1
|
Verstockt S, Hannes L, Jans DS, Deman S, Souche E, van der Werf I, Vandermeulen L, Lobaton T, Laukens D, Verstockt B, Van Houdt J, Hoischen A, Vermeire S, Cleynen I. MIP4IBD: An Easy and Rapid Genotyping-by-Sequencing Assay for the Inflammatory Bowel Diseases Risk Loci. Inflamm Bowel Dis 2025; 31:786-799. [PMID: 39657915 DOI: 10.1093/ibd/izae289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are polygenic, with many genetic variants contributing to disease risk. Knowing the genotype of specific variants or calculating a combined genetic risk score benefits translational and functional research. To address this, we developed MIP4IBD, a flexible and cost-effective genotyping-by-sequencing assay using molecular inversion probes (MIPs). METHODS The assay targets 463 IBD risk variants, and 77 additional relevant variants. Molecular inversion probes capture and library preparation were optimized using 15 IBD DNA samples, comparing genotypes with immunochip. A custom GitHub pipeline was created for data processing, performance testing, and genotype calling. The final design was validated on a larger scale (149 IBD patients, 104 non-IBD controls, and 3 external cell lines), incorporating post hoc quality control criteria. RESULTS The assay achieved a 3.5-day turnaround time at €15 per sample with optimal sample throughput, demonstrating a 92.6% success rate in variant capture and genotype concordance rates of 99.3% and 99.6% with Infinium Global Screening Array24 BeadChip and WGS, respectively. A downstream application involved the calculation of a weighted IBD polygenic risk score (PRS), which was significantly higher in IBD patients than controls (mean 0.42 vs -0.49, P = 1.95E-11). Individuals in the highest PRS quartile had a 15.7-fold (95% CI: 6.5-38.3) risk of developing IBD and an earlier age of onset (26 vs 37 years, P = 0.02), compared to the lowest quartile. CONCLUSIONS MIP4IBD is a validated, scalable genotyping assay targeting IBD risk loci, with an integrated bioinformatics pipeline from sequencing data to genotypes and PRS calculation. Its cost-effectiveness and flexibility for additional variants make it particularly appealing for translational and clinical applications.
Collapse
Affiliation(s)
- Sare Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
| | - Laurens Hannes
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Deborah Sarah Jans
- Department of Human Genetics, Laboratory for Complex Genetics Leuven, University of Leuven, Herestraat 49 Box 604, 3000 Leuven, Belgium
| | - Stephanie Deman
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Genomics Core, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Erika Souche
- Department of Human Genetics, University of Leuven, KU Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
- Genomics Core, University Hospitals Leuven, Herestraat 49 Box 602, 3000 Leuven, Belgium
| | - Ilse van der Werf
- Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Liv Vandermeulen
- Department of Gastroenterology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Triana Lobaton
- Department of Gastroenterology, University Hospital of Ghent, Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Debby Laukens
- Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Van Houdt
- J&J Innovative Medicine, Antwerpseweg 15-17, 2340 Beerse, Belgium
| | - Alexander Hoischen
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Department of Human Genetics, Heyendaalseweg 135 Box 9010, 6525 AJ Nijmegen, The Netherlands
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism (CHROMETA), University of Leuven, Herestraat 49 Box 701, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Isabelle Cleynen
- Department of Human Genetics, Laboratory for Complex Genetics Leuven, University of Leuven, Herestraat 49 Box 604, 3000 Leuven, Belgium
| |
Collapse
|
2
|
Ahadi M, Rouhbakhsh Zahmatkesh MR, Ebrahimi P, AkbariRad M. The Role of Dietary Habits in the Pathogenesis and Development of Inflammatory Bowel Disease: A Narrative Review. Middle East J Dig Dis 2024; 16:5-11. [PMID: 39050099 PMCID: PMC11264837 DOI: 10.34172/mejdd.2024.362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/25/2023] [Indexed: 07/27/2024] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a chronic immune-mediated disease. The incidence of IBD is influenced by various genetic and environmental factors, with dietary habits gaining significant scientific interest. While the role of diet in the pathogenesis and development of IBD is still debated, recent studies have demonstrated its potential impact. However, conflicting findings exist regarding the efficacy of dietary interventions in the treatment and control of IBD. This review aimed to summarize the current understanding of the relationship between diet and IBD, highlighting the different perspectives and reasonings observed in recent studies. Overall, it has been shown that dietary habits play a role in the incidence of IBD, and adopting a controlled dietary approach may help manage the disease. Consequently, diet can be considered a predictive and prognostic factor in IBD.
Collapse
Affiliation(s)
- Mitra Ahadi
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | | | - Parisa Ebrahimi
- Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Mina AkbariRad
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| |
Collapse
|
3
|
Cheng X, Shi J, Jia Z, Ha P, Soo C, Ting K, James AW, Shi B, Zhang X. NELL-1 in Genome-Wide Association Studies across Human Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:395-405. [PMID: 34890556 PMCID: PMC8895422 DOI: 10.1016/j.ajpath.2021.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/08/2023]
Abstract
Neural epidermal growth factor-like (EGFL)-like protein (NELL)-1 is a potent and key osteogenic factor in the development and regeneration of skeletal tissues. Intriguingly, accumulative data from genome-wide association studies (GWASs) have started unveiling potential broader roles of NELL-1 beyond its functions in bone and cartilage. With exploration of the genetic variants of the entire genome in large-scale disease cohorts, GWASs have been used for establishing the connection between specific single-nucleotide polymorphisms of NELL1, in addition to osteoporosis, metabolic diseases, inflammatory conditions, neuropsychiatric diseases, neurodegenerative disorders, and malignant tumors. This review summarizes the findings from GWASs on the manifestation, significance level, implications on function, and correlation of specific NELL1 single-nucleotide polymorphisms in various disorders in humans. By offering a unique and comprehensive correlation between genetic variants and plausible functions of NELL1 in GWASs, this review illustrates the wide range of potential effects of a single gene on the pathogenesis of multiple disorders in humans.
Collapse
Affiliation(s)
- Xu Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and the Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California
| | - Jiayu Shi
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California
| | - Zhonglin Jia
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and the Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pin Ha
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California-Los Angeles, Los Angeles, California
| | - Kang Ting
- Forsyth Institute, affiliate of the Harvard School of Dental Medicine, Boston, Massachusetts
| | - Aaron W James
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bing Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and the Department of Cleft Lip and Palate, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Xinli Zhang
- Section of Orthodontics, Division of Growth and Development, School of Dentistry, University of California-Los Angeles, Los Angeles, California.
| |
Collapse
|
4
|
Watanabe D, Kamada N. Contribution of the Gut Microbiota to Intestinal Fibrosis in Crohn's Disease. Front Med (Lausanne) 2022; 9:826240. [PMID: 35198577 PMCID: PMC8859331 DOI: 10.3389/fmed.2022.826240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
In Crohn's disease (CD), intestinal fibrosis is a critical determinant of a patient's prognosis. Although inflammation may be a prerequisite for the initiation of intestinal fibrosis, research shows that the progression or continuation of intestinal fibrosis can occur independently of inflammation. Thus, once initiated, intestinal fibrosis may persist even if medical treatment controls inflammation. Clearly, an understanding of the pathophysiological mechanisms of intestinal fibrosis is required to diminish its occurrence. Accumulating evidence suggests that the gut microbiota contributes to the pathogenesis of intestinal fibrosis. For example, the presence of antibodies against gut microbes can predict which CD patients will have intestinal complications. In addition, microbial ligands can activate intestinal fibroblasts, thereby inducing the production of extracellular matrix. Moreover, in various animal models, bacterial infection can lead to the development of intestinal fibrosis. In this review, we summarize the current knowledge of the link between intestinal fibrosis in CD and the gut microbiota. We highlight basic science and clinical evidence that the gut microbiota can be causative for intestinal fibrosis in CD and provide valuable information about the animal models used to investigate intestinal fibrosis.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
5
|
Wang X, Ni J, You Y, Feng G, Zhang S, Bao W, Hou H, Li H, Liu L, Zheng M, Wang Y, Zhou H, Shen W, Shen X. SNX10-mediated LPS sensing causes intestinal barrier dysfunction via a caspase-5-dependent signaling cascade. EMBO J 2021; 40:e108080. [PMID: 34747049 DOI: 10.15252/embj.2021108080] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 09/23/2021] [Accepted: 09/30/2021] [Indexed: 01/04/2023] Open
Abstract
Altered intestinal microbial composition promotes intestinal barrier dysfunction and triggers the initiation and recurrence of inflammatory bowel disease (IBD). Current treatments for IBD are focused on control of inflammation rather than on maintaining intestinal epithelial barrier function. Here, we show that the internalization of Gram-negative bacterial outer membrane vesicles (OMVs) in human intestinal epithelial cells promotes recruitment of caspase-5 and PIKfyve to early endosomal membranes via sorting nexin 10 (SNX10), resulting in LPS release from OMVs into the cytosol. Caspase-5 activated by cytosolic LPS leads to Lyn phosphorylation, which in turn promotes nuclear translocalization of Snail/Slug, downregulation of E-cadherin expression, and intestinal barrier dysfunction. SNX10 deletion or treatment with DC-SX029, a novel SNX10 inhibitor, rescues OMV-induced intestinal barrier dysfunction and ameliorates colitis in mice by blocking cytosolic LPS release, caspase-5 activation, and downstream signaling. Our results show that targeting SNX10 may be a new therapeutic approach for restoring intestinal epithelial barrier function and promising strategy for IBD treatment.
Collapse
Affiliation(s)
- Xu Wang
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiahui Ni
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China.,The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, China
| | - Yan You
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Guize Feng
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Sulin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Weilian Bao
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Hui Hou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haidong Li
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Lixin Liu
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yirui Wang
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Weixing Shen
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, China
| | - Xiaoyan Shen
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
6
|
The Candidate IBD Risk Gene CCNY Is Dispensable for Intestinal Epithelial Homeostasis. Cells 2021; 10:cells10092330. [PMID: 34571979 PMCID: PMC8471355 DOI: 10.3390/cells10092330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
The CCNY gene, which encodes cyclin Y, has been implicated in the pathogenesis of inflammatory bowel disease (IBD). Cyclin Y promotes Wnt/β-catenin signaling and autophagy, which are critical for intestinal epithelial cell (IEC) homeostasis, and may thereby contribute to wound repair in colitis. However, whether cyclin Y has an essential function in IECs is unknown. We, therefore, investigated the epithelial injury response and mucosal regeneration in mice with conditional knock-out of Ccny in the intestinal epithelium. We observed that Ccny-deficient mice did not exhibit any differences in cell proliferation and disease activity compared to wild-type littermates in the dextran sulfate sodium (DSS) colitis model. Complementary in vitro experiments showed that loss of CCNY in model IECs did not affect Wnt signaling, cell proliferation, or autophagy. Additionally, we observed that expression of the cyclin-Y-associated cyclin-dependent kinase (CDK) 14 is exceedingly low specifically in IEC. Collectively, these results suggest that cyclin Y does not contribute to intestinal epithelial homeostasis, possibly due to low levels of specific CDKs in these cells. Thus, it is unlikely that CCNY mutations are causatively involved in IBD pathogenesis.
Collapse
|
7
|
Germic N, Hosseini A, Yousefi S, Karaulov A, Simon HU. Regulation of eosinophil functions by autophagy. Semin Immunopathol 2021; 43:347-362. [PMID: 34019141 PMCID: PMC8241657 DOI: 10.1007/s00281-021-00860-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022]
Abstract
Eosinophils are granule-containing leukocytes which develop in the bone marrow. For many years, eosinophils have been recognized as cytotoxic effector cells, but recent studies suggest that they perform additional immunomodulatory and homeostatic functions. Autophagy is a conserved intracellular process which preserves cellular homeostasis. Autophagy defects have been linked to the pathogenesis of many human disorders. Evidence for abnormal regulation of autophagy, including decreased or increased expression of autophagy-related (ATG) proteins, has been reported in several eosinophilic inflammatory disorders, such as Crohn's disease, bronchial asthma, eosinophilic esophagitis, and chronic rhinosinusitis. Despite the increasing extent of research using preclinical models of immune cell-specific autophagy deficiency, the physiological relevance of autophagic pathway in eosinophils has remained unknown until recently. Owing to the increasing evidence that eosinophils play a role in keeping organismal homeostasis, the regulation of eosinophil functions is of considerable interest. Here, we discuss the most recent advances on the role of autophagy in eosinophils, placing particular emphasis on insights obtained in mouse models of infections and malignant diseases in which autophagy has genetically dismantled in the eosinophil lineage. These studies pointed to the possibility that autophagy-deficient eosinophils exaggerate inflammation. Therefore, the pharmacological modulation of the autophagic pathway in these cells could be used for therapeutic interventions.
Collapse
Affiliation(s)
- Nina Germic
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland
| | - Aref Hosseini
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Sechenov University, 119991, Moscow, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland. .,Department of Clinical Immunology and Allergology, Sechenov University, 119991, Moscow, Russia. .,Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420012, Kazan, Russia.
| |
Collapse
|
8
|
Wang J, Liu H, Wang Y, Wu J, Wang C, Liu K, Qin Q. The Polymorphisms of Interleukin-12B Gene and Susceptibility to Inflammatory Bowel Diseases: A Meta-analysis and Trial Sequential Analysis. Immunol Invest 2021; 50:987-1006. [PMID: 33390082 DOI: 10.1080/08820139.2020.1863981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objective: Inflammatory bowel disease (IBD) is a heterogeneous complex disease referring to two chronic disorders: Crohn's disease (CD) and ulcerative colitis (UC). To clarify the relationship between IL-12B gene polymorphisms and susceptibility to CD and UC, a meta-analysis was conducted.Methods: A comprehensive search of the PubMed, Web of Science, Embase and Cochrane databases was conducted up to Oct 2019. Studies evaluating the relationship between risk of IBD and variants of IL-12B (rs6887695, rs3212227 and rs10045431) were included. Odds ratio (OR) and 95% confidence interval (CI) were calculated. Trial sequential analysis (TSA) was implemented to estimate the required information size (RIS) and evaluate the credibility of the meta-analysis results.Results: Seventeen studies containing 9827 patients with CD, 7583 patients with UC and 16044 controls were included. The results showed significant association between rs6887695 polymorphism and susceptibility to CD (allele model: OR = 1.17, 95% CI: 1.12-1.22) and UC (allele model: OR = 1.16, 95% CI: 1.09-1.23), and "C" allele carriers had a higher risk, with TSA conclusive. For rs10045431, no significant association with CD susceptibility was identified, while a significantly increased risk in UC was found (allele mode: OR = 1.16, 95% CI: 1.07-1.25), both results were conclusive according to TSA. No significant association between rs3212227 and CD or UC susceptibility was found, and TSA research warranted further investigation to certify the results. No significant heterogeneity was found.Conclusion: IL-12B rs6887695 polymorphism was associated with increased risk of CD and UC, while IL-12B rs10045431 polymorphism might only be correlated with the risk of UC.Abbreviations: IBD: inflammatory bowel disease; CD: Crohn's disease; UC: ulcerative colitis; IL-12B: interleukin-12B; OR: odds ratio; CI: confidence interval; TSA: trial sequential analysis; RIS: required information size; DCs: dendritic cells; NK: nature killer; APCs: antigen-presenting cells; TNF: tumor necrosis factor; SNP: single nucleotide polymorphisms; HWE: Hardy-Weinberg equilibrium; NOS: Newcastle-Ottawa scale; RRR: relative risk reduction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hong Liu
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yadan Wang
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jing Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Canghai Wang
- Department of Gastroenterology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Kuiliang Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qin Qin
- Department of Prevention and Health Care, Zhongshan Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
9
|
Castro-Sanchez P, Teagle AR, Prade S, Zamoyska R. Modulation of TCR Signaling by Tyrosine Phosphatases: From Autoimmunity to Immunotherapy. Front Cell Dev Biol 2020; 8:608747. [PMID: 33425916 PMCID: PMC7793860 DOI: 10.3389/fcell.2020.608747] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Early TCR signaling is dependent on rapid phosphorylation and dephosphorylation of multiple signaling and adaptor proteins, leading to T cell activation. This process is tightly regulated by an intricate web of interactions between kinases and phosphatases. A number of tyrosine phosphatases have been shown to modulate T cell responses and thus alter T cell fate by negatively regulating early TCR signaling. Mutations in some of these enzymes are associated with enhanced predisposition to autoimmunity in humans, and mouse models deficient in orthologous genes often show T cell hyper-activation. Therefore, phosphatases are emerging as potential targets in situations where it is desirable to enhance T cell responses, such as immune responses to tumors. In this review, we summarize the current knowledge about tyrosine phosphatases that regulate early TCR signaling and discuss their involvement in autoimmunity and their potential as targets for tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Rose Zamoyska
- Ashworth Laboratories, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Finethy R, Dockterman J, Kutsch M, Orench‐Rivera N, Wallace GD, Piro AS, Luoma S, Haldar AK, Hwang S, Martinez J, Kuehn MJ, Taylor GA, Coers J. Dynamin-related Irgm proteins modulate LPS-induced caspase-11 activation and septic shock. EMBO Rep 2020; 21:e50830. [PMID: 33124745 PMCID: PMC7645254 DOI: 10.15252/embr.202050830] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/08/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammation associated with gram-negative bacterial infections is often instigated by the bacterial cell wall component lipopolysaccharide (LPS). LPS-induced inflammation and resulting life-threatening sepsis are mediated by the two distinct LPS receptors TLR4 and caspase-11 (caspase-4/-5 in humans). Whereas the regulation of TLR4 activation by extracellular and phago-endosomal LPS has been studied in great detail, auxiliary host factors that specifically modulate recognition of cytosolic LPS by caspase-11 are largely unknown. This study identifies autophagy-related and dynamin-related membrane remodeling proteins belonging to the family of Immunity-related GTPases M clade (IRGM) as negative regulators of caspase-11 activation in macrophages. Phagocytes lacking expression of mouse isoform Irgm2 aberrantly activate caspase-11-dependent inflammatory responses when exposed to extracellular LPS, bacterial outer membrane vesicles, or gram-negative bacteria. Consequently, Irgm2-deficient mice display increased susceptibility to caspase-11-mediated septic shock in vivo. This Irgm2 phenotype is partly reversed by the simultaneous genetic deletion of the two additional Irgm paralogs Irgm1 and Irgm3, indicating that dysregulated Irgm isoform expression disrupts intracellular LPS processing pathways that limit LPS availability for caspase-11 activation.
Collapse
Affiliation(s)
- Ryan Finethy
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
| | - Jacob Dockterman
- Department of ImmunologyDuke University Medical CenterDurhamNCUSA
| | - Miriam Kutsch
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
| | | | - Graham D Wallace
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
| | - Anthony S Piro
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
| | - Sarah Luoma
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
| | - Arun K Haldar
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
- Present address:
Division of BiochemistryCentral Drug Research Institute (CDRI)Council of Scientific and Industrial Research (CSIR)LucknowIndia
| | - Seungmin Hwang
- Department of PathologyThe University of ChicagoChicagoILUSA
- Present address:
VIR BiotechnologySan FranciscoCAUSA
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease LaboratoryNational Institute of Environmental Health SciencesNational Institutes of HealthResearch Triangle ParkNCUSA
| | - Meta J Kuehn
- Department of BiochemistryDuke University Medical CenterDurhamNCUSA
| | - Gregory A Taylor
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
- Department of ImmunologyDuke University Medical CenterDurhamNCUSA
- Division of GeriatricsDepartment of MedicineCenter for the Study of Aging and Human DevelopmentDuke University Medical CenterDurhamNCUSA
- Geriatric Research, Education, and Clinical Center, VA Medical CenterDurhamNCUSA
| | - Jörn Coers
- Department of Molecular Genetics and MicrobiologyDuke University Medical CenterDurhamNCUSA
- Department of ImmunologyDuke University Medical CenterDurhamNCUSA
| |
Collapse
|
11
|
Dohmen M, Krieg S, Agalaridis G, Zhu X, Shehata SN, Pfeiffenberger E, Amelang J, Bütepage M, Buerova E, Pfaff CM, Chanda D, Geley S, Preisinger C, Sakamoto K, Lüscher B, Neumann D, Vervoorts J. AMPK-dependent activation of the Cyclin Y/CDK16 complex controls autophagy. Nat Commun 2020; 11:1032. [PMID: 32098961 PMCID: PMC7042329 DOI: 10.1038/s41467-020-14812-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
The AMP-activated protein kinase (AMPK) is a master sensor of the cellular energy status that is crucial for the adaptive response to limited energy availability. AMPK is implicated in the regulation of many cellular processes, including autophagy. However, the precise mechanisms by which AMPK controls these processes and the identities of relevant substrates are not fully understood. Using protein microarrays, we identify Cyclin Y as an AMPK substrate that is phosphorylated at Serine 326 (S326) both in vitro and in cells. Phosphorylation of Cyclin Y at S326 promotes its interaction with the Cyclin-dependent kinase 16 (CDK16), thereby stimulating its catalytic activity. When expressed in cells, Cyclin Y/CDK16 is sufficient to promote autophagy. Moreover, Cyclin Y/CDK16 is necessary for efficient AMPK-dependent activation of autophagy. This functional interaction is mediated by AMPK phosphorylating S326 of Cyclin Y. Collectively, we define Cyclin Y/CDK16 as downstream effector of AMPK for inducing autophagy.
Collapse
Affiliation(s)
- Marc Dohmen
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
- Center for Translational & Clinical Research Aachen (CTC-A), Medical School, RWTH Aachen University, 52074, Aachen, Germany
| | - Sarah Krieg
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
| | - Georgios Agalaridis
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
- Miltenyi Biotec GmbH, Friedrich-Ebert-Straße 68, 51429, Bergisch Gladbach, Germany
| | - Xiaoqing Zhu
- CARIM School for Cardiovascular Diseases, Maastricht University, P.O. box 616, 6200 MD, Maastricht, The Netherlands
| | | | - Elisabeth Pfeiffenberger
- Division of Molecular Pathophysiology, Biocenter, Innsbruck Medical University, Innrain 80/82, 6020, Innsbruck, Austria
| | - Jan Amelang
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
| | - Mareike Bütepage
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
| | - Elena Buerova
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
| | - Carolina M Pfaff
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany
- AstraZeneca GmbH, Tinsdaler Weg 183, 22880, Wedel, Germany
| | - Dipanjan Chanda
- CARIM School for Cardiovascular Diseases, Maastricht University, P.O. box 616, 6200 MD, Maastricht, The Netherlands
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Innsbruck Medical University, Innrain 80/82, 6020, Innsbruck, Austria
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Center for Clinical Research (IZKF) Aachen, Medical School, RWTH Aachen University, 52074, Aachen, Germany
| | - Kei Sakamoto
- Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany.
| | - Dietbert Neumann
- CARIM School for Cardiovascular Diseases, Maastricht University, P.O. box 616, 6200 MD, Maastricht, The Netherlands.
- Department of Pathology, University Medical Center Maastricht, 6229 HX, Maastricht, The Netherlands.
| | - Jörg Vervoorts
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
12
|
García-Cano J, Martinez-Martinez A, Sala-Gaston J, Pedrazza L, Rosa JL. HERCing: Structural and Functional Relevance of the Large HERC Ubiquitin Ligases. Front Physiol 2019; 10:1014. [PMID: 31447701 PMCID: PMC6692442 DOI: 10.3389/fphys.2019.01014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 07/23/2019] [Indexed: 12/12/2022] Open
Abstract
Homologous to the E6AP carboxyl terminus (HECT) and regulator of chromosome condensation 1 (RCC1)-like domain-containing proteins (HERCs) belong to the superfamily of ubiquitin ligases. HERC proteins are divided into two subfamilies, Large and Small HERCs. Despite their similarities in terms of both structure and domains, these subfamilies are evolutionarily very distant and result from a convergence phenomenon rather than from a common origin. Large HERC genes, HERC1 and HERC2, are present in most metazoan taxa. They encode very large proteins (approximately 5,000 amino acid residues in a single polypeptide chain) that contain more than one RCC1-like domain as a structural characteristic. Accumulating evidences show that these unusually large proteins play key roles in a wide range of cellular functions which include neurodevelopment, DNA damage repair, and cell proliferation. To better understand the origin, evolution, and function of the Large HERC family, this minireview provides with an integrated overview of their structure and function and details their physiological implications. This study also highlights and discusses how dysregulation of these proteins is associated with severe human diseases such as neurological disorders and cancer.
Collapse
Affiliation(s)
- Jesús García-Cano
- Ubiquitylation and Cell Signalling Lab, IDIBELL, Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| | - Arturo Martinez-Martinez
- Ubiquitylation and Cell Signalling Lab, IDIBELL, Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| | - Joan Sala-Gaston
- Ubiquitylation and Cell Signalling Lab, IDIBELL, Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| | - Leonardo Pedrazza
- Ubiquitylation and Cell Signalling Lab, IDIBELL, Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| | - Jose Luis Rosa
- Ubiquitylation and Cell Signalling Lab, IDIBELL, Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Castro-Sánchez P, Aguilar-Sopeña O, Alegre-Gómez S, Ramirez-Munoz R, Roda-Navarro P. Regulation of CD4 + T Cell Signaling and Immunological Synapse by Protein Tyrosine Phosphatases: Molecular Mechanisms in Autoimmunity. Front Immunol 2019; 10:1447. [PMID: 31297117 PMCID: PMC6607956 DOI: 10.3389/fimmu.2019.01447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
T cell activation and effector function is mediated by the formation of a long-lasting interaction established between T cells and antigen-presenting cells (APCs) called immunological synapse (IS). During T cell activation, different signaling molecules as well as the cytoskeleton and the endosomal compartment are polarized to the IS. This molecular dynamics is tightly regulated by phosphorylation networks, which are controlled by protein tyrosine phosphatases (PTPs). While some PTPs are known to be important regulators of adhesion, ligand discrimination or the stimulation threshold, there is still little information about the regulatory role of PTPs in cytoskeleton rearrangements and endosomal compartment dynamics. Besides, spatial and temporal regulation of PTPs and substrates at the IS is only barely known. Consistent with an important role of PTPs in T cell activation, multiple mutations as well as altered expression levels or dynamic behaviors have been associated with autoimmune diseases. However, the precise mechanism for the regulation of T cell activation and effector function by PTPs in health and autoimmunity is not fully understood. Herein, we review the current knowledge about the regulatory role of PTPs in CD4+ T cell activation, IS assembly and effector function. The potential molecular mechanisms mediating the action of these enzymes in autoimmune disorders are discussed.
Collapse
Affiliation(s)
- Patricia Castro-Sánchez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Oscar Aguilar-Sopeña
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Sergio Alegre-Gómez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Rocio Ramirez-Munoz
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain.,Health Research Institute '12 de Octubre (imas12)', Madrid, Spain
| |
Collapse
|
14
|
Lee HS, Cleynen I. Molecular Profiling of Inflammatory Bowel Disease: Is It Ready for Use in Clinical Decision-Making? Cells 2019; 8:E535. [PMID: 31167397 PMCID: PMC6627070 DOI: 10.3390/cells8060535] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a heterogeneous disorder in terms of age at onset, clinical phenotypes, severity, disease course, and response to therapy. This underlines the need for predictive and precision medicine that can optimize diagnosis and disease management, provide more cost-effective strategies, and minimize the risk of adverse events. Ideally, we can leverage molecular profiling to predict the risk to develop IBD and disease progression. Despite substantial successes of genome-wide association studies in the identification of genetic variants affecting IBD susceptibility, molecular profiling of disease onset and progression as well as of treatment responses has lagged behind. Still, thanks to technological advances and good study designs, predicting phenotypes using genomics and transcriptomics in IBD has been rapidly evolving. In this review, we summarize the current status of prediction of disease risk, clinical course, and response to therapy based on clinical case presentations. We also discuss the potential and limitations of the currently used approaches.
Collapse
Affiliation(s)
- Ho-Su Lee
- Laboratory of Complex Genetics, Department of Human Genetics, KU Leuven, Herestraat 49 - box 610, 3000 Leuven, Belgium.
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea.
| | - Isabelle Cleynen
- Laboratory of Complex Genetics, Department of Human Genetics, KU Leuven, Herestraat 49 - box 610, 3000 Leuven, Belgium.
| |
Collapse
|
15
|
Heinrichs SKM, Hess T, Becker J, Hamann L, Vashist YK, Butterbach K, Schmidt T, Alakus H, Krasniuk I, Höblinger A, Lingohr P, Ludwig M, Hagel AF, Schildberg CW, Veits L, Gyvyte U, Weise K, Schüller V, Böhmer AC, Schröder J, Gehlen J, Kreuser N, Hofer S, Lang H, Lordick F, Malfertheiner P, Moehler M, Pech O, Vassos N, Rodermann E, Izbicki JR, Kruschewski M, Ott K, Schumann RR, Vieth M, Mangold E, Gasenko E, Kupcinskas L, Brenner H, Grimminger P, Bujanda L, Sopeña F, Espinel J, Thomson C, Pérez‐Aísa Á, Campo R, Geijo F, Collette D, Bruns C, Messerle K, Gockel I, Nöthen MM, Lippert H, Ridwelski K, Lanas A, Keller G, Knapp M, Leja M, Kupcinskas J, García‐González MA, Venerito M, Schumacher J. Evidence for PTGER4, PSCA, and MBOAT7 as risk genes for gastric cancer on the genome and transcriptome level. Cancer Med 2018; 7:5057-5065. [PMID: 30191681 PMCID: PMC6198243 DOI: 10.1002/cam4.1719] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022] Open
Abstract
Genetic associations between variants on chromosome 5p13 and 8q24 and gastric cancer (GC) have been previously reported in the Asian population. We aimed to replicate these findings and to characterize the associations at the genome and transcriptome level. We performed a fine-mapping association study in 1926 GC patients and 2012 controls of European descent using high dense SNP marker sets on both chromosomal regions. Next, we performed expression quantitative trait locus (eQTL) analyses using gastric transcriptome data from 143 individuals focusing on the GC associated variants. On chromosome 5p13 the strongest association was observed at rs6872282 (P = 2.53 × 10-04 ) and on chromosome 8q24 at rs2585176 (P = 1.09 × 10-09 ). On chromosome 5p13 we found cis-eQTL effects with an upregulation of PTGER4 expression in GC risk allele carrier (P = 9.27 × 10-11 ). On chromosome 8q24 we observed cis-eQTL effects with an upregulation of PSCA expression in GC risk allele carrier (P = 2.17 × 10-47 ). In addition, we found trans-eQTL effects for the same variants on 8q24 with a downregulation of MBOAT7 expression in GC risk allele carrier (P = 3.11 × 10-09 ). In summary, we confirmed and refined the previously reported GC associations at both chromosomal regions. Our data point to shared etiological factors between Asians and Europeans. Furthermore, our data imply an upregulated expression of PTGER4 and PSCA as well as a downregulated expression of MBOAT7 in gastric tissue as risk-conferring GC pathomechanisms.
Collapse
Affiliation(s)
- Sophie K. M. Heinrichs
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Timo Hess
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Jessica Becker
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Lutz Hamann
- Institute for Microbiology and HygieneCharité University Medical Center BerlinBerlinGermany
| | - Yogesh K. Vashist
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Katja Butterbach
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Hakan Alakus
- Department of General, Visceral and Cancer SurgeryUniversity of CologneCologneGermany
| | - Iurii Krasniuk
- Department of General and Visceral SurgeryMunicipal Hospital SolingenSolingenGermany
| | - Aksana Höblinger
- Department of Internal Medicine ICommunity Hospital MittelrheinKoblenzGermany
| | - Philipp Lingohr
- Department of General, Visceral, Thoracic and Vascular SurgeryUniversity of BonnBonnGermany
| | - Monika Ludwig
- Association for Oncological Studies (Gefos)DortmundGermany
| | - Alexander F. Hagel
- Department of Medicine I, Gastroenterology and Interventional EndoscopyUniversity of ErlangenErlangenGermany
| | | | - Lothar Veits
- Institute of PathologyKlinikum BayreuthBayreuthGermany
| | - Ugne Gyvyte
- Department of Gastroenterology and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Katharina Weise
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Vitalia Schüller
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
- Institute for Medical Biometry, Informatics and EpidemiologyUniversity of BonnBonnGermany
| | - Anne C. Böhmer
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Julia Schröder
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Jan Gehlen
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Nicole Kreuser
- Department of Visceral, Transplant, Thoracic and Vascular SurgeryUniversity Hospital of LeipzigLeipzigGermany
| | - Sebastian Hofer
- Department of General, Visceral and Transplant SurgeryUniversity Medical CenterUniversity of MainzMainzGermany
| | - Hauke Lang
- Department of General, Visceral and Transplant SurgeryUniversity Medical CenterUniversity of MainzMainzGermany
| | - Florian Lordick
- University Cancer Center Leipzig (UCCL)University Hospital of LeipzigLeipzigGermany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious DiseasesOtto‐von‐Guericke University HospitalMagdeburgGermany
| | - Markus Moehler
- First Medical Clinic and PoliclinicUniversity Medical CenterUniversity of MainzMainzGermany
| | - Oliver Pech
- Department of Gastroenterology and Interventional EndoscopySt. John of God HospitalRegensburgGermany
| | | | - Ernst Rodermann
- Association of Medical Practices in Hematology and Internal OncologyTroisdorfGermany
| | - Jakob R. Izbicki
- Department of General, Visceral and Thoracic SurgeryUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Martin Kruschewski
- Department of General and Visceral SurgeryHospital Frankfurt OderFrankfurt OderGermany
| | - Katja Ott
- Department of General, Visceral and Thorax SurgeryRoMed Hospital RosenheimRosenheimGermany
| | - Ralf R. Schumann
- Institute for Microbiology and HygieneCharité University Medical Center BerlinBerlinGermany
| | - Michael Vieth
- Institute of PathologyKlinikum BayreuthBayreuthGermany
| | - Elisabeth Mangold
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Evita Gasenko
- Institute of Clinical and Preventive MedicineUniversity of LatviaRigaLatvia
- Riga East University HospitalRigaLatvia
| | - Limas Kupcinskas
- Department of Gastroenterology and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging ResearchGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Division of Preventive OncologyGerman Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT)HeidelbergGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Grimminger
- Department of General, Visceral and Transplant SurgeryUniversity Medical CenterUniversity of MainzMainzGermany
| | - Luis Bujanda
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd)MadridSpain
- Department of GastroenterologyHospital Donostia/Instituto BiodonostiaUniversidad del País Vasco (UPV/EHU)San SebastiánSpain
| | - Federico Sopeña
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd)MadridSpain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain
- Department of GastroenterologyHospital Clínico Universitario Lozano BlesaZaragozaSpain
| | - Jesús Espinel
- Department of GastroenterologyComplejo HospitalarioLeónSpain
| | - Concha Thomson
- Department of GastroenterologyHospital Obispo PolancoTeruelSpain
| | | | - Rafael Campo
- Department of GastroenterologyHospital Parc TauliSabadellSpain
| | - Fernando Geijo
- Department of GastroenterologyHospital Clínico UniversitarioSalamancaSpain
| | - Daniela Collette
- Association of Medical Practices in Hematology and OncologyDortmundGermany
| | - Christiane Bruns
- Department of General, Visceral and Cancer SurgeryUniversity of CologneCologneGermany
| | - Katharina Messerle
- Department of General, Visceral and Cancer SurgeryUniversity of CologneCologneGermany
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular SurgeryUniversity Hospital of LeipzigLeipzigGermany
| | - Markus M. Nöthen
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
| | - Hans Lippert
- An‐Institute for Quality Control in SurgeryOtto‐von‐Guericke University HospitalMagdeburgGermany
| | - Karsten Ridwelski
- An‐Institute for Quality Control in SurgeryOtto‐von‐Guericke University HospitalMagdeburgGermany
- Department of SurgeryHospital MagdeburgMagdeburgGermany
| | - Angel Lanas
- CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd)MadridSpain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain
- Department of GastroenterologyHospital Clínico Universitario Lozano BlesaZaragozaSpain
| | - Gisela Keller
- Institute of PathologyTechnical University of MunichMunichGermany
| | - Michael Knapp
- Institute for Medical Biometry, Informatics and EpidemiologyUniversity of BonnBonnGermany
| | - Marcis Leja
- Institute of Clinical and Preventive MedicineUniversity of LatviaRigaLatvia
- Riga East University HospitalRigaLatvia
| | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive ResearchLithuanian University of Health SciencesKaunasLithuania
| | - Maria A. García‐González
- Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain
- Department of GastroenterologyHospital Clínico Universitario Lozano BlesaZaragozaSpain
- Instituto Aragonés de Ciencias de la Salud (IACS)ZaragozaSpain
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious DiseasesOtto‐von‐Guericke University HospitalMagdeburgGermany
| | - Johannes Schumacher
- Institute of Human GeneticsUniversity of BonnBonnGermany
- Department of Genomics, Life & Brain CenterUniversity of BonnBonnGermany
- Center of Human Genetics, University Hospital MarburgMarburgGermany
| |
Collapse
|
16
|
Bosca-Watts MM, Minguez M, Planelles D, Navarro S, Rodriguez A, Santiago J, Tosca J, Mora F. HLA-DQ: Celiac disease vs inflammatory bowel disease. World J Gastroenterol 2018; 24:96-103. [PMID: 29358886 PMCID: PMC5757130 DOI: 10.3748/wjg.v24.i1.96] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the genetic predisposition to celiac disease (CeD) in inflammatory bowel disease (IBD) patients by quantifying the frequency of CeD-related human leucocyte antigen (HLA) (HLA-CeD: HLA-DQ2 and -DQ8) in IBD patients globally, by type of IBD and gender, and by calculating the protective/risk contribution of these haplotypes in the development of the IBD disease.
METHODS We conducted a prospective study with IBD patients from our Unit. Clinical information was gathered and blood was tested for HLA-CeD. The control group was made up of unrelated Valencian organ donors.
RESULTS 1034 subjects were analyzed: 457 IBD [207 ulcerative coliti (UC) and 250 Crohn’s disease (CD)] patients and 577 healthy controls. 39% of the controls and 34% of the patients had HLA-CeD (P = 0.0852). HLA-DQ2 was less frequent in UC patients (P = 0.0287), and HLA-DQ8 in CD (P = 0.0217). In women with UC, the frequency of DQ2.5cis (DQB1*02:01-DQA1*05:01) was reduced ≥ 50% [P = 0.0344; preventive fraction (PF) = 13%]. PFs (7%-14%) were obtained with all HLA-CeD haplotypes. HLA DQB1*02:02-DQA1*02:01 (HLA-DQ2.2) was more frequent in CD patients with respect to controls (P = 0.001) and UC patients (etiological fraction = 15%).
CONCLUSION HLA-CeD is not more frequent in IBD patients, with an even lower frequency of HLA-DQ2 and -DQ8 in UC and CD respectively. HLA-DQ2.5 confers protection from the development of UC, especially in women, and HLA-DQ8 does so for the appearance of CD. HLA-DQ2.2 is present in 34% of the CD patients and may constitute a genetic risk factor for CD development.
Collapse
Affiliation(s)
- Marta Maia Bosca-Watts
- IBD Unit, Digestive Disease Department of the University Clinic Hospital of Valencia, University of Valencia, Valencia 46017, Spain
| | - Miguel Minguez
- IBD Unit, Digestive Disease Department of the University Clinic Hospital of Valencia, University of Valencia, Valencia 46017, Spain
| | - Dolores Planelles
- Histocompatibility Department of the Transfusion Center of the Valencian Community, Valencia 46014, Spain
| | - Samuel Navarro
- Pathology Department of the University Clinic Hospital of Valencia, University of Valencia, Valencia 46017, Spain
| | - Alejandro Rodriguez
- Digestive Disease Department of the Hospital Virgen del Castillo of Yecla, Yecla 30510, Spain
| | - Jesus Santiago
- Digestive Disease Department of the Hospital de Manises, Valencia 46940, Spain
| | - Joan Tosca
- IBD Unit, Digestive Disease Department of the University Clinic Hospital of Valencia, University of Valencia, Valencia 46017, Spain
| | - Francisco Mora
- Digestive Disease Department of the University Clinic Hospital of Valencia, University of Valencia, Valencia 46017, Spain
| |
Collapse
|
17
|
Hu J, Zhang W, Li X, Pan D, Li Q. Efficient estimation of disease odds ratios for follow-up genetic association studies. Stat Methods Med Res 2017; 28:1927-1941. [PMID: 29157118 DOI: 10.1177/0962280217741771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the past decade, genome-wide association studies have identified thousands of susceptible variants associated with complex human diseases and traits. Conducting follow-up genetic association studies has become a standard approach to validate the findings of genome-wide association studies. One problem of high interest in genetic association studies is to accurately estimate the strength of the association, which is often quantified by odds ratios in case-control studies. However, estimating the association directly by follow-up studies is inefficient since this approach ignores information from the genome-wide association studies. In this article, an estimator called GFcom, which integrates information from genome-wide association studies and follow-up studies, is proposed. The estimator includes both the point estimate and corresponding confidence interval. GFcom is more efficient than competing estimators regarding MSE and the length of confidence intervals. The superiority of GFcom is particularly evident when the genome-wide association study suffers from severe selection bias. Comprehensive simulation studies and applications to three real follow-up studies demonstrate the performance of the proposed estimator. An R package, "GFcom", implementing our method is publicly available at https://github.com/JiyuanHu/GFcom .
Collapse
Affiliation(s)
- Jiyuan Hu
- 1 Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, PR China.,2 Department of Population Health, New York University, New York, NY, USA
| | - Wei Zhang
- 3 Biostatistics and Bioinformatics Branch, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Xinmin Li
- 4 School of Mathematics and Statistics, Qingdao University, Qingdao, PR China
| | - Dongdong Pan
- 5 Yunnan Key Laboratory of Statistical Modeling and Data Analysis, Yunnan University, Kunming, PR China
| | - Qizhai Li
- 6 LSC, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
18
|
García-Heredia JM, Carnero A. The cargo protein MAP17 (PDZK1IP1) regulates the immune microenvironment. Oncotarget 2017; 8:98580-98597. [PMID: 29228712 PMCID: PMC5716752 DOI: 10.18632/oncotarget.21651] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/25/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a complex defensive response activated after various harmful stimuli allowing the clearance of damaged cells and initiating healing and regenerative processes. Chronic, or pathological, inflammation is also one of the causes of neoplastic transformation and cancer development. MAP17 is a cargo protein that transports membrane proteins from the endoplasmic reticulum. Therefore, its overexpression may be linked to an excess of membrane proteins that may be recognized as an unwanted signal, triggering local inflammation. Therefore, we analyzed whether its overexpression is related to an inflammatory phenotype. In this work, we found a correlation between MAP17 expression and inflammatory phenotype in tumors and in other inflammatory diseases such as Crohn's disease, Barrett's esophagus, COPD or psoriasis. MAP17 expression correlated also with the markers of inflammation HLAs, BBS10, HERC2, ADNP and PYCARD. Furthermore, we found that MAP17 expression directly regulates NFAT2 and IL-6 activation, inducing the differentiation of monocytes to dendritic cells and suggesting a causal role of MAP17 in inflammation. Immunohistochemistry confirms local inflammation, mainly CD45+ cells, at the site of expression of MAP17, at least in tumors, Crohn's and psoriasis. Therefore, our data indicates that the overexpression of the protein MAP17 plays important role in diseases involving chronic inflammation.
Collapse
Affiliation(s)
- José M García-Heredia
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Seville, Spain.,Department of Vegetal Biochemistry and Molecular Biology, University of Seville, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS/Hospital Universitario Virgen del Rocío/Universidad de Sevilla/Consejo Superior de Investigaciones Científicas, Seville, Spain.,CIBER de Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Wildenberg ME, Koelink PJ, Diederen K, Te Velde AA, Wolfkamp SCS, Nuij VJ, Peppelenbosch MP, Nobis M, Sansom OJ, Anderson KI, van der Woude CJ, D'Haens GRAM, van den Brink GR. The ATG16L1 risk allele associated with Crohn's disease results in a Rac1-dependent defect in dendritic cell migration that is corrected by thiopurines. Mucosal Immunol 2017; 10:352-360. [PMID: 27435106 DOI: 10.1038/mi.2016.65] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/18/2016] [Indexed: 02/04/2023]
Abstract
Thiopurines are commonly used drugs in the therapy of Crohn's disease, but unfortunately only show a 30% response rate. The biological basis for the thiopurine response is unclear, thus hampering patient selection prior to treatment. A genetic risk factor associated specifically with Crohn's disease is a variant in ATG16L1 that reduces autophagy. We have previously shown that autophagy is involved in dendritic cell (DC)-T-cell interactions and cytoskeletal regulation. Here we further investigated the role of autophagy in DC cytoskeletal modulation and cellular trafficking. Autophagy-deficient DC displayed loss of filopodia, altered podosome distribution, and increased membrane ruffling, all consistent with increased cellular adhesion. Consequently, autophagy-deficient DC showed reduced migration. The cytoskeletal aberrations were mediated through hyperactivation of Rac1, a known thiopurine target. Indeed thiopurines restored the migratory defects in autophagy-deficient DC. Clinically, the ATG16L1 risk variant associated with increased response to thiopurine treatment in patients with Crohn's disease but not ulcerative colitis. These results suggest that the association between ATG16L1 and Crohn's disease is mediated at least in part through Rac1 hyperactivation and subsequent defective DC migration. As this phenotype can be corrected using thiopurines, ATG16L1 genotyping may be useful in the identification of patients that will benefit most from thiopurine treatment.
Collapse
Affiliation(s)
- M E Wildenberg
- Tytgat Institute for Intestinal and Liver Research, Academic Medical Center, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - P J Koelink
- Tytgat Institute for Intestinal and Liver Research, Academic Medical Center, Amsterdam, The Netherlands
| | - K Diederen
- Tytgat Institute for Intestinal and Liver Research, Academic Medical Center, Amsterdam, The Netherlands
| | - A A Te Velde
- Tytgat Institute for Intestinal and Liver Research, Academic Medical Center, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - S C S Wolfkamp
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - V J Nuij
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - M P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - M Nobis
- Beatson Institute for Cancer Research, Bearsden, Glasgow, UK
| | - O J Sansom
- Beatson Institute for Cancer Research, Bearsden, Glasgow, UK
| | - K I Anderson
- Beatson Institute for Cancer Research, Bearsden, Glasgow, UK
| | - C J van der Woude
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, The Netherlands
| | - G R A M D'Haens
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | - G R van den Brink
- Tytgat Institute for Intestinal and Liver Research, Academic Medical Center, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Peng LL, Wang Y, Zhu FL, Xu WD, Ji XL, Ni J. IL-23R mutation is associated with ulcerative colitis: A systemic review and meta-analysis. Oncotarget 2017; 8:4849-4863. [PMID: 27902482 PMCID: PMC5354875 DOI: 10.18632/oncotarget.13607] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/12/2016] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Since a genome-wide association study revealed that Interleukin-23 receptor (IL-23R) gene is a candidate gene for Ulcerative Colitis (UC), many studies have investigated the association between the IL-23R polymorphisms and UC. However, the results were controversial. The aim of the study was to determine whether the IL-23R polymorphisms confer susceptibility to UC. METHODS A systematic literature search was carried out to identify all potentially relevant studies. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were used to estimate the strength of association. RESULTS A total of 33 studies in 32 articles, including 10,527 UC cases and 15,142 healthy controls, were finally involved in the meta-analysis. Overall, a significant association was found between all UC cases and the rs11209026A allele (OR = 0.665, 95% CI = 0.604~0.733, P < 0.001). Similarly, meta-analyses of the rs7517847, rs1004819, rs10889677, rs2201841, rs11209032, rs1495965, rs1343151 and rs11465804 polymorphisms also indicated significant association with all UC (all P < 0.05). Stratification by ethnicity revealed that the rs11209026, rs7517847, rs10889677, rs2201841 andrs11465804 polymorphisms were associated with UC in the Caucasian group, but not in Asians, while the rs1004819 and rs11209032 polymorphisms were found to be related to UC for both Caucasian and Asian groups. However, subgroup analysis failed to unveil any association between the rs1495965 and rs1343151 polymorphisms and UC in Caucasians or Asians. CONCLUSIONS The meta-analysis suggests significant association between IL-23R polymorphisms and UC, especially in Caucasians.
Collapse
Affiliation(s)
- Ling-Long Peng
- Department of Science and Education, The Second People's Hospital of Wuhu, Wuhu, Anhui 241000, China
| | - Ying Wang
- Department of Environmental Health, Suzhou Municipal Center for Disease Prevention and Control, Suzhou, Jiangsu 215004, China
| | - Feng-Ling Zhu
- Department of Science and Education, The Second People's Hospital of Wuhu, Wuhu, Anhui 241000, China
| | - Wang-Dong Xu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xue-Lei Ji
- Department of Science and Education, The Second People's Hospital of Wuhu, Wuhu, Anhui 241000, China
| | - Jing Ni
- The Teaching Centre for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
21
|
Uniken Venema WT, Voskuil MD, Dijkstra G, Weersma RK, Festen EA. The genetic background of inflammatory bowel disease: from correlation to causality. J Pathol 2016; 241:146-158. [PMID: 27785786 DOI: 10.1002/path.4817] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/13/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022]
Abstract
Recent studies have greatly improved our insight into the genetic background of inflammatory bowel disease (IBD). New high-throughput technologies and large-scale international collaborations have contributed to the identification of 200 independent genetic risk loci for IBD. However, in most of these loci, it is unclear which gene conveys the risk for IBD. More importantly, it is unclear which variant within or near the gene is causal to the disease. Using targeted GWAS, imputation, resequencing of risk loci, and in silico fine-mapping of densely typed loci, several causal variants have been identified in IBD risk genes, and various pathological pathways have been uncovered. Current research in the field of IBD focuses on the effect of these causal variants on gene expression and protein function. However, more elements than only the genome must be taken into account to disentangle the multifactorial pathology of IBD. The genetic risk loci identified to date only explain a small part of genetic variance in disease risk. Currently, large multi-omics studies are incorporating factors ranging from the gut microbiome to the environment. In this review, we present the progress that has been made in IBD genetic research and stress the importance of studying causality to increase our understanding of the pathogenesis of IBD. We highlight important causal genetic variants in the candidate genes NOD2, ATG16L1, IRGM, IL23R, CARD9, RNF186, and PRDM1. We describe their downstream effects on protein function and their direct effects on the gut immune system. Furthermore, we discuss the future role of genetics in unravelling disease mechanisms in IBD. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Werna Tc Uniken Venema
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel D Voskuil
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Eleonora Am Festen
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands.,Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Pilla-Moffett D, Barber MF, Taylor GA, Coers J. Interferon-Inducible GTPases in Host Resistance, Inflammation and Disease. J Mol Biol 2016; 428:3495-513. [PMID: 27181197 PMCID: PMC5010443 DOI: 10.1016/j.jmb.2016.04.032] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/23/2016] [Accepted: 04/30/2016] [Indexed: 01/18/2023]
Abstract
Cell-autonomous immunity is essential for host organisms to defend themselves against invasive microbes. In vertebrates, both the adaptive and the innate branches of the immune system operate cell-autonomous defenses as key effector mechanisms that are induced by pro-inflammatory interferons (IFNs). IFNs can activate cell-intrinsic host defenses in virtually any cell type ranging from professional phagocytes to mucosal epithelial cells. Much of this IFN-induced host resistance program is dependent on four families of IFN-inducible GTPases: the myxovirus resistance proteins, the immunity-related GTPases, the guanylate-binding proteins (GBPs), and the very large IFN-inducible GTPases. These GTPase families provide host resistance to a variety of viral, bacterial, and protozoan pathogens through the sequestration of microbial proteins, manipulation of vesicle trafficking, regulation of antimicrobial autophagy (xenophagy), execution of intracellular membranolytic pathways, and the activation of inflammasomes. This review discusses our current knowledge of the molecular function of IFN-inducible GTPases in providing host resistance, as well as their role in the pathogenesis of autoinflammatory Crohn's disease. While substantial advances were made in the recent past, few of the known functions of IFN-inducible GTPases have been explored in any depth, and new functions await discovery. This review will therefore highlight key areas of future exploration that promise to advance our understanding of the role of IFN-inducible GTPases in human diseases.
Collapse
Affiliation(s)
- Danielle Pilla-Moffett
- Department of Molecular Genetics and Microbiology, and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Matthew F Barber
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Gregory A Taylor
- Department of Medicine, Duke University, Durham, NC 27708, USA; Department of Molecular Genetics and Microbiology, and Immunology, Duke University, Durham, NC 27708, USA; Center for the Study of Aging, Duke University, Durham, NC 27708, USA; Geriatric Research and Education and Clinical Center, Veteran Affairs Medical Center, Durham, NC 27710, USA.
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, and Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
23
|
Hong SN, Park C, Park SJ, Lee CK, Ye BD, Kim YS, Lee S, Chae J, Kim JI, Kim YH. Deep resequencing of 131 Crohn's disease associated genes in pooled DNA confirmed three reported variants and identified eight novel variants. Gut 2016; 65:788-96. [PMID: 25731871 DOI: 10.1136/gutjnl-2014-308617] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/27/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Genome wide association studies (GWAS) and meta-analyses for Crohn's disease (CD) have not fully explained the heritability of CD, suggesting that additional loci are yet to be found and that the known loci may contain high effect rare risk variants that have thus far gone undetected by GWAS. While the cost of deep sequencing remains too high to analyse many samples, targeted sequencing of pooled DNA samples allows the efficient and cost effective capture of all variations in a target region. DESIGN We performed pooled sequencing in 500 Korean CD cases and 1000 controls to evaluate the coding exon and 5' and 3' untranslated regions of 131 CD associated genes. The identified genetic variants were validated using genotyping in an independent set of 500 CD cases and 1000 controls. RESULTS Pooled sequencing identified 30 common/low single nucleotide variants (SNVs) in 12 genes and 3 rare SNVs in 3 genes. Our results confirmed a significant association of CD with the following previously reported risk loci: rs3810936 in TNFSF15 (OR=1.83, p<2.2×10(-16)), rs76418789 in IL23R (OR=0.47, p=1.14×10(-8)) and rs2241880 in ATG16L1 (OR=1.30, p=5.28×10(-6)). In addition, novel loci were identified in TNFSF8 (rs3181374, OR=1.53, p=1.03×10(-14)), BTNL2 (rs28362680, OR=1.47, p=9.67×10(-11)), HLA-DQA2 (rs3208181, OR=1.36, p=4.66×10(-6)), STAT3 (rs1053004, OR=1.29, p=2.07×10(-5)), NFKBIA (rs2273650, OR=0.80, p=3.93×10(-4)), NKX2-3 (rs888208, OR=0.82, p=6.37×10(-4)) and DNAH12 (rs4462937, OR=1.13, p=3.17×10(-2)). A novel rare SNV, rs200735402 in CARD9, was shown to have a protective effect (OR=0.09, p=5.28×10(-5)). CONCLUSIONS Our deep resequencing of 131 CD associated genes confirmed 3 reported risk loci and identified 8 novel risk loci for CD in Koreans, providing new insights into the genetic architecture of CD.
Collapse
Affiliation(s)
- Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Changho Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Jung Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Kyun Lee
- Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Byong Duk Ye
- Department of Gastroenterology and Inflammatory Bowel Disease Center, Asan Medical Centre, University of Ulsan College of Medicine
| | - You Sun Kim
- Department of Internal Medicine, Seoul Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Seungbok Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Jeesoo Chae
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | |
Collapse
|
24
|
Zupančič K, Skok K, Repnik K, Weersma RK, Potočnik U, Skok P. Multi-locus genetic risk score predicts risk for Crohn's disease in Slovenian population. World J Gastroenterol 2016; 22:3777-3784. [PMID: 27076762 PMCID: PMC4814740 DOI: 10.3748/wjg.v22.i14.3777] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/01/2016] [Accepted: 03/13/2016] [Indexed: 02/07/2023] Open
Abstract
AIM To develop a risk model for Crohn's disease (CD) based on homogeneous population. METHODS In our study were included 160 CD patients and 209 healthy individuals from Slovenia. The association study was performed for 112 single nucleotide polymorphisms (SNPs). We generated genetic risk scores (GRS) based on the number of risk alleles using weighted additive model. Discriminatory accuracy was measured by area under ROC curve (AUC). For risk evaluation, we divided individuals according to positive and negative likelihood ratios (LR) of a test, with LR > 5 for high risk group and LR < 0.20 for low risk group. RESULTS The highest accuracy, AUC of 0.78 was achieved with GRS combining 33 SNPs with optimal sensitivity and specificity of 75.0% and 72.7%, respectively. Individuals with the highest risk (GRS > 5.54) showed significantly increased odds of developing CD (OR = 26.65, 95%CI: 11.25-63.15) compared to the individuals with the lowest risk (GRS < 4.57) which is a considerably greater risk captured than in one SNP with the highest effect size (OR = 3.24). When more than 33 SNPs were included in GRS, discriminatory ability was not improved significantly; AUC of all 74 SNPs was 0.76. CONCLUSION The authors proved the possibility of building accurate genetic risk score based on 33 risk variants on Slovenian CD patients which may serve as a screening tool in the targeted population.
Collapse
|
25
|
Abstract
Genome-wide association studies (GWAS) have associated many single variants with complex disease, yet the better part of heritable complex disease risk remains unexplained. Analytical tools designed to work under specific population genetic models are needed. Rare variants are increasingly shown to be important in human complex disease, but most existing GWAS data do not cover rare variants. Explicit population genetic models predict that genes contributing to complex traits and experiencing recurrent, unconditionally deleterious, mutation will harbor multiple rare, causative mutations of subtle effect. It is difficult to identify genes harboring rare variants of large effect that contribute to complex disease risk via the single marker association tests typically used in GWAS. Gene/region-based association tests may have the power detect associations by combining information from multiple markers, but have yielded limited success in practice. This is partially because many methods have not been widely applied. Here, we empirically demonstrate the utility of a procedure based on the rank truncated product (RTP) method, filtered to reduce the effects of linkage disequilibrium. We apply the procedure to the Wellcome Trust Case Control Consortium (WTCCC) data set, and uncover previously unidentified associations, some of which have been replicated in much larger studies. We show that, in the absence of significant rare variant coverage, RTP based methods still have the power to detect associated genes. We recommend that RTP-based methods be applied to all existing GWAS data to maximize the usefulness of those data. For this, we provide efficient software implementing our procedure.
Collapse
|
26
|
Rieder F, Bettenworth D, Imai J, Inagaki Y. Intestinal Fibrosis and Liver Fibrosis: Consequences of Chronic Inflammation or Independent Pathophysiology? Inflamm Intest Dis 2016; 1:41-49. [PMID: 29922656 DOI: 10.1159/000445135] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023] Open
Abstract
Background Intestinal fibrosis and liver fibrosis represent a significant burden for our patients and health-care systems. Despite the severe clinical problem and the observation that fibrosis is reversible, no specific antifibrotic therapies exist. Summary In this review, using an 'East-West' scientific collaboration, we summarize the current knowledge on principal mechanisms shared by intestinal fibrosis and liver fibrosis. We furthermore discuss inflammation as the cause of fibrogenesis in both entities, depict unique features of intestinal and hepatic fibrosis, and provide a future outlook on the development of antifibrotic therapies. Key Messages A collaborative effort in the field of fibrosis, covering multiple organ systems, will have the highest chance of leading to the development of a successful antifibrotic intervention.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland, Ohio, USA.,Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | | | - Jin Imai
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan.,Department of Gastroenterology, Tokai University School of Medicine, Isehara, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan.,Department of Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
27
|
Spalinger MR, McCole DF, Rogler G, Scharl M. Protein tyrosine phosphatase non-receptor type 2 and inflammatory bowel disease. World J Gastroenterol 2016; 22:1034-1044. [PMID: 26811645 PMCID: PMC4716018 DOI: 10.3748/wjg.v22.i3.1034] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/31/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
Genome wide association studies have associated single nucleotide polymorphisms within the gene locus encoding protein tyrosine phosphatase non-receptor type 2 (PTPN2) with the onset of inflammatory bowel disease (IBD) and other inflammatory disorders. Expression of PTPN2 is enhanced in actively inflamed intestinal tissue featuring a marked up-regulation in intestinal epithelial cells. PTPN2 deficient mice suffer from severe intestinal and systemic inflammation and display aberrant innate and adaptive immune responses. In particular, PTPN2 is involved in the regulation of inflammatory signalling cascades, and critical for protecting intestinal epithelial barrier function, regulating innate and adaptive immune responses, and finally for maintaining intestinal homeostasis. On one hand, dysfunction of PTPN2 has drastic effects on innate host defence mechanisms, including increased secretion of pro-inflammatory cytokines, limited autophagosome formation in response to invading pathogens, and disruption of the intestinal epithelial barrier. On the other hand, PTPN2 function is crucial for controlling adaptive immune functions, by regulating T cell proliferation and differentiation as well as maintaining T cell tolerance. In this way, dysfunction of PTPN2 contributes to the manifestation of IBD. The aim of this review is to present an overview of recent findings on the role of PTPN2 in intestinal homeostasis and the impact of dysfunctional PTPN2 on intestinal inflammation.
Collapse
|
28
|
Xu WD, Xie QB, Zhao Y, Liu Y. Association of Interleukin-23 receptor gene polymorphisms with susceptibility to Crohn's disease: A meta-analysis. Sci Rep 2015; 5:18584. [PMID: 26678098 PMCID: PMC4683513 DOI: 10.1038/srep18584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/20/2015] [Indexed: 02/05/2023] Open
Abstract
Studies investigating the association between Interleukin-23 receptor (IL-23R) gene polymorphisms and Crohn’s disease (CD) report conflicting results. Thus, a meta-analysis was carried out to assess the association between the IL-23R polymorphisms and CD. A systematic literature search was conducted to identify all relevant studies. Pooled odds ratio (ORs) with 95% confidence interval (CIs) was used to estimate the strength of association. Finally, a total of 60 case-control studies in 56 articles, involving 22,820 CD patients and 27,401 healthy controls, were included in the meta-analysis. Overall, a significant association was found between all CD and the rs7517847 polymorphism (OR = 0.699, 95% CI = 0.659 ~ 0.741, P < 0.001). Meta-analysis of the rs11209026, rs1343151, rs10489629 and rs11465804 polymorphisms indicated the same pattern as for rs7517847. Meta-analysis showed an association between the rs10889677A allele and CD (OR = 1.393, 95% CI = 1.328 ~ 1.461, P < 0.001). Similarly, meta-analysis of the rs2201840, rs1004819, rs1495965 and rs11209032 polymorphisms revealed the same pattern as that shown by meta-analysis of rs10889677. Stratification by ethnicity revealed that IL-23R gene polymorphisms were associated with CD in the Caucasian group, but not in Asians. In summary, the meta-analysis suggests a significant association between IL-23R polymorphisms and CD, especially in Caucasians.
Collapse
Affiliation(s)
- Wang-Dong Xu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan, 610041, PR China
| | - Qi-Bing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan, 610041, PR China
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan, 610041, PR China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, Sichuan, 610041, PR China
| |
Collapse
|
29
|
Rodriguez-Rodriguez L, Ivorra-Cortes J, Carmona FD, Martín J, Balsa A, van Steenbergen HW, van der Helm-van Mil AHM, González-Álvaro I, Fernandez-Gutiérrez B. PTGER4 gene variant rs76523431 is a candidate risk factor for radiological joint damage in rheumatoid arthritis patients: a genetic study of six cohorts. Arthritis Res Ther 2015; 17:306. [PMID: 26538147 PMCID: PMC4634155 DOI: 10.1186/s13075-015-0830-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction Prostaglandin E receptor 4 (PTGER4) is implicated in immune regulation and bone metabolism. The aim of this study was to analyze its role in radiological joint damage in rheumatoid arthritis (RA). Methods Six independent cohorts of patients with RA of European or North American descent were included, comprising 1789 patients with 5083 sets of X-rays. The Hospital Clínico San Carlos Rheumatoid Arthritis, Princesa Early Arthritis Register Longitudinal study, and Hospital Universitario de La Paz early arthritis (Spain) cohorts were used as discovery cohorts, and the Leiden Early Arthritis Clinic (The Netherlands), Wichita (United States), and National Databank for Rheumatic Diseases (United States and Canada) cohorts as replication cohorts. First, the PTGER4 rs6896969 single-nucleotide polymorphism (SNP) was genotyped using TaqMan assays and available Illumina Immunochip data and studied in the discovery and replication cohorts. Second, the PTGER4 gene and adjacent regions were analyzed using Immunochip genotyping data in the discovery cohorts. On the basis of pooled p values, linkage disequilibrium structure of the region, and location in regions with transcriptional properties, SNPs were selected for replication. The results from discovery, replication, and overall cohorts were pooled using inverse-variance–weighted meta-analysis. Influence of the polymorphisms on the overall radiological damage (constant effect) and on damage progression over time (time-varying effect) was analyzed. Results The rs6896969 polymorphism showed a significant association with radiological damage in the constant effect pooled analysis of the discovery cohorts, although no significant association was observed in the replication cohorts or the overall pooled analysis. Regarding the analysis of the PTGER4 region, 976 variants were analyzed in the discovery cohorts. From the constant and time-varying effect analyses, 12 and 20 SNPs, respectively, were selected for replication. Only the rs76523431 variant showed a significant association with radiographic progression in the time-varying effect pooled analysis of the discovery, replication, and overall cohorts. The overall pooled effect size was 1.10 (95 % confidence interval 1.05–1.14, p = 2.10 × 10−5), meaning that radiographic yearly progression was 10 % greater for each copy of the minor allele. Conclusions The PTGER4 gene is a candidate risk factor for radiological progression in RA. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0830-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luis Rodriguez-Rodriguez
- Rheumatology Department and Heath Research Institute (IdISSC), Hospital Clinico San Carlos, c/o Prof. Martin Lagos s/n, 28040, Madrid, Spain.
| | - Jose Ivorra-Cortes
- Rheumatology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain.
| | - F David Carmona
- Instituto de Parasitología y Biomedicina 'López-Neyra', CSIC, Granada, Spain.
| | - Javier Martín
- Instituto de Parasitología y Biomedicina 'López-Neyra', CSIC, Granada, Spain.
| | - Alejandro Balsa
- Rheumatology Department and Heath Research Institute (Idipaz), Hospital Universitario de La Paz, Madrid, Spain.
| | | | | | - Isidoro González-Álvaro
- Rheumatology Service and Heath Research Institute (IP), Hospital Universitario de La Princesa, Madrid, Spain.
| | - Benjamín Fernandez-Gutiérrez
- Rheumatology Department and Heath Research Institute (IdISSC), Hospital Clinico San Carlos, c/o Prof. Martin Lagos s/n, 28040, Madrid, Spain.
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Genome-wide association studies (GWAS) have helped to understand the genetic basis and pathogenesis of inflammatory bowel disease (IBD). However, understanding the functional and clinical consequences of the associated alleles has not followed the same pace. In this review, we discuss how studying the genetic predisposition to IBD has increased our understanding about IBD pathogenesis and how epigenetics is becoming more and more important. We describe the potential clinical applications of genetics, and provide important challenges in the field and the future steps to be taken. RECENT FINDINGS GWAS and meta-analyses have identified 163 loci associated with IBD, and have implicated key pathways in IBD pathogenesis. Only few of the association signals correspond to nonsynonymous coding variation with a clear effect on protein function. The majority of signals involve noncoding genetic variation, of which a large part is related to gene expression changes. More recently, expression and epigenetic studies in IBD are increasingly being reported, and have shown that many effects seem to be highly cell-type specific.Predictive genetic testing will not be for the immediate future for the majority of IBD patients. However, for the subset of patients with very-early onset IBD, several causal mutations have been found. Predictive genetic panels for these adolescents presenting with a very severe disease course, and/or families with high penetrance of disease will be of benefit. SUMMARY Genetic, transcriptomic and epigenetic studies have offered exciting clues about IBD pathogenesis but are unlikely to provide all answers. To fully grasp the function of disease-associated genetic variants, identifying causal genes and translating this knowledge into predictive biomarkers and new treatments, we should now integrate all these disciplines.
Collapse
|
31
|
Autophagy and inflammatory bowel disease: Association between variants of the autophagy-related IRGM gene and susceptibility to Crohn's disease. Dig Liver Dis 2015; 47:744-50. [PMID: 26066377 DOI: 10.1016/j.dld.2015.05.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/25/2015] [Accepted: 05/14/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Crohn's disease and ulcerative colitis are inflammatory bowel diseases involving a genetically determined inappropriate mucosal immune response towards luminal antigens, including resident bacterial flora. Recent studies identified susceptibility genes involved in autophagy. AIMS We analyzed known autophagic loci (IRGM, ULK1 and AMBRA1) previously described as associated with inflammatory bowel diseases or with other autoimmune and/or inflammatory disorders in a sample of Italian inflammatory bowel diseases patients in order to confirm their possible involvement and relative contribution in the disease. METHODS We performed a case-control association study, a sub-phenotype correlation and a haplotype analysis. The analysis included 263 Crohn's disease, 206 ulcerative colitis patients and 245 matched healthy controls. Five polymorphisms were genotyped by allelic discrimination assays. RESULTS IRGM was the most strongly associated with Crohn's disease susceptibility [rs13361189: P=0.011, OR=1.66 [95% CI: (1.12-2.45)]; rs4958847: P=0.05, OR=1.43 [95% CI: (1-2.03)]. The SNP rs13361189 was also found to increase the risk of Crohn's disease clinical sub-phenotype (fibrostricturing behaviour, ileal disease, perianal disease, intestinal resection). These findings suggest that IRGM variants may modulate clinical characteristics of Crohn's disease. CONCLUSIONS Our study confirms IRGM rs13361189 and rs4958847 polymorphisms to be important for Crohn's disease susceptibility and phenotype modulation, in accordance with previous findings.
Collapse
|
32
|
Leppkes M, Neurath MF, Herrmann M, Becker C. Immune deficiency vs. immune excess in inflammatory bowel diseases-STAT3 as a rheo-STAT of intestinal homeostasis. J Leukoc Biol 2015; 99:57-66. [PMID: 26232455 DOI: 10.1189/jlb.5mr0515-221r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/02/2015] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies have provided many genetic alterations, conferring susceptibility to multifactorial polygenic diseases, such as inflammatory bowel diseases. Yet, how specific genetic alterations functionally affect intestinal inflammation often remains elusive. It is noteworthy that a large overlap of genes involved in immune deficiencies with those conferring inflammatory bowel disease risk has been noted. This has provided new arguments for the debate on whether inflammatory bowel disease arises from either an excess or a deficiency in the immune system. In this review, we highlight the functional effect of an inflammatory bowel disease-risk allele, which cannot be deduced from genome-wide association studies data alone. As exemplified by the transcription factor signal transducer and activator of transcription 3 (STAT3), we show that a single gene can have a plethora of effects in various cell types of the gut. These effects may individually contribute to the restoration of intestinal homeostasis on the one hand or pave the way for excessive immunopathology on the other, as an inflammatory "rheo-STAT".
Collapse
Affiliation(s)
- Moritz Leppkes
- *Medical Clinic 1 and Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Markus F Neurath
- *Medical Clinic 1 and Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Martin Herrmann
- *Medical Clinic 1 and Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| | - Christoph Becker
- *Medical Clinic 1 and Medical Clinic 3, University Clinic, Friedrich Alexander University, Erlangen, Germany
| |
Collapse
|
33
|
Role of protein tyrosine phosphatases in regulating the immune system: implications for chronic intestinal inflammation. Inflamm Bowel Dis 2015; 21:645-55. [PMID: 25581833 PMCID: PMC4329025 DOI: 10.1097/mib.0000000000000297] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Current hypothesis suggests that genetic, immunological, and bacterial factors contribute essentially to the pathogenesis of inflammatory bowel disease. Variations within the gene loci encoding protein tyrosine phosphatases (PTPs) have been associated with the onset of inflammatory bowel disease. PTPs modulate the activity of their substrates by dephosphorylation of tyrosine residues and are critical for the regulation of fundamental cellular signaling processes. Evidence emerges that expression levels of PTPN2, PTPN11, and PTPN22 are altered in actively inflamed intestinal tissue. PTPN2 seems to be critical for protecting intestinal epithelial barrier function, regulating innate and adaptive immune responses and finally for maintaining intestinal homeostasis. These observations have been confirmed in PTPN2 knockout mice in vivo. Those animals are clearly more susceptible to intestinal and systemic inflammation and feature alterations in innate and adaptive immune responses. PTPN22 controls inflammatory signaling in lymphocytes and mononuclear cells resulting in aberrant cytokine secretion pattern and autophagosome formation. PTPN22 deficiency in vivo results in more severe colitis demonstrating the relevance of PTPN22 for intestinal homeostasis in vivo. Of note, loss of PTPN22 promotes mitogen-activated protein kinase-induced cytokine secretion but limits secretion of nuclear factor κB-associated cytokines and autophagy in mononuclear cells. Loss of PTPN11 is also associated with increased colitis severity in vivo. In summary, dysfunction of those PTPs results in aberrant and uncontrolled immune responses that result in chronic inflammatory conditions. This way, it becomes more and more evident that dysfunction of PTPs displays an important factor in the pathogenesis of chronic intestinal inflammation, in particular inflammatory bowel disease.
Collapse
|
34
|
Rieder F, de Bruyn JR, Pham BT, Katsanos K, Annese V, Higgins PDR, Magro F, Dotan I. Results of the 4th scientific workshop of the ECCO (Group II): markers of intestinal fibrosis in inflammatory bowel disease. J Crohns Colitis 2014; 8:1166-1178. [PMID: 24726695 DOI: 10.1016/j.crohns.2014.03.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/12/2014] [Accepted: 03/15/2014] [Indexed: 02/08/2023]
Abstract
The fourth scientific workshop of the European Crohn's and Colitis Organization (ECCO) focused on intestinal fibrosis in inflammatory bowel disease (IBD). The objective was to better understand basic mechanisms and markers of intestinal fibrosis as well as to suggest new therapeutic targets to prevent or treat fibrosis. The results of this workshop are presented in three separate manuscripts. This section describes markers of fibrosis in IBD, identifies unanswered questions in the field and provides a framework for future studies addressing the unmet needs in the field of intestinal fibrosis.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Department of Gastroenterology & Hepatology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Jessica R de Bruyn
- Academic Medical Center Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands.
| | - Bao Tung Pham
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, The Netherlands.
| | - Konstantinos Katsanos
- Department of Gastroenterology, University Hospital of Ioannina, Medical School of Ioannina, Greece.
| | - Vito Annese
- Division of Gastroenterology, University Hospital Careggi, Florence, Italy.
| | - Peter D R Higgins
- Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA.
| | - Fernando Magro
- Department of Pharmacology & Therapeutics, Institute for Molecular and Cell Biology, Faculty of Medicine University of Porto, Porto, Portugal; Department of Gastroenterology, Hospital de Sao Joao, Porto, Portugal.
| | - Iris Dotan
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
35
|
Abstract
Technological advances in the large scale analysis of human genetics have generated profound insights into possible genetic contributions to chronic diseases including the inflammatory bowel diseases (IBDs), Crohn's disease and ulcerative colitis. To date, 163 distinct genetic risk loci have been associated with either Crohn's disease or ulcerative colitis, with a substantial degree of genetic overlap between these 2 conditions. Although many risk variants show a reproducible correlation with disease, individual gene associations only affect a subset of patients, and the functional contribution(s) of these risk variants to the onset of IBD is largely undetermined. Although studies in twins have demonstrated that the development of IBD is not mediated solely by genetic risk, it is nevertheless important to elucidate the functional consequences of risk variants for gene function in relevant cell types known to regulate key physiological processes that are compromised in IBD. This article will discuss IBD candidate genes that are known to be, or are suspected of being, involved in regulating the intestinal epithelial barrier and several of the physiological processes presided over by this dynamic and versatile layer of cells. This will include assembly and regulation of tight junctions, cell adhesion and polarity, mucus and glycoprotein regulation, bacterial sensing, membrane transport, epithelial differentiation, and restitution.
Collapse
|
36
|
Serbati N, Senhaji N, Diakite B, Badre W, Nadifi S. IL23R and ATG16L1 variants in Moroccan patients with inflammatory bowel disease. BMC Res Notes 2014; 7:570. [PMID: 25159710 PMCID: PMC4162942 DOI: 10.1186/1756-0500-7-570] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/14/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are chronic diseases of the gastrointestinal tract. Although their pathogenesis is unclear, the combination of genetic predisposition and environmental components are believed to be the main cause of these diseases. Recently, many variants in interleukin 23 receptor (IL23R) and autophagy-related 16-like 1 (ATG16L1) genes have been associated with the disease. Our objective was to assess the frequency of ATG16L1 (T300A) and IL23R (L310P) variants in Moroccan IBD (Crohn's disease and Ulcerative Colitis) patients and to evaluate a possible effect of these variants on disease's phenotype and clinical course. METHODS 96 Moroccan IBD patients and 114 unrelated volunteers were genotyped for ATG16L1 (T300A) and IL23R (L310P) variants by PCR-restriction fragment length polymorphism. RESULTS This is the first report on the prevalence of ATG16L1 (T300A) and IL23R (L310P) variants in a Moroccan group. We found that IL23R (L310P) variant conferred a protective effect for crohn's disease (CD) but not ulcerative colitis (UC) patients. The presence of ATG16L1 (T300A) mutated alleles was associated with CD type but not with disease onset. In addition, the carriage of T300A variant alleles conferred a protective effect in UC. CONCLUSION Our results showed that the prevalence of ATG16L1 and IL23R variants was not significantly different between patients and controls. However a possible role of ATG16L1 (T300A) on CD phenotype was suggested.
Collapse
Affiliation(s)
- Nadia Serbati
- Laboratory of Medical Genetics- Medical school of Casablanca, Faculté de Médecine et de Pharmacie de Casablanca, 19, rue Tarik ibn ziad, Casablanca, Morocco.
| | | | | | | | | |
Collapse
|
37
|
Dolejsi E, Bodenstorfer B, Frommlet F. Analyzing genome-wide association studies with an FDR controlling modification of the Bayesian Information Criterion. PLoS One 2014; 9:e103322. [PMID: 25061809 PMCID: PMC4111553 DOI: 10.1371/journal.pone.0103322] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/01/2014] [Indexed: 01/24/2023] Open
Abstract
The prevailing method of analyzing GWAS data is still to test each marker individually, although from a statistical point of view it is quite obvious that in case of complex traits such single marker tests are not ideal. Recently several model selection approaches for GWAS have been suggested, most of them based on LASSO-type procedures. Here we will discuss an alternative model selection approach which is based on a modification of the Bayesian Information Criterion (mBIC2) which was previously shown to have certain asymptotic optimality properties in terms of minimizing the misclassification error. Heuristic search strategies are introduced which attempt to find the model which minimizes mBIC2, and which are efficient enough to allow the analysis of GWAS data. Our approach is implemented in a software package called MOSGWA. Its performance in case control GWAS is compared with the two algorithms HLASSO and d-GWASelect, as well as with single marker tests, where we performed a simulation study based on real SNP data from the POPRES sample. Our results show that MOSGWA performs slightly better than HLASSO, where specifically for more complex models MOSGWA is more powerful with only a slight increase in Type I error. On the other hand according to our simulations GWASelect does not at all control the type I error when used to automatically determine the number of important SNPs. We also reanalyze the GWAS data from the Wellcome Trust Case-Control Consortium and compare the findings of the different procedures, where MOSGWA detects for complex diseases a number of interesting SNPs which are not found by other methods.
Collapse
Affiliation(s)
- Erich Dolejsi
- Center for Medical Statistics, Informatics, and Intelligent Systems/Section of Medical Statistics, Medical University Vienna, Vienna, Austria
| | | | - Florian Frommlet
- Center for Medical Statistics, Informatics, and Intelligent Systems/Section of Medical Statistics, Medical University Vienna, Vienna, Austria
| |
Collapse
|
38
|
The expression of FAS-associated factor 1 and heat shock protein 70 in ovarian cancer. Obstet Gynecol Sci 2014; 57:281-90. [PMID: 25105101 PMCID: PMC4124089 DOI: 10.5468/ogs.2014.57.4.281] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE In this study, we evaluated the expression of FAS-associated factor 1 (FAF1) and heat shock protein 70 (HSP70) in normal ovary and ovarian cancer, and also analyzed the correlation between FAF1 and HSP70 in ovarian cancer. METHODS The patient group consisted of 29 unrelated Korean women diagnosed as ovarian cancers and control samples were obtained from 7 patients who underwent oophorectomy for benign disease of uterus, and normal ovary was confirmed histologically from biopsy. We examined FAF1 and HSP70 expression by western blot analysis and immunohistochemical staining in normal ovary and ovarian cancer. Furthermore, we examined a correlation between FAF1 and HSP70 in ovarian cancer. RESULTS The expression of FAF1 was lower in ovarian cancer than that in normal ovary (P=0.02), and the expression of HSP70 was increased in ovarian cancer in comparison to that in normal ovary (P=0.03). The expression of FAF1 was decreased in advanced stages (stage III or stage IV) as compared with early stages (stage I or stage II) (P=0.01). The expression of HSP70 was not significantly related with ovarian cancer histology (P=0.10), but the expression of HSP70 was most increased with papillary serous carcinomas and undifferentiated ovarian cancer. The expression of FAF1 was inversely correlated with the expression of HSP70 in ovarian cancer (Spearman correlation coefficience=-0.47). CONCLUSION We concluded that the expression of FAF1 or HSP70 each seems to have a meaning as a biomarker for early detection of ovarian cancer. The expressions of FAF1 and HSP70 seem to be more valuable in predicting ovarian cancer when used together because of their inverse correlation. This is the first study about the expression of FAF1 in ovarian cancer and the correlation between FAF1 and HSP70 expression in ovarian cancer.
Collapse
|
39
|
Contribution of NKX2-3 polymorphisms to inflammatory bowel diseases: a meta-analysis of 35358 subjects. Sci Rep 2014; 4:3924. [PMID: 24473197 PMCID: PMC5379238 DOI: 10.1038/srep03924] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/13/2014] [Indexed: 02/07/2023] Open
Abstract
Polymorphisms in NKX2-3 gene have been inconsistently associated with Crohn's disease (CD) and ulcerative colitis (UC). To generate large-scale evidence on whether NKX2-3 polymorphisms are associated with CD or UC susceptibility we have conducted a meta-analysis of 17 studies involving 17329 patients and 18029 controls. A significantly increased CD or UC risk was observed in persons carrying a G allele at rs10883365 polymorphism (A/G) compared with those with a A allele. (OR = 1.226, 95%CI: 1.177–1.277 and OR = 1.274, 95%CI: 1.175–1.382 respectively). In the subgroup analysis, a significantly increased CD risk was found in both Europeans and Asians. For rs11190140 polymorphism (C/T) and CD risk, the risk estimate for the allele contrast was OR = 1.201 (1.136–1.269). This meta-analysis provided a robust result that persons with a G or T allele may have a moderately increased risk of CD, and suggested that rs10883365 polymorphism was also a candidate gene polymorphism for UC susceptibility.
Collapse
|
40
|
Rieder F. The gut microbiome in intestinal fibrosis: environmental protector or provocateur? Sci Transl Med 2014; 5:190ps10. [PMID: 23785034 DOI: 10.1126/scitranslmed.3004731] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In individuals with inflammatory bowel diseases, intestinal fibrosis is a serious clinical complication with no specific therapies. Patients develop bowel fistulae and strictures that usually require surgery and often reoccur. The main driver of gut fibrogenesis is believed to be chronic inflammation, which leads to mesenchymal cell recruitment and activation. Recent findings suggest that the environment--in particular, the microbiome--plays a critical role in this process.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| |
Collapse
|
41
|
Lu XC, Tao Y, Wu C, Zhao PL, Li K, Zheng JY, Li LX. Association between variants of the autophagy related gene--IRGM and susceptibility to Crohn's disease and ulcerative colitis: a meta-analysis. PLoS One 2013; 8:e80602. [PMID: 24232856 PMCID: PMC3827440 DOI: 10.1371/journal.pone.0080602] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 10/04/2013] [Indexed: 01/17/2023] Open
Abstract
Background Polymorphisms in immunity-related GTPase family M (IRGM) gene may be associated with inflammatory bowel disease (IBD) by affecting autophagy. However, the genetic association studies on three common variants in IRGM gene (rs13361189, rs4958847 and rs10065172) have shown inconsistent results. Methodology/ Principal Findings The PubMed and Embase were searched up to June 5, 2013 for studies on the association between three IRGM polymorphisms and IBD risk. Data were extracted and the odd ratios (ORs) and 95% confidence intervals (95% CIs) were calculated. Finally, we performed a meta-analysis of 25 eligible studies in 3 SNPs located at IRGM gene by using a total of 20590 IBD cases and 27670 controls. The analysis showed modest significant association for the rs13361189, rs4958847 and rs10065172 variants in Crohn’s disease (CD): the risk estimates for the allele contrast were OR=1.306 (1.200-1.420), p=5.2×10-10, OR=1.182 (1.082-1.290), p=0.0002, and OR=1.248 (1.057-1.473), p=0.009 respectively (still significant when the p value was Bonferroni adjusted to 0.017). When stratified by ethnicity, significantly increased CD risk was observed in Europeans, but not in Asians. Conversely, there was no association of rs13361189 or rs4958847 variant with risk of ulcerative colitis (UC). Conclusions/ Significance These results indicated that autophagy gene-IRGM polymorphisms appear to confer susceptibility to CD but not UC, especially in Europeans. Our data may provide further understanding of the role of autophagy in the pathogenesis of CD.
Collapse
Affiliation(s)
- Xiao Cheng Lu
- Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
42
|
Long WY, Zhan LL, Chen L, Lv XP. Association of 10q21 gene rs10761659 polymorphism with susceptibility to inflammatory bowel disease in a Guangxi Zhuang population. Shijie Huaren Xiaohua Zazhi 2013; 21:3146-3152. [DOI: 10.11569/wcjd.v21.i29.3146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze the association of 10q21 gene rs10761659 polymorphism with susceptibility to inflammatory bowel disease (IBD) in a Zhuang population from Guangxi, China.
METHODS: A case-control study was conducted from August 2009 to October 2012, which included intestinal tissue samples of 73 Zhuang and 67 Han unrelated IBD patients and 70 Zhuang and 78 Han unrelated healthy volunteers in Guangxi. Genomic DNA was extracted from intestinal tissue using the phenol-chloroform method. The rs10761659 locus of the 10q21 gene was amplified by polymerase chain reaction (PCR), genotyped by restriction fragment length polymorphism (RFLP) and verified by gene sequencing.
RESULTS: The homozygous mutant, heterozygous mutant and wild-type genotypes of the rs10761659 locus were found in both IBD patients and healthy controls, but the distribution of genotypes and allele frequencies was not significantly different among Crohn's disease, ulcerative colitis and control groups (P > 0.05). No significant difference was detected in the 10q21 gene rs10761659 polymorphism between Zhuang and Han patients with Crohn's disease or ulcerative colitis (P > 0.05).
CONCLUSION: The rs10761659 polymorphism of the 10q21 gene is not associated with susceptibility to IBD in the Zhuang population in Guangxi.
Collapse
|
43
|
Maus B, Jung C, Mahachie John JM, Hugot JP, Génin E, Van Steen K. Molecular reclassification of Crohn's disease: a cautionary note on population stratification. PLoS One 2013; 8:e77720. [PMID: 24147066 PMCID: PMC3798408 DOI: 10.1371/journal.pone.0077720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/30/2013] [Indexed: 02/02/2023] Open
Abstract
Complex human diseases commonly differ in their phenotypic characteristics, e.g., Crohn's disease (CD) patients are heterogeneous with regard to disease location and disease extent. The genetic susceptibility to Crohn's disease is widely acknowledged and has been demonstrated by identification of over 100 CD associated genetic loci. However, relating CD subphenotypes to disease susceptible loci has proven to be a difficult task. In this paper we discuss the use of cluster analysis on genetic markers to identify genetic-based subgroups while taking into account possible confounding by population stratification. We show that it is highly relevant to consider the confounding nature of population stratification in order to avoid that detected clusters are strongly related to population groups instead of disease-specific groups. Therefore, we explain the use of principal components to correct for population stratification while clustering affected individuals into genetic-based subgroups. The principal components are obtained using 30 ancestry informative markers (AIM), and the first two PCs are determined to discriminate between continental origins of the affected individuals. Genotypes on 51 CD associated single nucleotide polymorphisms (SNPs) are used to perform latent class analysis, hierarchical and Partitioning Around Medoids (PAM) cluster analysis within a sample of affected individuals with and without the use of principal components to adjust for population stratification. It is seen that without correction for population stratification clusters seem to be influenced by population stratification while with correction clusters are unrelated to continental origin of individuals.
Collapse
Affiliation(s)
- Bärbel Maus
- UMR843, INSERM, Paris, France
- Bioinformatics and Modeling, GIGA-R, University of Liège, Liège, Belgium
| | - Camille Jung
- UMR843, Institut National de la Sante et de la recherche Medicale, Paris, France
- Service de Gastroentérologie Pédiatrique, Hôpital Robert Debré, APHP, Paris, France
- CRC-CRB, CHI Creteil, Creteil, France
| | - Jestinah M. Mahachie John
- UMR843, INSERM, Paris, France
- Bioinformatics and Modeling, GIGA-R, University of Liège, Liège, Belgium
| | - Jean-Pierre Hugot
- UMR843, Institut National de la Sante et de la recherche Medicale, Paris, France
- Service de Gastroentérologie Pédiatrique, Hôpital Robert Debré, APHP, Paris, France
- Labex Inflamex, Université Paris Diderot, Paris, France
| | - Emmanuelle Génin
- UMR1078, Génétique, Génomique fonctionnelle et Biotechnologies, INSERM, Brest, France
- Centre Hospitalier Régional Universitaire de Brest, Brest, France
| | - Kristel Van Steen
- UMR843, INSERM, Paris, France
- Bioinformatics and Modeling, GIGA-R, University of Liège, Liège, Belgium
| |
Collapse
|
44
|
Associations between PTPN2 polymorphisms and susceptibility to ulcerative colitis and Crohn's disease: a meta-analysis. Inflamm Res 2013; 63:71-9. [PMID: 24127071 DOI: 10.1007/s00011-013-0673-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/29/2013] [Accepted: 10/03/2013] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Ulcerative colitis (UC) and Crohn's disease (CD) result from an interaction between genetic and environmental factors. Though several polymorphisms have been identified in PTPN2, their roles in the incidence of UC and CD are conflicting. This meta-analysis was aimed to clarify the impact of these polymorphisms on UC and CD risk. METHOD PubMed, EMBASE, Cochrane Library and CBM were searched until 23 July 2013 for eligible studies on three PTPN2 polymorphisms: rs2542151, rs1893217 and rs7234029. Data were extracted, and pooled odd ratios (ORs) as well as 95 % confidence intervals (95 % CIs) were calculated. CONCLUSION The meta-analysis indicated that rs2542151, rs1893217 and rs1893217 were associated with increased CD risk, while the former was associated with increased UC risk. The differences in age of onset and ethnic groups may influence the associations. Gene-gene and gene-environment interactions should be investigated in the future. RESULTS Seventeen studies with 18,308 cases and 20,406 controls were included. Significant associations were found between rs2542151 polymorphism and CD susceptibility (OR = 1.22, 95 % CI, 1.15-1.30, I (2) = 32 %), as well as between rs2542151 and UC susceptibility (OR = 1.16, 95 % CI, 1.07-1.25, I (2) = 39 %). A similar result was found in Caucasians, but not in Asians. Moreover, a significant increase in CD risk for all carriers of the minor allele of rs1893217 (OR = 1.45, 95 % CI, 1.23-1.70, I (2) = 0 %) and rs7234029 (OR = 1.36, 95 % CI, 1.16-1.59, I (2) = 0 %) were found. For children, the rs1893217 polymorphism appeared to confer susceptibility to CD (OR = 1.56, 95 % CI, 1.28-1.89, I (2) = 0 %).
Collapse
|
45
|
Moon CM, Shin DJ, Son NH, Shin ES, Hong SP, Kim TI, Kim WH, Cheon JH. Genetic variants in the IL12B gene are associated with inflammatory bowel diseases in the Korean population. J Gastroenterol Hepatol 2013; 28:1588-94. [PMID: 23573954 DOI: 10.1111/jgh.12214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/01/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIM Recent genomic studies have identified genetic variants in the IL12B gene, which encodes the p40 subunit shared by interleukin 12 and interleukin 23, as susceptibility loci for inflammatory bowel disease (IBD). The study aimed to identify additional novel genetic variants in IL12B and investigated whether variants confer susceptibility to the development of Crohn's disease (CD) or ulcerative colitis (UC) in the Korean population. METHODS To detect single nucleotide polymorphisms (SNPs) in IL12B, direct sequencing of all coding exons, exon-intron boundaries, promoter region, and 5' untranslated region was performed in 24 randomly selected samples. Selected haplotype-tagging SNPs were subsequently genotyped in 493 IBD patients (245 patients with CD and 248 with UC) and 504 healthy controls. RESULTS Two haplotype-tagging SNPs (rs2288831 and rs919766) were selected through direct sequencing and were genotyped. Of them, SNP rs2288831 in the IL12B gene was significantly associated with CD susceptibility in allelic association analysis (odds ratio = 1.30; 95% confidence interval 1.04-1.62; P = 0.019). This significant association with CD was also observed for a haplotype consisting of SNP rs919766 and rs2288831 (odds ratio = 1.29; 95% confidence interval 1.03-1.60; P = 0.025). However, none of IL12B SNPs were associated with UC susceptibility. Finally, no specific associations between genetic variants and disease phenotype of CD were identified. CONCLUSIONS This study is first to identify SNP rs2288831 in the IL12B gene as a susceptible variation for CD. Further studies in other ethnic groups are warranted to validate the association of this genetic variant with IBD.
Collapse
Affiliation(s)
- Chang Mo Moon
- Department of Internal Medicine, Graduate School, Yonsei University College of Medicine, Seoul, Korea; Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The clinical course of inflammatory bowel disease (IBD) is highly heterogeneous and often unpredictable, with multiple and serious complications that range from stricture formation to bowel obstruction or perforation, fistula formation and the need for surgery. All these problems are manifestations of tissue remodeling, a secondary but universal response to the insults of chronic inflammation. The factors involved in tissue remodeling are several, including the site and duration of inflammation, soluble molecules, the gut microbiota, and the type of mesenchymal cell response. The prototypical and most common type of tissue remodeling in IBD, and Crohn's disease (CD) in particular, is a fibrotic response, and this review will focus on the factors and mechanisms involved in fibrogenesis, and speculate on what is needed for the development of a rational treatment of intestinal fibrosis.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Pathobiology, Lerner Research Institute, and Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | | |
Collapse
|
47
|
Abstract
BACKGROUND Genome-wide association studies have identified at least 71 Crohn's disease (CD) genetic risk loci, but the role of gene-gene interactions is unclear. The value of genetic variants in clinical practice is not defined because of limited explained heritability. METHODS We examined model predictability of combining the 71 CD risk alleles and genetic interactions in an ongoing inflammatory bowel disease genome-wide association study. The Wellcome Trust Case Control Consortium inflammatory bowel disease genome-wide association study was used as a replicate cohort. We used logic regression, an adaptive regression methodology, to search for high-order binary predictors (e.g., single-nucleotide polymorphism [SNP] interactions). RESULTS The combined 71 CD SNPs had good CD risk predictability (area under the curve of 0.75 and 0.73 in the 2 cohorts). Higher cumulative allele score predicted higher CD risk, but a relatively small difference in cumulative allele scores was observed between CD and controls (49 versus 47, P < 0.001). Through LR, we identified high-order genetic interactions and significantly improved the model predictability (area under the curve, from 0.75 to 0.77, P < 0.0001). A genetic interaction model, including NOD2, ATG16L1, IL10/IL19, C13orf31, and chr21q loci, was discovered and successfully replicated in the independent Wellcome Trust Case Control Consortium cohort. The explained heritability of the 71 CD SNPs alone was 24% and increased to 27% after adding the genetic interactions. CONCLUSIONS A novel approach allowed the identification and replication of genetic interactions among NOD2, ATG16L1, IL10/IL19, C13orf31, and chr21q loci. CD risk can be predicted by a model of 71 CD loci and improved by adding genetic interactions.
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW The inflammatory bowel diseases (IBDs) are chronic disabling conditions. Despite the benefits of anti-tumor necrosis factor (TNF)-α agents in improving quality of life and reducing the need for surgeries, overall only one-third of patients are in clinical remission at 1 year and loss of response is frequent. It seems clear that treatment must go beyond alleviation of symptoms in IBD. It is important that treatment targets in IBD will ensure mucosal healing and deep remission. RECENT FINDINGS The induction of deep remission might be the best way to alter the natural course of these diseases by preventing disability and bowel damage. New disability indices and the new Crohn's disease damage score have recently been developed and they can be used to evaluate the long-term effect on patients and as new endpoints in trials. Early intervention with disease-modifying anti-IBD drugs (DMAIDs) should be considered in patients with poor prognostic factors. SUMMARY New therapeutic targets in IBD patients who failed anti-TNF-α therapy are urgently required, and tofacitinib, vedolizumab and ustekinumab appear to be the most promising drugs. Herein, we review the new and current trends in IBD therapy, with the final aim of changing disease course and patients' lives by both improving quality of life and avoiding disability.
Collapse
|
49
|
IRGM variants and susceptibility to inflammatory bowel disease in the German population. PLoS One 2013; 8:e54338. [PMID: 23365659 PMCID: PMC3554777 DOI: 10.1371/journal.pone.0054338] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 12/12/2012] [Indexed: 01/11/2023] Open
Abstract
Background & Aims Genome-wide association studies identified the autophagy gene IRGM to be strongly associated with Crohn's disease (CD) but its impact in ulcerative colitis (UC), its phenotypic effects and potential epistatic interactions with other IBD susceptibility genes are less clear which we therefore analyzed in this study. Methodology/Principal Findings Genomic DNA from 2060 individuals including 817 CD patients, 283 UC patients, and 961 healthy, unrelated controls (all of Caucasian origin) was analyzed for six IRGM single nucleotide polymorphisms (SNPs) (rs13371189, rs10065172 = p.Leu105Leu, rs4958847, rs1000113, rs11747270, rs931058). In all patients, a detailed genotype-phenotype analysis and testing for epistasis with the three major CD susceptibility genes NOD2, IL23R and ATG16L1 were performed. Our analysis revealed an association of the IRGM SNPs rs13371189 (p = 0.02, OR 1.31 [95% CI 1.05–1.65]), rs10065172 = p.Leu105Leu (p = 0.016, OR 1.33 [95% CI 1.06–1.66]) and rs1000113 (p = 0.047, OR 1.27 [95% CI 1.01–1.61]) with CD susceptibility. There was linkage disequilibrium between these three IRGM SNPs. In UC, several IRGM haplotypes were weakly associated with UC susceptibility (p<0.05). Genotype-phenotype analysis revealed no significant associations with a specific IBD phenotype or ileal CD involvement. There was evidence for weak gene-gene-interaction between several SNPs of the autophagy genes IRGM and ATG16L1 (p<0.05), which, however, did not remain significant after Bonferroni correction. Conclusions/Significance Our results confirm IRGM as susceptibility gene for CD in the German population, supporting a role for the autophagy genes IRGM and ATG16L1 in the pathogenesis of CD.
Collapse
|
50
|
Almeida A. Genetic determinants of neuronal vulnerability to apoptosis. Cell Mol Life Sci 2013; 70:71-88. [PMID: 22695677 PMCID: PMC11113535 DOI: 10.1007/s00018-012-1029-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 04/22/2012] [Accepted: 05/07/2012] [Indexed: 12/18/2022]
Abstract
Apoptosis is a common mode of cell death that contributes to neuronal loss associated with neurodegeneration. Single-nucleotide polymorphisms (SNPs) in chromosomal DNA are contributing factors dictating natural susceptibility of humans to disease. Here, the most common SNPs affecting neuronal vulnerability to apoptosis are reviewed in the context of neurological disorders. Polymorphic variants in genes encoding apoptotic proteins, either from the extrinsic (FAS, TNF-α, CASP8) or the intrinsic (BAX, BCL2, CASP3, CASP9) pathways could be highly valuable in the diagnosis of neurodegenerative diseases and stroke. Interestingly, the Arg72Pro SNP in TP53, the gene encoding tumor suppressor p53, was recently revealed a biomarker of poor prognosis in stroke due to its ability to modulate neuronal apoptotic death. Search for new SNPs responsible for genetic variability to apoptosis will ensure the implementation of novel diagnostic and prognostic tools, as well as therapeutic strategies against neurological diseases.
Collapse
Affiliation(s)
- Angeles Almeida
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|