1
|
Bai Z, Gholipourshahraki T, Shrestha M, Hjelholt A, Hu S, Kjolby M, Rohde PD, Sørensen P. Evaluation of Bayesian Linear Regression derived gene set test methods. BMC Genomics 2024; 25:1236. [PMID: 39716056 DOI: 10.1186/s12864-024-11026-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/08/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Gene set tests can pinpoint genes and biological pathways that exert small to moderate effects on complex diseases like Type 2 Diabetes (T2D). By aggregating genetic markers based on biological information, these tests can enhance the statistical power needed to detect genetic associations. RESULTS Our goal was to develop a gene set test utilizing Bayesian Linear Regression (BLR) models, which account for both linkage disequilibrium (LD) and the complex genetic architectures intrinsic to diseases, thereby increasing the detection power of genetic associations. Through a series of simulation studies, we demonstrated how the efficacy of BLR derived gene set tests is influenced by several factors, including the proportion of causal markers, the size of gene sets, the percentage of genetic variance explained by the gene set, and the genetic architecture of the traits. By using KEGG pathways, eQTLs, and regulatory elements as different kinds of gene sets with T2D results, we also assessed the performance of gene set tests in explaining more about real phenotypes. CONCLUSIONS Comparing our method with other approaches, such as the gold standard MAGMA (Multi-marker Analysis of Genomic Annotation) approach, our BLR gene set test showed superior performance. Combining performance of our method in simulated and real phenotypes, this suggests that our BLR-based approach could more accurately identify genes and biological pathways underlying complex diseases.
Collapse
Affiliation(s)
- Zhonghao Bai
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark.
| | | | - Merina Shrestha
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Astrid Hjelholt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Sile Hu
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Palle Duun Rohde
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
2
|
Gholipourshahraki T, Bai Z, Shrestha M, Hjelholt A, Hu S, Kjolby M, Rohde PD, Sørensen P. Evaluation of Bayesian Linear Regression models for gene set prioritization in complex diseases. PLoS Genet 2024; 20:e1011463. [PMID: 39495786 PMCID: PMC11563439 DOI: 10.1371/journal.pgen.1011463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/14/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
Genome-wide association studies (GWAS) provide valuable insights into the genetic architecture of complex traits, yet interpreting their results remains challenging due to the polygenic nature of most traits. Gene set analysis offers a solution by aggregating genetic variants into biologically relevant pathways, enhancing the detection of coordinated effects across multiple genes. In this study, we present and evaluate a gene set prioritization approach utilizing Bayesian Linear Regression (BLR) models to uncover shared genetic components among different phenotypes and facilitate biological interpretation. Through extensive simulations and analyses of real traits, we demonstrate the efficacy of the BLR model in prioritizing pathways for complex traits. Simulation studies reveal insights into the model's performance under various scenarios, highlighting the impact of factors such as the number of causal genes, proportions of causal variants, heritability, and disease prevalence. Comparative analyses with MAGMA (Multi-marker Analysis of GenoMic Annotation) demonstrate BLR's superior performance, especially in highly overlapped gene sets. Application of both single-trait and multi-trait BLR models to real data, specifically GWAS summary data for type 2 diabetes (T2D) and related phenotypes, identifies significant associations with T2D-related pathways. Furthermore, comparison between single- and multi-trait BLR analyses highlights the superior performance of the multi-trait approach in identifying associated pathways, showcasing increased statistical power when analyzing multiple traits jointly. Additionally, enrichment analysis with integrated data from various public resources supports our results, confirming significant enrichment of diabetes-related genes within the top T2D pathways resulting from the multi-trait analysis. The BLR model's ability to handle diverse genomic features, perform regularization, conduct variable selection, and integrate information from multiple traits, genders, and ancestries demonstrates its utility in understanding the genetic architecture of complex traits. Our study provides insights into the potential of the BLR model to prioritize gene sets, offering a flexible framework applicable to various datasets. This model presents opportunities for advancing personalized medicine by exploring the genetic underpinnings of multifactorial traits.
Collapse
Affiliation(s)
| | - Zhonghao Bai
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Merina Shrestha
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Astrid Hjelholt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Sile Hu
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, United Kingdom
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Palle Duun Rohde
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Du W, Wang X, Zhang D, Zuo X. Genotype-Guided Model for Prediction of Tacrolimus Initial Dosing After Lung Transplantation. J Clin Pharmacol 2024; 64:719-727. [PMID: 38327217 DOI: 10.1002/jcph.2411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
The determination of the appropriate initial dose for tacrolimus is crucial in achieving the target concentration promptly and avoiding adverse effects and poor prognosis. However, the trial-and-error approach is still common practice. This study aimed to establish a prediction model for an initial dosing algorithm of tacrolimus in patients receiving a lung transplant. A total of 210 lung transplant recipients were enrolled, and 26 single nucleotide polymorphisms (SNP) from 18 genes that could potentially affect tacrolimus pharmacokinetics were genotyped. Associations between SNPs and tacrolimus concentration/dose ratio were analyzed. SNPs that remained significant in pharmacogenomic analysis were further combined with clinical factors to construct a prediction model for tacrolimus initial dose. The dose needed to reach steady state tacrolimus concentrations and achieve the target range was used to validate model prediction efficiency. Our final model consisted of 7 predictors-CYP3A5 rs776746, SLCO1B3 rs4149117, SLC2A2 rs1499821, NFATc4 rs1955915, alanine aminotransferase, direct bilirubin, and hematocrit-and explained 41.4% variance in the tacrolimus concentration/dose ratio. It achieved an area under the receiver operating characteristic curve of 0.804 (95% confidence interval, 0.746-0.861). The Hosmer-Lemeshow test yielded a nonsignificant P value of .790, suggesting good fit of the model. The predicted dose exhibited good correlation with the observed dose in the early postoperative period (r = 0.748, P less than .001). Our study provided a genotype-guided prediction model for tacrolimus initial dose, which may help to guide individualized dosing of tacrolimus in the lung transplant population in clinical practice.
Collapse
Affiliation(s)
- Wenwen Du
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoxing Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Dan Zhang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Xianbo Zuo
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
- Department of Dermatology, Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
4
|
Dallali H, Hechmi M, Morjane I, Elouej S, Jmel H, Ben Halima Y, Abid A, Bahlous A, Barakat A, Jamoussi H, Abdelhak S, Kefi R. Association of HNF1A gene variants and haplotypes with metabolic syndrome: a case-control study in the Tunisian population and a meta-analysis. Diabetol Metab Syndr 2022; 14:25. [PMID: 35109885 PMCID: PMC8812021 DOI: 10.1186/s13098-022-00794-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/15/2022] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Variants in the Hepatocyte Nuclear Factor 1 Alpha gene (HNF1A) are associated with lipoproteins levels and type 2 diabetes. In this study, we aimed to assess the association of HNF1A gene and haplotypes with the metabolic syndrome (MetS) and its components through an association study in the Tunisian population as well as by a meta-analysis. METHODS A total of 594 Tunisian individuals were genotyped for three variants (rs1169288, rs2464196 and rs735396) located in HNF1A gene using KASPar technology. Statistical analyses were performed with R software. The association was furthermore evaluated through a meta-analysis of our results with those obtained in a Moroccan population. RESULTS Our results showed no association between HNF1A variants and MetS in the Tunisian population. However, a significant association was observed between the variant rs735396 and a higher waist circumference. The stratified analysis according to the sex highlighted a significant association between the variant rs1169288 and high cholesterol levels only in women. Similarly, Haplotype analysis showed an association between the HNF1A minor haplotype and high total cholesterol mainly in women. Finally, our meta-analysis showed no association between HNF1A variants and MetS. CONCLUSIONS Our findings exclude the involvement of the three HNF1A variants rs1169288, rs2464196 and rs735396 in the susceptibility to MetS in our studied Tunisian population but emphasize the role of these variants in the cholesterol homeostasis with sex-specific differences, which may serve to rise clinical consideration to early statin therapy in women carrying these genetic variants.
Collapse
Affiliation(s)
- Hamza Dallali
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
| | - Meriem Hechmi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
- University of Carthage, National Institute of Applied Science and Technology, Tunis, Tunisia
| | - Imane Morjane
- Human Molecular Genetics Laboratory, Institut Pasteur du Maroc, Place Louis Pasteur, Casablanca, Morocco
| | - Sahar Elouej
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
| | - Haifa Jmel
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
| | - Yosra Ben Halima
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
| | - Abdelmajid Abid
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
- National Institute of Nutrition and Food Technology, 11 rue Jebel Lakhdar, Bab Saadoun, 1007, Tunis, Tunisia
| | - Afef Bahlous
- Central Laboratory of Medical Biology, Institut Pasteur in Tunis, 13 Place Pasteur, BP 74, 1002, Tunis, Tunisia
| | - Abdelhamid Barakat
- Human Molecular Genetics Laboratory, Institut Pasteur du Maroc, Place Louis Pasteur, Casablanca, Morocco
| | - Henda Jamoussi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
- National Institute of Nutrition and Food Technology, 11 rue Jebel Lakhdar, Bab Saadoun, 1007, Tunis, Tunisia
| | - Sonia Abdelhak
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia
- University of Tunis El Manar, 2092, El Manar I Tunis, Tunisia
| | - Rym Kefi
- Laboratory of Biomedical Genomics and Oncogenetics, Institut Pasteur in Tunis, BP 74, 13 Place Pasteur, Belvedere, 1002, Tunis, Tunisia.
- University of Tunis El Manar, 2092, El Manar I Tunis, Tunisia.
| |
Collapse
|
5
|
Bonetti S, Zusi C, Rinaldi E, Boselli ML, Csermely A, Malerba G, Trabetti E, Bonora E, Bonadonna R, Trombetta M. Role of monogenic diabetes genes on beta cell function in Italian patients with newly diagnosed type 2 diabetes. The Verona Newly Diagnosed Type 2 Diabetes Study (VNDS) 13. DIABETES & METABOLISM 2022; 48:101323. [DOI: 10.1016/j.diabet.2022.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/27/2021] [Accepted: 11/25/2021] [Indexed: 10/19/2022]
|
6
|
Shi D, Xie T, Deng J, Niu P, Wu W. CYP3A4 and GCK genetic polymorphisms are the risk factors of tacrolimus-induced new-onset diabetes after transplantation in renal transplant recipients. Eur J Clin Pharmacol 2018; 74:723-729. [DOI: 10.1007/s00228-018-2442-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 03/08/2018] [Indexed: 12/23/2022]
|
7
|
A label-free genetic biosensor for diabetes based on AuNPs decorated ITO with electrochemiluminescent signaling. Anal Chim Acta 2017; 982:62-71. [DOI: 10.1016/j.aca.2017.05.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 01/19/2023]
|
8
|
Billings LK, Jablonski KA, Warner AS, Cheng YC, McAteer JB, Tipton L, Shuldiner AR, Ehrmann DA, Manning AK, Dabelea D, Franks PW, Kahn SE, Pollin TI, Knowler WC, Altshuler D, Florez JC, for the Diabetes Prevention Program Research Group . Variation in Maturity-Onset Diabetes of the Young Genes Influence Response to Interventions for Diabetes Prevention. J Clin Endocrinol Metab 2017; 102:2678-2689. [PMID: 28453780 PMCID: PMC5546852 DOI: 10.1210/jc.2016-3429] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/21/2017] [Indexed: 11/19/2022]
Abstract
Context Variation in genes that cause maturity-onset diabetes of the young (MODY) has been associated with diabetes incidence and glycemic traits. Objectives This study aimed to determine whether genetic variation in MODY genes leads to differential responses to insulin-sensitizing interventions. Design and Setting This was a secondary analysis of a multicenter, randomized clinical trial, the Diabetes Prevention Program (DPP), involving 27 US academic institutions. We genotyped 22 missense and 221 common variants in the MODY-causing genes in the participants in the DPP. Participants and Interventions The study included 2806 genotyped DPP participants randomized to receive intensive lifestyle intervention (n = 935), metformin (n = 927), or placebo (n = 944). Main Outcome Measures Association of MODY genetic variants with diabetes incidence at a median of 3 years and measures of 1-year β-cell function, insulinogenic index, and oral disposition index. Analyses were stratified by treatment group for significant single-nucleotide polymorphism × treatment interaction (Pint < 0.05). Sequence kernel association tests examined the association between an aggregate of rare missense variants and insulinogenic traits. Results After 1 year, the minor allele of rs3212185 (HNF4A) was associated with improved β-cell function in the metformin and lifestyle groups but not the placebo group; the minor allele of rs6719578 (NEUROD1) was associated with an increase in insulin secretion in the metformin group but not in the placebo and lifestyle groups. Conclusions These results provide evidence that genetic variation among MODY genes may influence response to insulin-sensitizing interventions.
Collapse
Affiliation(s)
- Liana K. Billings
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois 60201
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
| | | | - A. Sofia Warner
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Yu-Chien Cheng
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois 60201
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Jarred B. McAteer
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Laura Tipton
- Biostatistics Center, George Washington University, Rockville, Maryland 20852
| | - Alan R. Shuldiner
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - David A. Ehrmann
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
| | - Alisa K. Manning
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, Colorado 80045
| | - Paul W. Franks
- Department of Clinical Sciences, Genetic, and Molecular Epidemiology Unit, Lund University Diabetes Center, Skåne University Hospital Malmö, SE-205 02 Malmö, Sweden
| | - Steven E. Kahn
- Division of Metabolism, Endocrinology, and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98195
| | - Toni I. Pollin
- Departments of Medicine (Division of Endocrinology, Diabetes, and Nutrition) and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - William C. Knowler
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85014
| | - David Altshuler
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
- Vertex Pharmaceuticals, Boston, Massachusetts 02210
| | - Jose C. Florez
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - for the Diabetes Prevention Program Research Group
- Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02114
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114
- Department of Medicine, NorthShore University HealthSystem, Evanston, Illinois 60201
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637
- Biostatistics Center, George Washington University, Rockville, Maryland 20852
- Department of Medicine, Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, Colorado 80045
- Department of Clinical Sciences, Genetic, and Molecular Epidemiology Unit, Lund University Diabetes Center, Skåne University Hospital Malmö, SE-205 02 Malmö, Sweden
- Division of Metabolism, Endocrinology, and Nutrition, VA Puget Sound Health Care System and University of Washington, Seattle, Washington 98195
- Departments of Medicine (Division of Endocrinology, Diabetes, and Nutrition) and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85014
- Vertex Pharmaceuticals, Boston, Massachusetts 02210
| |
Collapse
|
9
|
Piccolo P, Annunziata P, Soria LR, Attanasio S, Barbato A, Castello R, Carissimo A, Quagliata L, Terracciano LM, Brunetti-Pierri N. Down-regulation of hepatocyte nuclear factor-4α and defective zonation in livers expressing mutant Z α1-antitrypsin. Hepatology 2017; 66:124-135. [PMID: 28295475 DOI: 10.1002/hep.29160] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/09/2017] [Accepted: 03/10/2017] [Indexed: 12/19/2022]
Abstract
UNLABELLED α1 -Antitrypsin (AAT) deficiency is one of the most common genetic disorders and the liver disease due to the Z mutant of AAT (ATZ) is a prototype of conformational disorder due to protein misfolding with consequent aberrant intermolecular protein aggregation. In the present study, we found that livers of PiZ transgenic mice expressing human ATZ have altered expression of a network of hepatocyte transcriptional factors, including hepatocyte nuclear factor-4α, that is early down-regulated and induces a transcriptional repression of ATZ expression. Reduced hepatocyte nuclear factor-4α was associated with activation of β-catenin, which regulates liver zonation. Livers of PiZ mice and human patients with AAT deficiency were both found to have a severe perturbation of liver zonation. Functionally, PiZ mice showed a severe defect of ureagenesis, as shown by increased baseline ammonia, and reduced urea production and survival after an ammonia challenge. Down-regulation of hepatocyte nuclear factor-4α expression and defective zonation in livers have not been recognized so far as features of the liver disease caused by ATZ and are likely involved in metabolic disturbances and in the increased risk of hepatocellular carcinoma in patients with AAT deficiency. CONCLUSION The findings of this study are consistent with the concept that abnormal AAT protein conformation and intrahepatic accumulation have broad effects on metabolic liver functions. (Hepatology 2017;66:124-135).
Collapse
Affiliation(s)
- Pasquale Piccolo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | | | - Leandro R Soria
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Sergio Attanasio
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Anna Barbato
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Raffaele Castello
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | | | - Luca Quagliata
- Molecular Pathology Division, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Luigi M Terracciano
- Molecular Pathology Division, Institute of Pathology, University of Basel, Basel, Switzerland
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Department of Translational Medicine, Federico II University, Naples, Italy
| |
Collapse
|
10
|
Pharmacogenetics of posttransplant diabetes mellitus. THE PHARMACOGENOMICS JOURNAL 2017; 17:209-221. [DOI: 10.1038/tpj.2017.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/04/2016] [Accepted: 01/09/2017] [Indexed: 02/08/2023]
|
11
|
Morita K, Saruwatari J, Tanaka T, Oniki K, Kajiwara A, Miyazaki H, Yoshida A, Jinnouchi H, Nakagawa K. Common variants of HNF1A gene are associated with diabetic retinopathy and poor glycemic control in normal-weight Japanese subjects with type 2 diabetes mellitus. J Diabetes Complications 2017; 31:483-488. [PMID: 27445006 DOI: 10.1016/j.jdiacomp.2016.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 01/30/2023]
Abstract
AIM This study investigated the associations between the common hepatocyte nuclear factor-1A (HNF1A) variants and the risk of diabetic retinopathy (DR) in relation to the glycemic control and weight status. METHODS A retrospective longitudinal analysis was conducted among 354 Japanese patients with type 2 diabetes mellitus (T2DM) (mean follow-up duration: 5.8±2.5 years). The multivariable-adjusted hazard ratio (HR) for the cumulative incidence of DR was calculated using a Cox proportional hazard model. During the observation period, the longitudinal associations of the HNF1A diplotypes with the risk of DR and the clinical parameters were also analyzed using the generalized estimating equations approach. RESULTS The combination of risk variants, i.e., rs1169288-C, rs1183910-A and rs2464196-A, was defined as the H1 haplotype. The incidence of DR was higher in the H1/H1 diplotype cases than in the others (HR 2.75 vs. non-H1/non-H1; p=0.02). Only in normal-weight subjects, the risks of DR and poor glycemic control were higher in the H1/H1 diplotype cases than in the others [odds ratio 4.08 vs. non-H1/non-H1, p=0.02; odds ratio 3.03, p=0.01; respectively]. CONCLUSIONS This study demonstrated that the common HNF1A diplotype of three risk variants may be an independent risk factor for the development of DR resulting from poor glycemic control in normal-weight patients with T2DM. These results need to be replicated in larger and more varied study populations.
Collapse
Affiliation(s)
- Kazunori Morita
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Junji Saruwatari
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahiro Tanaka
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kentaro Oniki
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ayami Kajiwara
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroko Miyazaki
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Akira Yoshida
- Jinnouchi Clinic, Diabetes Care Center, Kumamoto, Japan
| | | | - Kazuko Nakagawa
- Division of Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan; Center for Clinical Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
12
|
Type 2 Diabetes Susceptibility in the Greek-Cypriot Population: Replication of Associations with TCF7L2, FTO, HHEX, SLC30A8 and IGF2BP2 Polymorphisms. Genes (Basel) 2017; 8:genes8010016. [PMID: 28067832 PMCID: PMC5295011 DOI: 10.3390/genes8010016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/13/2016] [Accepted: 12/30/2016] [Indexed: 01/17/2023] Open
Abstract
Type 2 diabetes (T2D) has been the subject of numerous genetic studies in recent years which revealed associations of the disease with a large number of susceptibility loci. We hereby initiate the evaluation of T2D susceptibility loci in the Greek-Cypriot population by performing a replication case-control study. One thousand and eighteen individuals (528 T2D patients, 490 controls) were genotyped at 21 T2D susceptibility loci, using the allelic discrimination method. Statistically significant associations of T2D with five of the tested single nucleotide polymorphisms (SNPs) (TCF7L2 rs7901695, FTO rs8050136, HHEX rs5015480, SLC30A8 rs13266634 and IGF2BP2 rs4402960) were observed in this study population. Furthermore, 14 of the tested SNPs had odds ratios (ORs) in the same direction as the previously published studies, suggesting that these variants can potentially be used in the Greek-Cypriot population for predictive testing of T2D. In conclusion, our findings expand the genetic assessment of T2D susceptibility loci and reconfirm five of the worldwide established loci in a distinct, relatively small, newly investigated population.
Collapse
|
13
|
Yang J, Jiang F, Guo H, Soniya T, Yan CX, Tian ZF, Shi BY. Studies of genetic variability of the hepatocyte nuclear factor-1α gene in an Indian maturity-onset diabetes of the young family. Cell Biosci 2016; 6:29. [PMID: 27148439 PMCID: PMC4855895 DOI: 10.1186/s13578-016-0095-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/19/2016] [Indexed: 11/28/2022] Open
Abstract
Maturity-onset diabetes of the young (MODY), one of the specific types of diabetes mellitus, is a monogenetic disorder characterized by an autosomal dominant (AD) inheritance and β-cell dysfunction. To study an Indian family with clinical diagnosis of MODY and detect the genetic mutations in the aspect of molecular mechanism, seven blood samples were obtained from the diabetic patients of this pedigree and genomic DNA was extracted from peripheral leukocytes. The exon1, exon2 and exon4 of hepatocyte nuclear factor-1α (HNF-1α) gene were amplified by polymerase chain reaction. Then the products were sequenced and compared with standard sequences on gene bank. As a result, two mutations were detected in exon1. That was CTC → CTG (Leu → Leu) in codon17 and ATC → CTC (Ile → Leu) in codon27. I27L was speculated to have a close relationship with the glycometabolism and the pathogenesis of diabetes mellitus together with the putative novel mutation existed in this Indian pedigree. Meanwhile, one mutation of GGG → GGC (Gly → Gly) in codon288 of exon4 was detected in the proband. No mutations were found in exon2 but a G → T base substitution in the intron4 region among all seven samples was detected. It may have some potential effects on the onset of diabetes in this family, but we do not have any evidence right now. Although it requires further investigation on the function of mutations found in the intron region, our research may provide some clue for this issue and it deserves more attention.
Collapse
Affiliation(s)
- Jing Yang
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Feng Jiang
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Hui Guo
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Thadimacca Soniya
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Chun-xia Yan
- />Department of Forensic Medicine, Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Zhu-fang Tian
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| | - Bing-yin Shi
- />Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, 710061 People’s Republic of China
| |
Collapse
|
14
|
Khan IA, Vattam KK, Jahan P, Hasan Q, Rao P. Importance of glucokinase -258G/A polymorphism in Asian Indians with post-transplant and type 2 diabetes mellitus. Intractable Rare Dis Res 2016; 5:25-30. [PMID: 26989645 PMCID: PMC4761580 DOI: 10.5582/irdr.2015.01040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and post-transplant diabetes mellitus (PTDM) are non-synonymous forms of diabetes. Glucokinase (GCK) plays a key role in glucose metabolism. The relationship between the GCK promoter and specific types of diabetes, such as PTDM and T2DM, in the Asian Indian population is unknown. We examined the occurrence of a specific GCK promoter variant (-258G/A) in patients with T2DM and PTDM. The case-control study enrolled 640 Asian Indian subjects, including controls (n = 250) and T2DM (n = 250), PTDM (n = 42), and non-post-transplant diabetes mellitus (non-PTDM) (n = 98) patients. Purified Deoxyribonucleic acid (DNA) was genotyped with the polymerase chain reaction (PCR) and restriction fragment length polymorphism (RFLP) analysis. The digested PCR products were analyzed on 12% polyacrylamide gels. The anthropometric, biochemical, and clinical details of each group were documented. GCK -258G/A alleles and genotypes were not associated with T2DM. However, among PTDM subjects, we detected a higher frequency of heterozygotes (52.4%) and a positive association with alleles/genotypes. The results suggest that the promoter region (-258G/A) of GCK plays an important role in PTDM in Asian Indians.
Collapse
Affiliation(s)
- Imran Ali Khan
- Department of Genetics and Molecular medicine, Kamineni Hospitals, Hyderabad, India
- Department of Genetics, Vasavi Medical and Research Centre, Khairathabad, Hyderabad, India
- Department of Genetics and Biotechnology, Osmania University, Tarnaka, Hyderabad, India
| | - Kiran Kumar Vattam
- Department of Genetics and Molecular medicine, Kamineni Hospitals, Hyderabad, India
| | - Parveen Jahan
- Department of Genetics and Biotechnology, Osmania University, Tarnaka, Hyderabad, India
| | - Qurratulain Hasan
- Department of Genetics and Molecular medicine, Kamineni Hospitals, Hyderabad, India
- Department of Genetics, Vasavi Medical and Research Centre, Khairathabad, Hyderabad, India
| | - Pragna Rao
- Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
- Address correspondence to: Dr. Pragna Rao, Department of Biochemistry, Kasturba Medical College, Manipal University, Manipal-576104, Karnataka, India. E-mail:
| |
Collapse
|
15
|
Liu HP, Zhao Q, Jin GZ, Qian YW, Gu YJ, Dong H, Lu XY, Cong WM, Wu MC. Unique genetic alterations and clinicopathological features of hepatocellular adenoma in Chinese population. Pathol Res Pract 2015; 211:918-24. [DOI: 10.1016/j.prp.2015.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 09/02/2015] [Indexed: 01/09/2023]
|
16
|
Three missense variants of metabolic syndrome-related genes are associated with alpha-1 antitrypsin levels. Nat Commun 2015; 6:7754. [PMID: 26174136 PMCID: PMC4518310 DOI: 10.1038/ncomms8754] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 06/06/2015] [Indexed: 01/18/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) encoded by SERPINA1 is an acute-phase inflammation marker, and AAT deficiency (AATD) is known as one of the common genetic disorders in European populations. However, no genetic determinants to AAT levels apart from the SERPINA gene clusters have been identified to date. Here we perform a genome-wide association study of serum AAT levels followed by a two-staged replication study recruiting a total of 9,359 Japanese community-dwelling population. Three missense variants of metabolic syndrome-related genes, namely, rs671 in ALDH2, rs1169288 in HNF1A and rs1260326 in GCKR, significantly associate with AAT levels (P≤1.5 × 10(-12)). Previous reports have shown the functional relevance of ALDH2 and HNF1A to AAT. We observe a significant interaction of rs671 and alcohol consumption on AAT levels. We confirm the association between AAT and rs2896268 in SERPINA1, which is independent of known causative variants of AATD. These findings would support various AAT functions including metabolic processes.
Collapse
|
17
|
Hesami O, Kasmaei HD, Matini F, Assarzadegan F, Mansouri B, Jabbehdari S. Relationship between intracerebral hemorrhage and diabetes mellitus: a case-control study. JOURNAL OF CLINICAL AND DIAGNOSTIC RESEARCH : JCDR 2015; 9:OC08-10. [PMID: 26023579 DOI: 10.7860/jcdr/2015/12226.3741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/26/2015] [Indexed: 01/14/2023]
Abstract
INTRODUCTION The role of diabetes mellitus in the pathogenesis of intracerebral hemorrhage (ICH) is controversial. Underlying comorbidities such as diabetes mellitus may increase the risk of intracerebral hemorrhage. In this study, we sought to assess the role of diabetes mellitus in the occurrence of intracerebral hemorrhage. MATERIALS AND METHODS In this case-control study, the prevalence of diabetes mellitus was evaluated in 120 patients presenting with intracerebral hemorrhage and in a control group of 135 patients with low back pain. All patients were treated at the Departments of Neurology and Neurosurgery, Shohadaye Tajrish University Hospital, Tehran, Iran between 2008 and 2012. T-test was applied for analysing the quantitative variables and chi-square and Fisher's exact tests were used to analyse qualitative variables. RESULTS The mean age was 67.5±12.7 y in patients with intracerebral hemorrhage and 70.5±12.6 y in the control group (p=0.201). Diabetes mellitus was found in 39 patients with intracerebral hemorrhage (33.1%) and 30 (22.2%) control subjects (p=0.054). The prevalence of diabetes mellitus in patients younger than 60 y was 7.4% in the control group and 27.8% in the case group (p=0.042). CONCLUSION In our study, no significant relationship was found between diabetes mellitus and intracerebral hemorrhage (except in patients younger than 60 years) and diabetes mellitus did not cause bleeding in certain brain areas. However, more studies are required on the correlation of diabetes mellitus and intracerebral hemorrhage in the same population to reach a definite conclusion.
Collapse
Affiliation(s)
- Omid Hesami
- Faculty, Department of Neurology, Imam Hosein Hospital, Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Hosein Delavar Kasmaei
- Faculty, Department of Neurology, Shohada Hospital, Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Fateme Matini
- Faculty, Department of Neurology, Shohada Hospital, Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Farhad Assarzadegan
- Faculty, Department of Neurology, Imam Hosein Hospital, Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Behnam Mansouri
- Faculty, Department of Neurology, Imam Hosein Hospital, Shahid Beheshti University of Medical Sciences , Tehran, Iran
| | - Sayena Jabbehdari
- Students' Research Committee, Faculty of Medicine, Shahid beheshti University of Medical Sciences , Tehran, Iran
| |
Collapse
|
18
|
Ali O, Cerjak D, Kent JW, James R, Blangero J, Carless MA, Zhang Y. An epigenetic map of age-associated autosomal loci in northern European families at high risk for the metabolic syndrome. Clin Epigenetics 2015; 7:12. [PMID: 25806089 PMCID: PMC4372177 DOI: 10.1186/s13148-015-0048-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The prevalence of chronic diseases such as cancer, type 2 diabetes, metabolic syndrome (MetS), and cardiovascular disease increases with age in all populations. Epigenetic features are hypothesized to play important roles in the pathophysiology of age-associated diseases, but a map of these markers is lacking. We searched for genome-wide age-associated methylation signatures in peripheral blood of individuals at high risks for MetS by profiling 485,000 CpG sites in 192 individuals of Northern European ancestry using the Illumina HM450 array. Subjects (ages 6-85 years) were part of seven extended families, and 73% of adults and 32% of children were overweight or obese. RESULTS We found 22,122 genome-wide significant age-associated CpG sites (P α=0.05 = 3.65 × 10(-7) after correction for multiple testing) of which 14,155 are positively associated with age while 7,967 are negatively associated. By applying a positional density-based clustering algorithm, we generated a map of epigenetic 'hot-spots' of age-associated genomic segments, which include 290 age-associated differentially methylated CpG clusters (aDMCs), of which 207 are positively associated with age. Gene/pathway enrichment analyses were performed on these clusters using FatiGO. Genes localized to both the positively (n = 241) and negatively (n = 16) age-associated clusters are significantly enriched in specific KEGG pathways and GO terms. The most significantly enriched pathways are the hedgehog signaling pathway (adjusted P = 3.96 × 10(-3)) and maturity-onset diabetes of the young (MODY) (adjusted P = 6.26 × 10(-3)) in the positive aDMCs and type I diabetes mellitus (adjusted P = 3.69 × 10(-7)) in the negative aDMCs. We also identified several epigenetic loci whose age-associated change rates differ between subjects diagnosed with MetS and those without. CONCLUSION We conclude that in a family cohort at high risk for MetS, age-associated epigenetic features enrich in biological pathways important for determining the fate of fat cells and for insulin production. We also observe that several genes known to be related to MetS show differential epigenetic response to age in individuals with and without MetS.
Collapse
Affiliation(s)
- Omar Ali
- />Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin USA
| | - Diana Cerjak
- />TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin USA
- />Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Wisconsin USA
| | - Jack W Kent
- />Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas USA
| | - Roland James
- />TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin USA
- />Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Wisconsin USA
| | - John Blangero
- />Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas USA
| | - Melanie A Carless
- />Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas USA
| | - Yi Zhang
- />TOPS Obesity and Metabolic Research Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin USA
- />Human and Molecular Genetics Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Wisconsin USA
| |
Collapse
|
19
|
Association of the glucokinase gene promoter polymorphism -30G > A (rs1799884) with gestational diabetes mellitus susceptibility: a case-control study and meta-analysis. Arch Gynecol Obstet 2015; 292:291-8. [PMID: 25633883 DOI: 10.1007/s00404-015-3635-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/22/2015] [Indexed: 12/23/2022]
Abstract
PURPOSE Several studies have examined the association between glucokinase (GCK)-30G > A polymorphism and gestational diabetes mellitus (GDM). However, the results are still controversial. We performed the case-control study to investigate whether GCK-30G > A polymorphism correlates with the susceptibility of GDM in Chinese populations, and then conducted a meta-analysis by combining the previous studies. METHODS We recruited 948 GDM patients and 975 controls from May 2011 to August 2013. All the subjects were genotyped using the PCR-based invader assay. The differences of allelic frequencies and genotype distributions between GDM patients and controls were investigated in case-control study. A systematic search of all relevant studies was conducted. The observational studies that were related to an association between the glucokinase (GCK)-30G > A polymorphism and GDM were identified. The association between the glucokinase (GCK)-30G > A polymorphism and GDM susceptibility was assessed using genetic models. RESULTS The case-control study showed that GCK-30G > A polymorphism was associated with the susceptibility of GDM in a Chinese population. Furthermore, other six previously reported studies were included to perform meta-analysis. The meta-analysis showed that GCK-30G > A polymorphism was associated with GDM in Caucasian and Asian. CONCLUSIONS This study suggested that GCK-30G > A polymorphism may be associated with the susceptibility of GDM in a Chinese population. The further meta-analysis provides additional evidence supporting the above result that the risk allele of the GCK-30G > A polymorphism may increase GDM risk.
Collapse
|
20
|
Muller YL, Piaggi P, Hoffman D, Huang K, Gene B, Kobes S, Thearle MS, Knowler WC, Hanson RL, Baier LJ, Bogardus C. Common genetic variation in the glucokinase gene (GCK) is associated with type 2 diabetes and rates of carbohydrate oxidation and energy expenditure. Diabetologia 2014; 57:1382-90. [PMID: 24728127 PMCID: PMC4052004 DOI: 10.1007/s00125-014-3234-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/14/2014] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Glucokinase (GCK) plays a role in glucose metabolism and glucose-stimulated insulin secretion. Rare mutations in GCK cause MODY. We investigated whether common variation (minor allele frequency ≥0.01) in GCK is associated with metabolic traits and type 2 diabetes. METHODS Four exonic single-nucleotide polymorphisms (SNPs) and three SNPs predicted to cause loss of promoter function were identified in whole-genome sequence data from 234 Pima Indians. These seven tag SNPs and rs4607517, a type 2 diabetes variant established in other studies, were analysed in 415 full-heritage non-diabetic Pima Indians characterised for metabolic traits, and 7,667 American Indians who had data on type 2 diabetes and BMI. RESULTS A novel 3' untranslated region (3'UTR) SNP, chr7:44184184-G/A, was associated with the rate of carbohydrate oxidation post-absorptively (β = 0.22 mg [kg estimated metabolic body size (EMBS)](-1) min(-1), p = 0.005) and during a hyperinsulinaemic-euglycaemic clamp (β = 0.24 mg [kg EMBS](-1) min(-1), p = 0.0002), the rate of carbohydrate oxidation in a respiratory chamber (β = 311 kJ/day, p = 0.03) and 24 h energy expenditure, which was attributable to the thermic effect of food (β = 520 kJ/day, p = 3.39 × 10(-6)). This 3'UTR SNP was also associated with diabetes (OR 1.36, 95% CI 1.11, 1.65, p = 0.002), where the A allele (allele frequency 0.05) was associated with a lower rate of carbohydrate oxidation, lower 24 h energy expenditure and higher risk for diabetes. In a Cox proportional hazards model, a rate of insulin-stimulated carbohydrate oxidation lower than the mean rate at baseline predicted a higher risk for developing diabetes than for those above the mean (hazard rate ratio 2.2, 95% CI 1.3, 3.6, p = 0.002). CONCLUSIONS/INTERPRETATION Common variation in GCK influences the rate of carbohydrate oxidation, 24 h energy expenditure and diabetes risk in Pima Indians.
Collapse
Affiliation(s)
- Yunhua L. Muller
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Duncan Hoffman
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Ke Huang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Brittany Gene
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Marie S. Thearle
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - William C. Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Robert L. Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Leslie J. Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| | - Clifton Bogardus
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, 445 North 5th street, Phoenix, AZ 85004 USA
| |
Collapse
|
21
|
Santiago JA, Potashkin JA. System-based approaches to decode the molecular links in Parkinson's disease and diabetes. Neurobiol Dis 2014; 72 Pt A:84-91. [PMID: 24718034 DOI: 10.1016/j.nbd.2014.03.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 03/24/2014] [Accepted: 03/28/2014] [Indexed: 12/17/2022] Open
Abstract
A growing body of evidence indicates an increased risk for developing Parkinson's disease (PD) among people with type 2 diabetes (T2DM). The relationship between the etiology and development of both chronic diseases is beginning to be uncovered and recent studies show that PD and T2DM share remarkably similar dysregulated pathways. It has been proposed that a cascade of events including mitochondrial dysfunction, impaired insulin signaling, and metabolic inflammation trigger neurodegeneration in T2DM models. Network-based approaches have elucidated a potential molecular framework linking both diseases. Further, transcriptional signatures that modulate the neurodegenerative phenotype in T2DM have been identified. Here we contextualize the current experimental approaches to dissect the mechanisms underlying the association between PD and T2DM and discuss the existing challenges toward the understanding of the coexistence of these devastating aging diseases.
Collapse
Affiliation(s)
- Jose A Santiago
- The Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Judith A Potashkin
- The Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
22
|
Bouchard-Mercier A, Rudkowska I, Lemieux S, Couture P, Vohl MC. An interaction effect between glucokinase gene variation and carbohydrate intakes modulates the plasma triglyceride response to a fish oil supplementation. GENES AND NUTRITION 2014; 9:395. [PMID: 24643341 DOI: 10.1007/s12263-014-0395-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/27/2014] [Indexed: 02/08/2023]
Abstract
A large inter-individual variability in the plasma triglyceride (TG) response to fish oil consumption has been observed. The objective was to investigate the gene-diet interaction effects between single-nucleotide polymorphisms (SNPs) within glucokinase (GCK) gene and dietary carbohydrate intakes (CHO) on the plasma TG response to a fish oil supplementation. Two hundred and eight participants were recruited in the greater Quebec City area. The participants completed a 6-week fish oil supplementation (5 g fish oil/day: 1.9-2.2 g EPA and 1.1 g DHA). Thirteen SNPs within GCK gene were genotyped using TAQMAN methodology. A gene-diet interaction effect on the plasma TG response was observed with rs741038 and CHO adjusted for age, sex and BMI (p = 0.008). In order to compare the plasma TG response between genotypes according to CHO, participants were divided according to median CHO. Homozygotes of the minor C allele of rs741038 with high CHO >48.59 % had a greater decrease in their plasma TG concentrations following the intake of fish oil (p < 0.05) than C/C homozygotes with low CHO and also than the other genotypes either with high or low CHO. The plasma TG response to a fish oil supplementation may be modulated by gene-diet interaction effects involving GCK gene and CHO.
Collapse
Affiliation(s)
- Annie Bouchard-Mercier
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd., Quebec, G1V 0A6, Canada
| | | | | | | | | |
Collapse
|
23
|
Hamdan AL, Kurban Z, Azar ST. Prevalence of phonatory symptoms in patients with type 2 diabetes mellitus. Acta Diabetol 2013; 50:731-6. [PMID: 22527095 DOI: 10.1007/s00592-012-0392-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 03/19/2012] [Indexed: 10/28/2022]
Abstract
To look at the prevalence of phonatory symptoms in patients with type 2 diabetes mellitus. The correlation between these symptoms with duration of the disease, glycemic control, and neuropathy will be described. A total of 105 consecutive patients diagnosed with type 2 diabetes mellitus by their primary endocrinologist were evaluated. A control group consisting of 33 healthy subjects was recruited for this study. Demographic data included: age, gender, allergy, smoking, duration of the disease, glycemic control, and presence or absence of neuropathy. Subjects were also asked about the presence or absence of the following symptoms: hoarseness, vocal tiring or fatigue, vocal straining, and aphonia or complete loss of voice. Patients were also asked to fill out the Voice Handicap Index 10. The mean age of patients with diabetes was 53.21 + 9.68 years with male-to-female ratio of 2/3. The most common phonatory symptoms were vocal tiring or fatigue and hoarseness (34.3 and 33.3 %). There was a significant difference in the prevalence of hoarseness and vocal straining (p value 0.045 and 0.015, respectively) compared to controls. There was a significant correlation between glycemic control, neuropathy, and hoarseness (p value 0.030 and 0.001, respectively). Vocal straining and aphonia also correlated significantly with the presence of neuropathy. Close to 16 % of diabetic patients had a VHI-10 above or equal to 7. Diabetic patients are more likely to have phonatory symptoms compared to controls, namely straining and hoarseness. One out of seven patients with diabetes has reported that phonatory symptoms had a significant impact on their quality of life. The presence of neuropathy and poor glycemic control should alert the treating physician to these vocal complaints.
Collapse
Affiliation(s)
- Abdul-Latif Hamdan
- Department of Otolaryngology and Head and Neck Surgery, American University of Beirut-Medical Center, New York, NY, USA
| | | | | |
Collapse
|
24
|
Large scale meta-analyses of fasting plasma glucose raising variants in GCK, GCKR, MTNR1B and G6PC2 and their impacts on type 2 diabetes mellitus risk. PLoS One 2013; 8:e67665. [PMID: 23840762 PMCID: PMC3695948 DOI: 10.1371/journal.pone.0067665] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 05/22/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The evidence that the variants GCK rs1799884, GCKR rs780094, MTNR1B rs10830963 and G6PC2 rs560887, which are related to fasting plasma glucose levels, increase the risk of type 2 diabetes mellitus (T2DM) is contradictory. We therefore performed a meta-analysis to derive a more precise estimation of the association between these polymorphisms and T2DM. METHODS All the publications examining the associations of these variants with risk of T2DM were retrieved from the MEDLINE and EMBASE databases. Using the data from the retrieved articles, we computed summary estimates of the associations of the four variants with T2DM risk. We also examined the studies for heterogeneity, as well as for bias of the publications. RESULTS A total of 113,025 T2DM patients and 199,997 controls from 38 articles were included in the meta-analysis. Overall, the pooled results indicated that GCK (rs1799884), GCKR (rs780094) and MTNR1B (rs10830963) were significantly associated with T2DM susceptibility (OR, 1.04; 95%CI, 1.01-1.08; OR, 1.08; 95%CI, 1.05-1.12 and OR, 1.05; 95%CI, 1.02-1.08, respectively). After stratification by ethnicity, significant associations for the GCK, MTNR1B and G6PC2 variants were detected only in Caucasians (OR, 1.09; 95%CI, 1.02-1.16; OR, 1.10; 95%CI, 1.08-1.13 and OR, 0.97; 95%CI, 0.95-0.99, respectively), but not in Asians (OR, 1.02, 95% CI 0.98-1.05; OR, 1.01; 95%CI, 0.98-1.04 and OR, 1.12; 95%CI, 0.91-1.32, respectively). CONCLUSIONS Our meta-analyses demonstrated that GCKR rs780094 variant confers high cross-ethnicity risk for the development of T2DM, while significant associations between GCK, MTNR1B and G6PC2 variants and T2DM risk are limited to Caucasians.
Collapse
|
25
|
Colclough K, Bellanne-Chantelot C, Saint-Martin C, Flanagan SE, Ellard S. Mutations in the genes encoding the transcription factors hepatocyte nuclear factor 1 alpha and 4 alpha in maturity-onset diabetes of the young and hyperinsulinemic hypoglycemia. Hum Mutat 2013; 34:669-85. [PMID: 23348805 DOI: 10.1002/humu.22279] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 01/08/2013] [Indexed: 12/16/2022]
Abstract
Maturity-onset diabetes of the young (MODY) is a monogenic disorder characterized by autosomal dominant inheritance of young-onset (typically <25 years), noninsulin-dependent diabetes due to defective insulin secretion. MODY is both clinically and genetically heterogeneous with mutations in at least 10 genes. Mutations in the HNF1A gene encoding hepatocyte nuclear factor-1 alpha are the most common cause of MODY in most adult populations studied. The number of different pathogenic HNF1A mutations totals 414 in 1,247 families. Mutations in the HNF4A gene encoding hepatocyte nuclear factor-4 alpha are a rarer cause of MODY with 103 different mutations reported in 173 families to date. Sensitivity to treatment with sulfonylurea tablets is a feature of both HNF1A and HNF4A mutations. The HNF4A MODY phenotype has been expanded by the reports of macrosomia in ∼50% of babies, and more rarely, neonatal hyperinsulinemic hypoglycemia. The identification of an HNF1A or HNF4A gene mutation has important implications for clinical management in diabetes and pregnancy, but MODY is significantly underdiagnosed. Current research is focused on identifying biomarkers and developing probability models to identify those patients most likely to have MODY, until next generation sequencing technology enables cost-effective gene analysis for all patients with young onset diabetes.
Collapse
Affiliation(s)
- Kevin Colclough
- Department of Molecular Genetics, Royal Devon & Exeter NHS Foundation Trust, Exeter, UK
| | | | | | | | | |
Collapse
|
26
|
Shared dysregulated pathways lead to Parkinson's disease and diabetes. Trends Mol Med 2013; 19:176-86. [PMID: 23375873 DOI: 10.1016/j.molmed.2013.01.002] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/12/2012] [Accepted: 01/05/2013] [Indexed: 12/11/2022]
|
27
|
Genetic polymorphism of glucokinase on the risk of type 2 diabetes and impaired glucose regulation: evidence based on 298,468 subjects. PLoS One 2013; 8:e55727. [PMID: 23441155 PMCID: PMC3575415 DOI: 10.1371/journal.pone.0055727] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 12/29/2012] [Indexed: 12/23/2022] Open
Abstract
Background Glucokinase (GCK) is the key glucose phosphorylation enzyme which has attracted considerable attention as a candidate gene for type 2 diabetes (T2D) based on its enzyme function as the first rate-limiting step in the glycolysis pathway and regulates glucose-stimulated insulin secretion. In the past decade, the relationship between GCK and T2D has been reported in various ethnic groups. To derive a more precise estimation of the relationship and the effect of factors that might modify the risk, we performed this meta-analysis. Methods Databases including Pubmed, EMBASE, Web of Science and China National Knowledge Infrastructure (CNKI) were searched to find relevant studies. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of association. Results A total of 24 articles involving 88, 229 cases and 210, 239 controls were included. An overall random-effects per-allele OR of 1.06 (95% CI: 1.03–1.09; P<10−4) was found for the GCK −30G>A polymorphism. Significant results were also observed using dominant or recessive genetic models. In the subgroup analyses by ethnicity, significant results were found in Caucasians; whereas no significant associations were found among Asians. In addition, we found that the −30G>A polymorphism is a risk factor associated with increased impaired glucose regulation susceptibility. Besides, −30G>A homozygous was found to be significantly associated with increased fasting plasma glucose level with weighted mean difference (WMD) of 0.15 (95%: 0.05–0.24, P = 0.001) compared with G/G genotype. Conclusions This meta-analysis demonstrated that the −30G>A polymorphism of GCK is a risk factor associated with increased T2D susceptibility, but these associations vary in different ethnic populations.
Collapse
|
28
|
Association of single nucleotide polymorphisms in TCF2 with type 2 diabetes susceptibility in a Han Chinese population. PLoS One 2012; 7:e52938. [PMID: 23300827 PMCID: PMC3534126 DOI: 10.1371/journal.pone.0052938] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 11/22/2012] [Indexed: 12/15/2022] Open
Abstract
Hepatocyte nuclear factor 1β (HNF1β), a transcription factor encoded by the transcription factor 2 gene (TCF2), plays a critical role in pancreatic cell formation and glucose homeostasis. It has been suggested that single nucleotide polymorphisms (SNPs) of TCF2 are associated with susceptibility to type 2 diabetes (T2D). However, published results are inconsistent and inclusive. To further investigate the role of these common variants, we examined the association of TCF2 polymorphisms with the risk of T2D in a Han population in northeastern China. We genotyped five SNPs in 624 T2D patients and 630 healthy controls by using a SNaPshot method, and evaluated the T2D risk conferred by individual SNPs and haplotypes. In the single-locus analysis, we found that rs752010, rs4430796 and rs7501939 showed allelic differences between T2D patients and healthy controls, with an OR of 1.26 (95% CI 1.08-1.51, P = 0.003), an OR of 1.23 (95% CI 1.06-1.55, P = 0.001) and an OR of 1.28 (95% CI 1.10-1.61, P = 0.001), respectively. Genotype association analysis of each locus also revealed that the homozygous carriers of the at-risk allele had a significant increased T2D risk compared to homozygous carriers of the other allele (OR 1.78, 95% CI 1.20-2.64 for rs752010; OR 1.82, 95% CI 1.24-2.67 for rs4430796; OR 1.95, 95% CI 1.31-2.90 for rs7501939), even after Bonferroni correction for multiple comparisons. Besides, the haplotype-based analysis demonstrated that AGT in block rs752010-rs4430796-rs7501939 was associated with about 30% increase in T2D risk (OR 1.31, 95% CI 1.09-1.57, P = 0.01). Our findings suggested that TCF2 variants may be involved in T2D risk in a Han population of northeastern China. Larger studies with ethnically diverse populations are warranted to confirm the results reported in this investigation.
Collapse
|
29
|
Hamdan AL, Jabbour J, Nassar J, Dahouk I, Azar ST. Vocal characteristics in patients with type 2 diabetes mellitus. Eur Arch Otorhinolaryngol 2012; 269:1489-95. [PMID: 22302159 DOI: 10.1007/s00405-012-1933-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 01/12/2012] [Indexed: 11/25/2022]
Abstract
The objective of this study is to report the vocal characteristics of patients with type 2 diabetes mellitus in relation to disease duration, glycemic control, and neuropathy. This is a prospective study. The setting is institutional setting. A total of 82 patients were recruited for this study, and a healthy control group matched according to age and gender was recruited. Subjects underwent acoustic analysis and perceptual evaluation using the GRABS classification where G stands for grading, R for roughness, A for asthenia, B for breathiness, and S for straining using a scale of 0–3 where o stands for normal and three for severe deviation from normal. There was no significant difference in any of the acoustic variables between diabetic patients and control. There was no significant difference in the mean score of any of the perceptual evaluation parameters between diabetic patients and control, despite the fact that the mean scores were all higher in the diseased group except for roughness. When looking at subgroups, we see that diabetic patients with poor glycemic control and with neuropathy had significantly higher mean score for the G overall grade of the voice compared to controls with P values of 0.005 and 0.009, respectively. What is also worth noting is that diabetic patients with poor glycemic control had more straining compared to controls, P value 0.043. Patients with type 2 diabetes mellitus and poor glycemic control or neuropathy have a significant difference in the grade of their voice compared to controls.
Collapse
Affiliation(s)
- Abdul-latif Hamdan
- Department of Otolaryngology, American University of Beirut Medical Center, Beirut, Lebanon
| | | | | | | | | |
Collapse
|
30
|
Genetic polymorphisms of the transcription factor NFATc4 and development of new-onset diabetes after transplantation in Hispanic kidney transplant recipients. Transplantation 2012; 93:325-30. [PMID: 22234350 DOI: 10.1097/tp.0b013e31823f7f26] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Transcription factors of the nuclear factor of activated T cells (NFAT) family regulate both immune activation and insulin production. Calcineurin inhibitors (CNIs) target NFAT activation. Hence, CNIs not only prevent organ transplant rejection but also contribute to the development of new-onset diabetes after transplantation (NODAT). Given individual variation in the susceptibility to NODAT, we hypothesized that polymorphisms in the cytoplasmic NFAT (NFATc)4 gene, which is expressed in pancreatic islets, may be associated with NODAT. Haplotype-tagging single-nucleotide polymorphisms (SNPs) of the NFATc4 gene were genotyped in Hispanic renal transplant patients. Cumulative incidences of NODAT were compared between recipients of different NFATc4 genotypes and haplotypes. The Cox proportional hazard model was used to examine risks for NODAT. Nongenetic and genetic characteristics were included in the multivariate risk model. The SNP (rs10141896) T allele was associated with a lower cumulative incidence of NODAT (P=0.02). This is a tagging SNP for one of the five dominant NFATc4 haplotypes, T-T-T-T-G, and CNI-treated recipients with this haplotype had a reduced adjusted risk for NODAT (hazard ratio: 0.45; 95% confidence interval: 0.19-1.01). Conversely, patients homozygous for the C-C-C-G-G haplotype were at an increased risk (hazard ratio: 2.13; 95% confidence interval: 1.01-4.46) for NODAT in subanalysis. Of the nongenetic factors, use of tacrolimus, sirolimus, and older age were associated with increased risk for NODAT. Polymorphisms in the NFATc4 gene may confer certain protection or predisposition for NODAT.
Collapse
|
31
|
Swen JJ, Baak-Pablo RF, Guchelaar HJ, van der Straaten T. Alternative methods to a TaqMan assay to detect a tri-allelic single nucleotide polymorphism rs757210 in the HNF1β gene. Clin Chem Lab Med 2011; 50:279-84. [PMID: 22022982 DOI: 10.1515/cclm.2011.758] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 10/03/2011] [Indexed: 11/15/2022]
Abstract
BACKGROUND Several studies report difficulties in genotyping HNF1β rs757210 using TaqMan probes. This is possibly due to the tri-allelic nature of this single nucleotide polymorphism (SNP). The aim of the present research was to develop alternative methods for genotyping rs757210. METHODS Pyrosequencing and high resolution melting analysis of small amplicons (HRM) were developed and tested in panels of type 2 diabetes mellitus patients (n=258) and healthy blood donors (n=183). Results were confirmed by Sanger sequencing. RESULTS With pyrosequencing, allele frequencies for the A, G and C allele of 0.42, 0.56, 0.02 and 0.37, 0.62, 0.01 were established in the panel of type 2 diabetes mellitus patients and healthy blood donors, respectively. Similar results were found using the more routinely available HRM method. Results for pyrosequencing and HRM were in 99.6% concordance. CONCLUSIONS Pyrosequencing and HRM can be used to genotype the tri-allelic SNP rs757210 in the HNF1β gene and have the advantage over the commercially available TaqMan analysis that they can determine the rare C-allele variant.
Collapse
Affiliation(s)
- Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden, University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
32
|
Association of Hepatocyte Nuclear Factor 4 Alpha Polymorphisms with Type 2 Diabetes With or Without Metabolic Syndrome in Malaysia. Biochem Genet 2011; 50:298-308. [DOI: 10.1007/s10528-011-9472-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 05/27/2011] [Indexed: 10/17/2022]
|
33
|
Bahíllo Curieses P, Hermoso López F, Garrote Molpeceres R, Zurita Muñoz O, Campos Barros A. [Fasting hyperglycaemia and polymorphism in glucokinase promoter (rs1799884)]. An Pediatr (Barc) 2011; 75:273-6. [PMID: 21697023 DOI: 10.1016/j.anpedi.2011.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/05/2011] [Accepted: 05/06/2011] [Indexed: 11/29/2022] Open
Abstract
Glucokinase is one of the most important regulators of fasting glucose levels. There are several mutations in the glucokinase gene (GCK) which are linked with monogenic diabetes. Recently, a polymorphism in its promoter has been described, which is associated with impaired fasting glucose levels. We present a 7 years and 7 months old boy with overweight and a familial background of diabetes in two previous generations. In the oral glucose tolerance test, he had impaired fasting glucose levels and after two hours, with a high insulin response. Laboratory abnormalities improved after weight loss, but he maintains a slight fasting hyperglycaemia. The molecular study of the most common monogenic diabetes forms, MODY subtypes 1, 2, and 3, was negative. The allelic variant G/A was however detected at the GCK promoter polymorphism rs1799884.
Collapse
Affiliation(s)
- P Bahíllo Curieses
- Departamento de Endocrinología Pediátrica, Hospital Clínico Universitario, Valladolid, España.
| | | | | | | | | |
Collapse
|
34
|
Handelsman Y, Mechanick JI, Blonde L, Grunberger G, Bloomgarden ZT, Bray GA, Dagogo-Jack S, Davidson JA, Einhorn D, Ganda O, Garber AJ, Hirsch IB, Horton ES, Ismail-Beigi F, Jellinger PS, Jones KL, Jovanovič L, Lebovitz H, Levy P, Moghissi ES, Orzeck EA, Vinik AI, Wyne KL, Hurley DL, Zangeneh F. American Association of Clinical Endocrinologists Medical Guidelines for Clinical Practice for developing a diabetes mellitus comprehensive care plan. Endocr Pract 2011; 17 Suppl 2:1-53. [PMID: 21474420 DOI: 10.4158/ep.17.s2.1] [Citation(s) in RCA: 292] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Mirasierra M, Fernández-Pérez A, Díaz-Prieto N, Vallejo M. Alx3-deficient mice exhibit decreased insulin in beta cells, altered glucose homeostasis and increased apoptosis in pancreatic islets. Diabetologia 2011; 54:403-14. [PMID: 21104068 DOI: 10.1007/s00125-010-1975-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 10/20/2010] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Homeodomain transcription factors play an important role in the regulation of pancreatic islet function. In previous studies we determined that aristaless-like homeobox 3 (ALX3) is produced in islet cells, binds to the promoter of the insulin gene and regulates its expression. The purpose of the present study was to investigate the functional role of ALX3 in pancreatic islets and its possible involvement in the regulation of glucose homeostasis in vivo. METHODS Alx3-knockout mice were used. Glucose and insulin tolerance tests were carried out, and serum insulin concentrations were determined. Isolated islets were used to test insulin secretion and gene expression. The pancreatic islets were also studied using both confocal and conventional microscopy. RESULTS ALX3 deficiency resulted in increased blood glucose levels and impaired glucose tolerance in the presence of normal serum insulin concentrations. Insulin, glucagon and glucokinase expression were reduced in Alx3-null pancreatic islets. Reduced insulin content was reflected by decreased insulin secretion from isolated islets. Alx3-deficient islets also showed increased apoptosis, and morphometric analyses indicated that they were, on average, of smaller size than islets from control mice. ALX3 deficiency resulted in reduced beta cell mass. Finally, mature Alx3-null mice developed age-dependent insulin resistance due to impaired peripheral insulin receptor signalling. CONCLUSIONS/INTERPRETATION ALX3 participates in the regulation of the expression of essential genes for the function of pancreatic islets, and its deficiency alters the regulation of glucose homeostasis in vivo. We suggest that ALX3 constitutes a potential candidate to consider in the aetiopathogenesis of diabetes mellitus.
Collapse
Affiliation(s)
- M Mirasierra
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas /Universidad Autónoma de Madrid, Calle Arturo Duperier 4, 28029 Madrid, Spain
| | | | | | | |
Collapse
|
36
|
Abstract
BACKGROUND
Type 2 diabetes (T2D) is a complex disorder that is affected by multiple genetic and environmental factors. Extensive efforts have been made to identify the disease-affecting genes to better understand the disease pathogenesis, find new targets for clinical therapy, and allow prediction of disease.
CONTENT
Our knowledge about the genes involved in disease pathogenesis has increased substantially in recent years, thanks to genomewide association studies and international collaborations joining efforts to collect the huge numbers of individuals needed to study complex diseases on a population level. We have summarized what we have learned so far about the genes that affect T2D risk and their functions. Although more than 40 loci associated with T2D or glycemic traits have been reported and reproduced, only a minor part of the genetic component of the disease has been explained, and the causative variants and affected genes are unknown for many of the loci.
SUMMARY
Great advances have recently occurred in our understanding of the genetics of T2D, but much remains to be learned about the disease etiology. The genetics of T2D has so far been driven by technology, and we now hope that next-generation sequencing will provide important information on rare variants with stronger effects. Even when variants are known, however, great effort will be required to discover how they affect disease risk.
Collapse
Affiliation(s)
- Emma Ahlqvist
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Tarunveer Singh Ahluwalia
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
37
|
Lopez AP, Foscaldi SA, Perez MS, Rodriguez M, Traversa M, Puchulu FM, Bergada I, Frechtel GD. HNF1 alpha gene coding regions mutations screening, in a Caucasian population clinically characterized as MODY from Argentina. Diabetes Res Clin Pract 2011; 91:208-12. [PMID: 21168233 DOI: 10.1016/j.diabres.2010.11.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 10/18/2010] [Accepted: 11/16/2010] [Indexed: 11/26/2022]
Abstract
INTRODUCTION There are at least six subtypes of Maturity Onset Diabetes of the Young (MODY) with distinctive genetic causes. MODY 3 is caused by mutations in HNF1A gene, an insulin transcription factor, so mutations in this gene are associated with impaired insulin secretion. MODY 3 prevalence differs according to the population analyzed, but it is one of the most frequent subtypes. Therefore, our aims in this work were to find mutations present in the HNF1A gene and provide information on their prevalence. MATERIAL AND METHODS Mutations screening was done in a group of 80 unrelated patients (average age 17.1 years) selected by clinical characterization of MODY, by SSCP electrophoresis followed by sequenciation. RESULTS We found eight mutations, of which six were novel and four sequence variants, which were all novel. Therefore the prevalence of MODY 3 in this group was 10%. Compared clinical data between the non-MODY 3 patients and the MODY 3 diagnosed patients did not show any significant difference. DISCUSSION Eight patients were diagnosed as MODY 3 and new data about the prevalence of that subtype is provided. Our results contribute to reveal novel mutations, providing new data about the prevalence of that subtype.
Collapse
Affiliation(s)
- Ariel Pablo Lopez
- Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, 1113 Buenos Aires, Argentina.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ley SH, Hegele RA, Harris SB, Mamakeesick M, Cao H, Connelly PW, Gittelsohn J, Retnakaran R, Zinman B, Hanley AJ. HNF1A G319S variant, active cigarette smoking and incident type 2 diabetes in Aboriginal Canadians: a population-based epidemiological study. BMC MEDICAL GENETICS 2011; 12:1. [PMID: 21208426 PMCID: PMC3022797 DOI: 10.1186/1471-2350-12-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 01/05/2011] [Indexed: 12/02/2022]
Abstract
Background In a recent report of large-scale association analysis, a type 2 diabetes susceptibility locus near HNF1A was identified in predominantly European descent populations. A population-specific G319S polymorphism in HNF1A was previously identified in Aboriginal Canadians who have a high prevalence of type 2 diabetes. We aimed to investigate the association of the HNF1A G319S polymorphism with incident type 2 diabetes and to assess whether clinical risk variables for type 2 diabetes influence the association in an Aboriginal population. Methods Of 606 participants who were free of diabetes at baseline in 1993-1995, 540 (89.1%) participated in 10-year follow-up assessments in 2003-2005. Fasting glucose and a 75-g oral glucose tolerance test were obtained to determine incident type 2 diabetes. Participants were genotyped for the HNF1A G319S polymorphism. Interviewers administered questionnaires on smoking behavior. Results The incidence rates of type 2 diabetes were 14.2% (55/388) in major allele homozygotes and 31.2% (29/93) in minor allele carriers (p < 0.001). The HNF1A G319S carrier status was associated with incident type 2 diabetes (odds ratio [OR] 3.78 [95% CI 2.13-6.69]) after adjustment for age, sex, hypertension, triglyceride, HDL cholesterol, and waist circumference. A statistical interaction was observed between HNF1A G319S and baseline active cigarette smoking on the development of type 2 diabetes with similar adjustment (p = 0.006). When participants were stratified by baseline smoking status, HNF1A G319S carriers who were active smokers had increased risk of developing diabetes (OR 6.91 [95% CI 3.38-14.12]), while the association was attenuated to non-significance among non-smokers (1.11 [0.40-3.08]). Conclusions The HNF1A G319S variant is associated with incident type 2 diabetes in Aboriginal Canadians. Furthermore, cigarette smoking appears to amplify incident diabetes risk in carriers of HNF1A G319S.
Collapse
Affiliation(s)
- Sylvia H Ley
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Nuli R, Mohemaiti P, Yimamu Y, Taxitiemuer A. Sequencing MODY1-6 genes in Uyghur Early-onset diabetes pedigree. Indian J Endocrinol Metab 2011; 15:60-61. [PMID: 21584173 PMCID: PMC3079876 DOI: 10.4103/2230-8210.77591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Rebiya Nuli
- Occupational and Environmental Health Department, School of Public Health, Xinjiang Medical University, Xinyi Road, 393, Urumqi. Xinjiang Province 830011, P.R. China
| | - Patamu Mohemaiti
- Occupational and Environmental Health Department, School of Public Health, Xinjiang Medical University, Xinyi Road, 393, Urumqi. Xinjiang Province 830011, P.R. China
| | - Yilihamujan Yimamu
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University. Urumqi. Xinjiang Province 830011, P.R. China
| | - Aierken Taxitiemuer
- Occupational and Environmental Health Department, School of Public Health, Xinjiang Medical University, Xinyi Road, 393, Urumqi. Xinjiang Province 830011, P.R. China
| |
Collapse
|
40
|
Klein K, Winter S, Turpeinen M, Schwab M, Zanger UM. Pathway-Targeted Pharmacogenomics of CYP1A2 in Human Liver. Front Pharmacol 2010; 1:129. [PMID: 21918647 PMCID: PMC3171976 DOI: 10.3389/fphar.2010.00129] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 09/28/2010] [Indexed: 01/08/2023] Open
Abstract
The human drug metabolizing cytochrome P450 (CYP) 1A2, is one of the major P450 isoforms contributing by about 5–20% to the hepatic P450 pool and catalyzing oxidative biotransformation of up to 10% of clinically relevant drugs including clozapine and caffeine. CYP1A2 activity is interindividually highly variable and although twin studies have suggested a high heritability, underlying genetic factors are still unknown. Here we adopted a pathway-oriented approach using a large human liver bank (n = 150) to elucidate whether variants in candidate genes of constitutive, ligand-inducible, and pathophysiological inhibitory regulatory pathways may explain different hepatic CYP1A2 phenotypes. Samples were phenotyped for phenacetin O-deethylase activity, and the expression of CYP1A2 protein and mRNA was determined. CYP1A2 expression and function was increased in smokers and decreased in patients with inflammation and cholestasis. Of 169 SNPs in 17 candidate genes including the CYP1A locus, 136 non-redundant SNPs with minor allele frequency >5% were analyzed by univariate and multivariate methods. A total of 13 strong significant associations were identified, of which 10 SNPs in the ARNT, AhRR, HNF1α, IL1β, SRC-1, and VDR genes showed consistent changes for at least two phenotypes by univariate analysis. Multivariate linear modeling indicated that the polymorphisms and non-genetic factors together explained 42, 38, and 33% of CYP1A2 variation at activity, protein and mRNA levels, respectively. In conclusion, we identified novel trans-associations between regulatory genes and hepatic CYP1A2 function and expression, but additional genetic factors must be assumed to explain the full extent of CYP1A2 heritability.
Collapse
Affiliation(s)
- Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, University of Tuebingen Stuttgart, Germany
| | | | | | | | | |
Collapse
|
41
|
Abstract
Three and a half decades after the clinical description of "Maturity Onset Diabetes of the Young" (MODY), and despite its low prevalence, important knowledge has been gathered concerning its genetic basis, molecular pathways, clinical phenotypes and pharmacogenetic issues. This knowledge has proved to be important not only for the attention of subjects carrying a mutation but also for the insight provided in Type 2 diabetes mellitus. In recent years, a shift from the term "MODY" to "monogenic diabetes" has taken place, the latter term being a better and more comprehensive descriptor. We stick to the "old" term because information on other types of monogenic diabetes and pregnancy is scarce. In this review we perform an overview of the entity, the prevalence rates reported in women with gestational diabetes mellitus and the specific impact of each type on pregnancy outcome.
Collapse
Affiliation(s)
- Cristina Colom
- Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain
| | | |
Collapse
|
42
|
Ahmed MH, Byrne CD. Ezetimibe as a potential treatment for non-alcoholic fatty liver disease: is the intestine a modulator of hepatic insulin sensitivity and hepatic fat accumulation? Drug Discov Today 2010; 15:590-5. [DOI: 10.1016/j.drudis.2010.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 05/30/2010] [Accepted: 06/14/2010] [Indexed: 01/12/2023]
|
43
|
The glucokinase gene promoter polymorphism -30G>A (rs1799884) is associated with fasting glucose in healthy pregnant women but not with gestational diabetes. Clin Chim Acta 2010; 411:892-3. [PMID: 20227404 DOI: 10.1016/j.cca.2010.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 03/08/2010] [Accepted: 03/08/2010] [Indexed: 11/23/2022]
|
44
|
Osbak KK, Colclough K, Saint-Martin C, Beer NL, Bellanné-Chantelot C, Ellard S, Gloyn AL. Update on mutations in glucokinase (GCK), which cause maturity-onset diabetes of the young, permanent neonatal diabetes, and hyperinsulinemic hypoglycemia. Hum Mutat 2010; 30:1512-26. [PMID: 19790256 DOI: 10.1002/humu.21110] [Citation(s) in RCA: 364] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glucokinase is a key regulatory enzyme in the pancreatic beta-cell. It plays a crucial role in the regulation of insulin secretion and has been termed the glucose sensor in pancreatic beta-cells. Given its central role in the regulation of insulin release it is understandable that mutations in the gene encoding glucokinase (GCK) can cause both hyper- and hypoglycemia. Heterozygous inactivating mutations in GCK cause maturity-onset diabetes of the young (MODY) subtype glucokinase (GCK), characterized by mild fasting hyperglycemia, which is present at birth but often only detected later in life during screening for other purposes. Homozygous inactivating GCK mutations result in a more severe phenotype presenting at birth as permanent neonatal diabetes mellitus (PNDM). A growing number of heterozygous activating GCK mutations that cause hypoglycemia have also been reported. A total of 620 mutations in the GCK gene have been described in a total of 1,441 families. There are no common mutations, and the mutations are distributed throughout the gene. The majority of activating mutations cluster in a discrete region of the protein termed the allosteric activator site. The identification of a GCK mutation in patients with both hyper- and hypoglycemia has implications for the clinical course and clinical management of their disorder.
Collapse
Affiliation(s)
- Kara K Osbak
- Diabetes Research Laboratories, Oxford Centre for Diabetes Endocrinology & Metabolism, University of Oxford, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
45
|
Wallerman O, Motallebipour M, Enroth S, Patra K, Bysani MSR, Komorowski J, Wadelius C. Molecular interactions between HNF4a, FOXA2 and GABP identified at regulatory DNA elements through ChIP-sequencing. Nucleic Acids Res 2010; 37:7498-508. [PMID: 19822575 PMCID: PMC2794179 DOI: 10.1093/nar/gkp823] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Gene expression is regulated by combinations of transcription factors, which can be mapped to regulatory elements on a genome-wide scale using ChIP experiments. In a previous ChIP-chip study of USF1 and USF2 we found evidence also of binding of GABP, FOXA2 and HNF4a within the enriched regions. Here, we have applied ChIP-seq for these transcription factors and identified 3064 peaks of enrichment for GABP, 7266 for FOXA2 and 18783 for HNF4a. Distal elements with USF2 signal was frequently bound also by HNF4a and FOXA2. GABP peaks were found at transcription start sites, whereas 94% of FOXA2 and 90% of HNF4a peaks were located at other positions. We developed a method to accurately define TFBS within peaks, and found the predicted sites to have an elevated conservation level compared to peak centers; however the majority of bindings were not evolutionary conserved. An interaction between HNF4a and GABP was seen at TSS, with one-third of the HNF4a positive promoters being bound also by GABP, and this interaction was verified by co-immunoprecipitations.
Collapse
Affiliation(s)
- Ola Wallerman
- Department of Genetics and Pathology, Rudbeck Laboratory, SE-751 85 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
46
|
Sookoian S, Gemma C, Pirola CJ. Influence of hepatocyte nuclear factor 4alpha (HNF4alpha) gene variants on the risk of type 2 diabetes: a meta-analysis in 49,577 individuals. Mol Genet Metab 2010; 99:80-9. [PMID: 19748811 DOI: 10.1016/j.ymgme.2009.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Revised: 08/14/2009] [Accepted: 08/14/2009] [Indexed: 12/19/2022]
Abstract
BACKGROUND The nuclear receptor hepatocyte nuclear factor 4alpha (HNF4alpha) contributes to the regulation of a large fraction of liver and pancreatic islet transcriptomes. AIM To evaluate the influence of HNF4alpha polymorphisms across the entire locus on the occurrence of type 2 diabetes (T2D) by means of a meta-analysis. METHODS We evaluated haplotype block structure of HNF4alpha variants owing to linkage disequilibrium (LD). From 1455 reports, we evaluated 21 observational studies. RESULTS Six haplotype blocks of LD were constructed with SNPs with r(2)>0.8; there were also 14 unlinked SNPs. Overall, we included 22,920 cases and 26.657 controls. Among 17 heterogeneous studies (21,881 cases and 24,915 controls), including 3 SNPs of P2 promoter region in block 1, we observed a significant association with T2D in fixed (OR 0.94, 95%CI: 0.905-0.975, p=0.001) and random (OR 0.988, 95%CI: 0.880-0.948, p=0.000012) model. Three homogeneous studies were evaluated in block 2 (2684 cases and 2059 controls), and a significant association with T2D was also observed: OR: 1.121, 95%CI 1.013-1.241, p=0.027. Three additional variants were associated with T2D: two intronic SNPs (rs4810424: OR: 1.080, 95%CI: 1.010-1.154, p<0.03 and rs3212183: OR: 0.843, 95%CI: 0.774-0.918, p<0.00009) and one missense variant (rs1800961: OR: 0.770, 95%CI: 0.595-0.995, p<0.05, 6562 cases and 6723 controls). CONCLUSIONS In addition to HNF4alpha variants in the promoter region, other SNPs may be involved on the occurrence of T2D.
Collapse
Affiliation(s)
- Silvia Sookoian
- Molecular Genetics and Biology of Complex Diseases Department, Institute of Medical Research A. Lanari, University of Buenos Aires--National Council of Scientific and Technological Research, Combatientes de Malvinas 3150, Buenos Aires (1427), Argentina
| | | | | |
Collapse
|
47
|
Corrales PJP, López Garrido MP, Rodríguez SA, Rubio LL, López Jiménez LM, Oliveira CL, Alfaro Martínez JJ, Lozano García JJ, López AH, Castillo RR, Martínez JE, Romero FB. Clinical differences between patients with MODY-3, MODY-2 and type 2 diabetes mellitus with I27L polymorphism in the HNF1α gene. ACTA ACUST UNITED AC 2010; 57:4-8. [DOI: 10.1016/s1575-0922(10)70002-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Accepted: 12/01/2009] [Indexed: 01/20/2023]
|
48
|
Abstract
Although the genetic causes of monogenic disorders have been successfully identified in the past, the success in dissecting the genetics of complex polygenic diseases has until now been limited. With the introduction of whole genome wide association studies (WGAS) in 2007, the picture has been dramatically changed. Today we know of about 20 genetic variants increasing the risk of type 2 diabetes (T2D). Most of them seem to influence the capacity of beta-cells to increase insulin secretion to meet the demands imposed by an increase in body weight and insulin resistance. This probably represents only the tip of the iceberg, and over the next few years refined tools will provide a more complete picture of the genetic complexity of T2D. This will not only include the current dissection of common variants increasing the susceptibility of the disease but also rare variants with stronger effects, copy number variations and epigenetic effects like DNA methylation and histone acetylation. For the first time, we can anticipate with some confidence that the genetics of a complex disease like T2D really can be dissected.
Collapse
Affiliation(s)
- L Groop
- Department of Clinical Sciences/Diabetes and Endocrinology, and Lund University Diabetes Centre, Lund University, University Hospital Malmoe, Sweden.
| | | |
Collapse
|
49
|
Abstract
Type 2 diabetes mellitus is a complex metabolic disease that is caused by insulin resistance and beta-cell dysfunction. Furthermore, type 2 diabetes has an evident genetic component and represents a polygenic disease. During the last decade, considerable progress was made in the identification of type 2 diabetes risk genes. This was crucially influenced by the development of affordable high-density single nucleotide polymorphism (SNP) arrays that prompted several successful genome-wide association scans in large case-control cohorts. Subsequent to the identification of type 2 diabetes risk SNPs, cohorts thoroughly phenotyped for prediabetic traits with elaborate in vivo methods allowed an initial characterization of the pathomechanisms of these SNPs. Although the underlying molecular mechanisms are still incompletely understood, a surprising result of these pathomechanistic investigations was that most of the risk SNPs affect beta-cell function. This favors a beta-cell-centric view on the genetics of type 2 diabetes. The aim of this review is to summarize the current knowledge about the type 2 diabetes risk genes and their variants' pathomechanisms.
Collapse
Affiliation(s)
- Harald Staiger
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, D-72076 Tübingen, Germany
| | | | | | | |
Collapse
|
50
|
Reiner AP, Gross MD, Carlson CS, Bielinski SJ, Lange LA, Fornage M, Jenny NS, Walston J, Tracy RP, Williams OD, Jacobs DR, Nickerson DA. Common coding variants of the HNF1A gene are associated with multiple cardiovascular risk phenotypes in community-based samples of younger and older European-American adults: the Coronary Artery Risk Development in Young Adults Study and The Cardiovascular Health Study. CIRCULATION. CARDIOVASCULAR GENETICS 2009; 2:244-54. [PMID: 20031592 PMCID: PMC2841292 DOI: 10.1161/circgenetics.108.839506] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The transcription factor hepatocyte nuclear factor (HNF)-1 alpha regulates the activity of a number of genes involved in innate immunity, blood coagulation, lipid and glucose transport and metabolism, and cellular detoxification. Common polymorphisms of the HNF-1 alpha gene (HNF1A) were recently associated with plasma C-reactive protein and gamma-glutamyl transferase concentration in middle-aged to older European Americans (EA). METHODS AND RESULTS We assessed whether common variants of HNF1A are associated with C-reactive protein, gamma-glutamyl transferase, and other atherosclerotic and metabolic risk factors, in the large, population-based Coronary Artery Risk Development in Young Adults Study of healthy young EA (n=2154) and African American (AA; n=2083) adults. The minor alleles of Ile27Leu (rs1169288) and Ser486Asn (rs2464196) were associated with 0.10 to 0.15 standard deviation units lower C-reactive protein and gamma-glutamyl transferase levels in EA. The same HNF1A coding variants were associated with higher low-density lipoprotein cholesterol, apolipoprotein B, creatinine, and fibrinogen in EA. We replicated the associations between HNF1A coding variants and C-reactive protein, fibrinogen, low-density lipoprotein cholesterol, and renal function in a second population-based sample of EA adults 65 years and older from the Cardiovascular Health Study. The HNF1A Ser486Asn and/or Ile27Leu variants were also associated with increased risk of subclinical coronary atherosclerosis in Coronary Artery Risk Development in Young Adults and with incident coronary heart disease in Cardiovascular Health Study. The Ile27Leu and Ser486Asn variants were 3-fold less common in AA than in EA. There was little evidence of association between HNF1A genotype and atherosclerosis-related phenotypes in AA. CONCLUSIONS Common polymorphisms of HNF1A seem to influence multiple phenotypes related to cardiovascular risk in the general population of younger and older EA adults.
Collapse
Affiliation(s)
- Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|