the central role of microbiota in disease development and perpetuation. Many genetically susceptible models do not develop colitis when raised in a germ-free or Helicobacter-free environment. In fact, disease in most models can be attenuated or completely abolished with antibiotic treatment. Moreover, an interplay between intestinal microbiota and mucosal immune activation is suggested by the presence of serum antibodies against the Cbir1 flagellin, an immunodominant antigen that activates TLR5, in certain models of spontaneous colitis as well as in human patients. Furthermore, T cells reactive to Cbir1 are able to induce disease in recipient mice upon adoptive cell transfer, demonstrating the pro-inflammatory properties of certain bacterial products. In fact, it has been shown that transfer of certain intestinal bacteria from a specific genetically altered mouse model with spontaneous colitis can induce disease in wild-type mice upon co-housing or direct feeding. These observations demonstrate the pathogenic potential of intestinal microbiota in IBD. However, intestinal bacteria are not always maladaptive in mucosal homeostasis. Both Bacteroides fragilis and Clostridium species promote the number and function of a certain regulatory T cell subset in the colon leading to protection against murine colitis. In fact, normal development of regulatory cells and epithelial cell integrity are abolished in the absence of an intestinal flora, suggestive of the need for certain microbial components to induce beneficial anti-inflammatory mechanisms. All in all, altered immune responses to microbes play a crucial role in IBD pathogenesis. However, certain components of the microbiota are also likely critical for normal development of regulatory mechanisms that contribute to mucosal homeostasis. Findings in animal models highlight the concept that IBD is a disease that results from the interplay of genetics and microbial/environmental factors.
Collapse
Affiliation(s)
- Joanna M Peloquin
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
6496
|
Kaser A, Adolph TE, Blumberg RS. The unfolded protein response and gastrointestinal disease. Semin Immunopathol 2013; 35:307-19. [PMID: 23588234 DOI: 10.1007/s00281-013-0377-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 03/19/2013] [Indexed: 12/22/2022]
Abstract
As the inner lining of the gastrointestinal tract, the intestinal epithelium serves an essential role in innate immune function at the interface between the host and microbiota. Given the unique environmental challenges and thus physiologic secretory functions of this surface, it is exquisitely sensitive to perturbations that affect its capacity to resolve endoplasmic reticulum (ER) stress. Genetic deletion of factors involved in the unfolded protein response (UPR), which functions in the resolution of ER stress that arises from misfolded proteins, result in spontaneous intestinal inflammation closely mimicking human inflammatory bowel disease (IBD). This is demonstrated by observations wherein deletion of genes such as Xbp1 and Agr2 profoundly affects the intestinal epithelium and results in spontaneous intestinal inflammation. Moreover, both genes, along with others (e.g., ORDML3) represent genetic risk factors for human IBD, both Crohn's disease and ulcerative colitis. Here, we review the current mechanistic understanding for how unresolved ER stress can lead to intestinal inflammation and highlight the findings that implicate ER stress as a genetically affected biological pathway in IBD. We further discuss environmental and microbial factors that might impact on the epithelium's capacity to resolve ER stress and which may constitute exogenous factors that may precipitate disease in genetically susceptible individuals.
Collapse
Affiliation(s)
- Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | | | | |
Collapse
|
6497
|
Antonioli L, Colucci R, Pellegrini C, Giustarini G, Tuccori M, Blandizzi C, Fornai M. The role of purinergic pathways in the pathophysiology of gut diseases: pharmacological modulation and potential therapeutic applications. Pharmacol Ther 2013; 139:157-88. [PMID: 23588157 DOI: 10.1016/j.pharmthera.2013.04.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/15/2013] [Indexed: 02/08/2023]
Abstract
Gut homeostasis results from complex neuro-immune interactions aimed at triggering stereotypical and specific programs of coordinated mucosal secretion and powerful motor propulsion. A prominent role in the regulation of this highly integrated network, comprising a variety of immune/inflammatory cells and the enteric nervous system, is played by purinergic mediators. The cells of the digestive tract are literally plunged into a "biological sea" of functionally active nucleotides and nucleosides, which carry out the critical task of driving regulatory interventions on cellular functions through the activation of P1 and P2 receptors. Intensive research efforts are being made to achieve an integrated view of the purinergic system, since it is emerging that the various components of purinergic pathways (i.e., enzymes, transporters, mediators and receptors) are mutually linked entities, deputed to finely modulating the magnitude and the duration of purinergic signaling, and that alterations occurring in this balanced network could be intimately involved in the pathophysiology of several gut disorders. This review article intends to provide a critical appraisal of current knowledge on the purinergic system role in the regulation of gastrointestinal functions, considering these pathways as a whole integrated network, which is capable of finely controlling the levels of bioactive nucleotides and nucleosides in the biophase of their respective receptors. Special attention is paid to the mechanisms through which alterations in the various compartments of the purinergic system could contribute to the pathophysiology of gut disorders, and to the possibility of counteracting such dysfunctions by means of pharmacological interventions on purinergic molecular targets.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Italy.
| | | | | | | | | | | | | |
Collapse
|
6498
|
Yamazaki K, Umeno J, Takahashi A, Hirano A, Johnson TA, Kumasaka N, Morizono T, Hosono N, Kawaguchi T, Takazoe M, Yamada T, Suzuki Y, Tanaka H, Motoya S, Hosokawa M, Arimura Y, Shinomura Y, Matsui T, Matsumoto T, Iida M, Tsunoda T, Nakamura Y, Kamatani N, Kubo M. A genome-wide association study identifies 2 susceptibility Loci for Crohn's disease in a Japanese population. Gastroenterology 2013; 144:781-8. [PMID: 23266558 DOI: 10.1053/j.gastro.2012.12.021] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 12/08/2012] [Accepted: 12/17/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Crohn's disease is an inflammatory bowel disease induced by multiple genetic and environmental factors. Genome-wide association studies have identified genetic factors that affect the risk for Crohn's disease in European populations, but information from other ethnic groups is scarce. We therefore investigated genetic factors associated with Crohn's disease in the Japanese population. METHODS We performed a genome-wide association study with 372 individuals with Crohn's disease (cases) and 3389 controls, all from the Japanese population. To confirm identified associations, we performed a replication study with an independent panel of 1151 Crohn's disease cases and 15,800 controls. We also performed an association analysis using genome-wide genotype imputation in the discovery cohort. RESULTS We confirmed associations of Crohn's disease with variants in MHC (rs7765379, P = 2.11 × 10(-59)), TNFSF15 (rs6478106, P = 3.87 × 10(-45)), and STAT3 (rs9891119, P = 2.24 × 10(-14)). We identified 2 new susceptibility loci: on chromosome 4p14 (rs1487630, P = 2.40 × 10(-11); odds ratio, 1.33), and in the SLC25A15-ELF1-WBP4 region on 13q14 (rs7329174 in ELF1, P = 5.12 × 10(-9); odds ratio, 1.27). CONCLUSIONS In a genome-wide association study, we identified 2 new susceptibility loci for Crohn's disease in a Japanese population. These findings could increase our understanding of the pathogenesis of Crohn's disease.
Collapse
Affiliation(s)
- Keiko Yamazaki
- Laboratory for Genotyping Development, Center for Genomic Medicine, The Institute of Physical and Chemical Research (RIKEN), Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6499
|
Abstract
BACKGROUND Our previous studies have demonstrated that B cells in human inflammatory bowel disease (IBD) are highly activated and produce copious amounts of chemokines. Here, we showed that B cells produce eotaxin-1, a selective chemokine for acute eosinophilia. Increased levels of activated eosinophils have been found in the intestinal mucosa in patients with IBD, but their role(s) and the regulation of their migration patterns remain poorly defined. METHODS To determine how B-cell secretion of eotaxin-1 influences eosinophil activation and migration, we performed immunoepidemiological approaches coupled with in vitro studies. B cells and eosinophils from patients with Crohn's disease and ulcerative colitis were isolated, and responses to Toll-like receptor ligands (TLR) were measured and assessed for the relationship with clinical disease. RESULTS Eotaxin-1 from recirculating B cells, and TLR ligands, regulated eosinophil homing mechanisms in IBD. B cells stimulated with hypo-acylated lipopolysaccharide (LPS) produced copious amounts of eotaxin-1, which influenced eosinophil activation profiles in the bloodstream. We also found that hexa-acylated LPS, such Escherichia coli LPS, directly activated TLR2-expressing and TLR4-expressing eosinophils from patients with IBD to express a different repertoire of mucosal homing receptors, namely CCR9 and CCR10. Whereas B-cell production of eotaxin-1 was correlated with reduced disease activity, eosinophil activation by hexa-acylated LPS was associated with increased disease activity. CONCLUSIONS These results suggest that systemic TLR ligands influence eosinophil migration patterns, both directly and indirectly, through B cells. Our report uncovers unexpected mechanisms of cross talk between certain immune cells that shed new light on IBD immunology.
Collapse
|
6500
|
Abstract
Innate immunity involves direct interactions between the host and microorganisms, both pathogenic and symbiotic, so natural selection is expected to strongly influence genes involved in these processes. Population genetics investigates the impact of past natural selection events on the genome of present-day human populations, and it complements immunological as well as clinical and epidemiological genetic studies. Recent data show that the impact of selection on the different families of innate immune receptors and their downstream signalling molecules varies considerably. This Review discusses these findings and highlights how they help to delineate the relative functional importance of innate immune pathways, which can range from being essential to being redundant.
Collapse
|