1
|
Imamura M, Maeda S. Perspectives on genetic studies of type 2 diabetes from the genome-wide association studies era to precision medicine. J Diabetes Investig 2024; 15:410-422. [PMID: 38259175 PMCID: PMC10981147 DOI: 10.1111/jdi.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Genome-wide association studies (GWAS) have facilitated a substantial and rapid increase in the number of confirmed genetic susceptibility variants for complex diseases. Approximately 700 variants predisposing individuals to the risk for type 2 diabetes have been identified through GWAS until 2023. From 2018 to 2022, hundreds of type 2 diabetes susceptibility loci with smaller effect sizes were identified through large-scale GWAS with sample sizes of 200,000 to >1 million. The clinical translation of genetic information for type 2 diabetes includes the development of novel therapeutics and risk predictions. Although drug discovery based on loci identified in GWAS remains challenging owing to the difficulty of functional annotation, global efforts have been made to identify novel biological mechanisms and therapeutic targets by applying multi-omics approaches or searching for disease-associated coding variants in isolated founder populations. Polygenic risk scores (PRSs), comprising up to millions of associated variants, can identify individuals with higher disease risk than those in the general population. In populations of European descent, PRSs constructed from base GWAS data with a sample size of approximately 450,000 have predicted the onset of diseases well. However, European GWAS-derived PRSs have limited predictive performance in non-European populations. The predictive accuracy of a PRS largely depends on the sample size of the base GWAS data. The results of GWAS meta-analyses for multi-ethnic groups as base GWAS data and cross-population polygenic prediction methodology have been applied to establish a universal PRS applicable to small isolated ethnic populations.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| |
Collapse
|
2
|
Teerawattanapong N, Srisawat L, Narkdontri T, Yenchitsomanus PT, Tangjittipokin W, Plengvidhya N. The effects of transcription factor 7-like 2 rs7903146 and paired box 4 rs2233580 variants associated with type 2 diabetes on the therapeutic efficacy of hypoglycemic agents. Heliyon 2024; 10:e27047. [PMID: 38439836 PMCID: PMC10909763 DOI: 10.1016/j.heliyon.2024.e27047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 12/11/2023] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
Aim This study aims to investigate the effects of the TCF7L2 rs7903146 and PAX4 rs2233580 (R192H) variants associated with T2D on the therapeutic efficacies of various HAs in patients with T2D after follow-up for 3 years. Methods A total of 526 patients who were followed up at the Diabetic Clinic of Siriraj Hospital during 2016-2019 were enrolled. The variants TCF7L2 rs7903146 and PAX4 rs2233580 (R192H) were genotyped using the RNase H2 enzyme-based amplification (rhAmp) technique and the associations between genotypes and glycemic control after treatments with different combinations HA were evaluated using Generalized Estimating Equations (GEE) analysis. Results Patients who carried TCF7L2 rs7903146C/T + T/T genotypes when they were treated with biguanide alone had significantly lower fasting plasma glucose (FPG) than those of the patients who carried the C/C genotype (p = 0.01). Patients who carried the PAX4 rs2233580 G/G genotype when they were treated with sulfonylurea alone had significantly lower FPG than those of the patients who carried G/A + A/A genotypes (p = 0.04). Conclusion Genotypes of TCF7L2 rs7903146 and PAX4 rs2233580 (R192H) variants associated with T2D influence the therapeutic responses to biguanide and sulfonylurea. Different genotypes of these two variants might distinctively affect the therapeutic effects of HAs. This finding provides evidence of pharmacogenetics in the treatment of diabetes.
Collapse
Affiliation(s)
- Nipaporn Teerawattanapong
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence for Diabetes and Obesity (SiCORE-DO), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Lanraphat Srisawat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence for Diabetes and Obesity (SiCORE-DO), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tassanee Narkdontri
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence for Diabetes and Obesity (SiCORE-DO), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-thai Yenchitsomanus
- Siriraj Center of Research Excellence for Diabetes and Obesity (SiCORE-DO), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Watip Tangjittipokin
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence for Diabetes and Obesity (SiCORE-DO), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nattachet Plengvidhya
- Siriraj Center of Research Excellence for Diabetes and Obesity (SiCORE-DO), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
3
|
Valvi D, Christiani DC, Coull B, Højlund K, Nielsen F, Audouze K, Su L, Weihe P, Grandjean P. Gene-environment interactions in the associations of PFAS exposure with insulin sensitivity and beta-cell function in a Faroese cohort followed from birth to adulthood. ENVIRONMENTAL RESEARCH 2023; 226:115600. [PMID: 36868448 PMCID: PMC10101920 DOI: 10.1016/j.envres.2023.115600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Exposure to perfluoroalkyl substances (PFAS) has been associated with changes in insulin sensitivity and pancreatic beta-cell function in humans. Genetic predisposition to diabetes may modify these associations; however, this hypothesis has not been yet studied. OBJECTIVES To evaluate genetic heterogeneity as a modifier in the PFAS association with insulin sensitivity and pancreatic beta-cell function, using a targeted gene-environment (GxE) approach. METHODS We studied 85 single-nucleotide polymorphisms (SNPs) associated with type 2 diabetes, in 665 Faroese adults born in 1986-1987. Perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) were measured in cord whole blood at birth and in participants' serum from age 28 years. We calculated the Matsuda-insulin sensitivity index (ISI) and the insulinogenic index (IGI) based on a 2 h-oral glucose tolerance test performed at age 28. Effect modification was evaluated in linear regression models adjusted for cross-product terms (PFAS*SNP) and important covariates. RESULTS Prenatal and adult PFOS exposures were significantly associated with decreased insulin sensitivity and increased beta-cell function. PFOA associations were in the same direction but attenuated compared to PFOS. A total of 58 SNPs were associated with at least one PFAS exposure variable and/or Matsuda-ISI or IGI in the Faroese population and were subsequently tested as modifiers in the PFAS-clinical outcome associations. Eighteen SNPs showed interaction p-values (PGxE) < 0.05 in at least one PFAS-clinical outcome association, five of which passed False Discovery Rate (FDR) correction (PGxE-FDR<0.20). SNPs for which we found stronger evidence for GxE interactions included ABCA1 rs3890182, FTO rs9939609, FTO rs3751812, PPARG rs170036314 and SLC12A3 rs2289116 and were more clearly shown to modify the PFAS associations with insulin sensitivity, rather than with beta-cell function. DISCUSSION Findings from this study suggest that PFAS-associated changes in insulin sensitivity could vary between individuals as a result of genetic predisposition and warrant replication in independent larger populations.
Collapse
Affiliation(s)
- Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Brent Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Flemming Nielsen
- Department of Public Health, Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Pal Weihe
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands; Centre of Health Science, Faculty of Health Sciences, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Philippe Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Public Health, Clinical Pharmacology, Pharmacy and Environmental Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
4
|
Salauddin A, Chakma K, Hasan MM, Akter F, Chowdhury NA, Chowdhury SR, Mannan A. Association between TCF7L2 polymorphism and type 2 diabetes mellitus susceptibility: a case-control study among the Bangladeshi population. Mol Biol Rep 2023; 50:609-619. [PMID: 36369331 DOI: 10.1007/s11033-022-08081-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Diabetes is a severe health burden for Bangladesh. Genetic polymorphism has been reported to be one of the major risk factors for diabetes in various studies. TCF7L2 (transcription factor 7 like 2) transcripts in the human β-cell have effects on β-cell survival, function, and Wnt signaling activation. This study aimed to evaluate the frequency and association of various polymorphisms namely TCF7L2 rs12255372 and rs7903146 among Bangladeshi patients with T2DM (Type 2 Diabetes Mellitus). METHODS This case-control study included 300 patients with T2DM and 234 healthy individuals from two health facilities in the Chattogram Division of Bangladesh. Anthropometric measurements were assessed using a self-reported, structured, eight-item questionnaire. The polymorphisms were identified by PCR-RFLP and sequencing method. RESULTS A strong association of T2DM with polymorphisms was observed, including rs12255372 (p = 0.0004) and rs7903146 (p = 0.005). It was observed that the risk genotype at rs12255372 was associated with age (p = 0.009), a family history of diabetes (p < 0.0001), and HbA1C (p < 0.0001). Furthermore, it was found that rs12255372 was substantially associated with hypertension (p = 0.03), eye problems (p = 0.01), and neurological abnormalities (p = 0.02). CONCLUSION This study postulates that TCF7L2 genetic polymorphism is associated with the risk of T2DM among the studied Bangladeshi population. The findings should be replicated through more studies with a large number of samples and in different populations.
Collapse
Affiliation(s)
- Asma Salauddin
- Department of Genetic Engineering & Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, 4331, Bangladesh.,Disease Biology and Molecular Epidemiology Research Group, Chattogram, Bangladesh
| | - Kallyan Chakma
- Department of Genetic Engineering & Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, 4331, Bangladesh.,Disease Biology and Molecular Epidemiology Research Group, Chattogram, Bangladesh
| | - Md Mahbub Hasan
- Department of Genetic Engineering & Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, 4331, Bangladesh.,Disease Biology and Molecular Epidemiology Research Group, Chattogram, Bangladesh
| | - Farhana Akter
- Department of Endocrinology, Chittagong Medical College, Chattogram, 4203, Bangladesh.,Disease Biology and Molecular Epidemiology Research Group, Chattogram, Bangladesh
| | | | | | - Adnan Mannan
- Department of Genetic Engineering & Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram, 4331, Bangladesh. .,Disease Biology and Molecular Epidemiology Research Group, Chattogram, Bangladesh.
| |
Collapse
|
5
|
Cui Y, Cruz M, Palatnik A, Olivier-Van Stichelen S. O-GlcNAc transferase contributes to sex-specific placental deregulation in gestational diabetes. Placenta 2023; 131:1-12. [PMID: 36442303 PMCID: PMC9839643 DOI: 10.1016/j.placenta.2022.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Gestational diabetes (GDM) is traditionally thought to emerge from placental endocrine dysregulations, but recent evidence suggests that fetal sex can also impact GDM development. Understanding the molecular mechanisms through which sex modulates placenta physiology can help identify novel molecular targets for future clinical care. Thus, we investigated the nutrient-sensing O-GlcNAc pathway as a potential mediator of sex-specific placenta dysfunction in GDM. METHODS Expression levels of O-GlcNAc enzymes were measured in male and female (n = 9+/gender) human placentas based on the maternal diagnosis of GDM. We then simulated the observed differences in both BeWo cells and human syncytiotrophoblasts primary cells (SCT) from male and female origins (n = 6/gender). RNA sequencing and targeted qPCR were performed to characterize the subsequent changes in the placenta transcriptome related to gestational diabetes. RESULTS O-GlcNAc transferase (OGT) expression was significantly reduced only in male placenta collected from mothers with GDM compared to healthy controls. Similar downregulation of OGT in trophoblast-like BeWo male cells demonstrated significant gene expression deregulations that overlapped with known GDM-related genes. Notably, placental growth hormone (GH) production was significantly elevated, while compensatory factors against GH-related insulin resistance were diminished. Inflammatory and immunologic factors with toxic effects on pancreatic β cell mass were also increased, altogether leaning toward a decompensatory diabetic profile. Similar changes in hormone expression were confirmed in male human primary SCTs transfected with siOGT. However, down-regulating OGT in female primary SCTs did not impact hormone production. CONCLUSION Our study demonstrated the significant deregulation of placental OGT levels in mothers with GDM carrying a male fetus. When simulated in vitro, such deregulation impacted hormonal production in BeWo trophoblast cells and primary SCTs purified from male placentas. Interestingly, female placentas were only modestly impacted by OGT downregulation, suggesting that the sex-specific presentation observed in gestational diabetes could be related to O-GlcNAc-mediated regulation of placental hormone production.
Collapse
Affiliation(s)
- Yiwen Cui
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Meredith Cruz
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Anna Palatnik
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Stephanie Olivier-Van Stichelen
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA; Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
6
|
Sánchez-Maldonado JM, Collado R, Cabrera-Serrano AJ, Ter Horst R, Gálvez-Montosa F, Robles-Fernández I, Arenas-Rodríguez V, Cano-Gutiérrez B, Bakker O, Bravo-Fernández MI, García-Verdejo FJ, López JAL, Olivares-Ruiz J, López-Nevot MÁ, Fernández-Puerta L, Cózar-Olmo JM, Li Y, Netea MG, Jurado M, Lorente JA, Sánchez-Rovira P, Álvarez-Cubero MJ, Sainz J. Type 2 Diabetes-Related Variants Influence the Risk of Developing Prostate Cancer: A Population-Based Case-Control Study and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14102376. [PMID: 35625981 PMCID: PMC9139180 DOI: 10.3390/cancers14102376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 02/06/2023] Open
Abstract
In this study, we have evaluated whether 57 genome-wide association studies (GWAS)-identified common variants for type 2 diabetes (T2D) influence the risk of developing prostate cancer (PCa) in a population of 304 Caucasian PCa patients and 686 controls. The association of selected single nucleotide polymorphisms (SNPs) with the risk of PCa was validated through meta-analysis of our data with those from the UKBiobank and FinnGen cohorts, but also previously published genetic studies. We also evaluated whether T2D SNPs associated with PCa risk could influence host immune responses by analysing their correlation with absolute numbers of 91 blood-derived cell populations and circulating levels of 103 immunological proteins and 7 steroid hormones. We also investigated the correlation of the most interesting SNPs with cytokine levels after in vitro stimulation of whole blood, peripheral mononuclear cells (PBMCs), and monocyte-derived macrophages with LPS, PHA, Pam3Cys, and Staphylococcus Aureus. The meta-analysis of our data with those from six large cohorts confirmed that each copy of the FTOrs9939609A, HNF1Brs7501939T, HNF1Brs757210T, HNF1Brs4430796G, and JAZF1rs10486567A alleles significantly decreased risk of developing PCa (p = 3.70 × 10-5, p = 9.39 × 10-54, p = 5.04 × 10-54, p = 1.19 × 10-71, and p = 1.66 × 10-18, respectively). Although it was not statistically significant after correction for multiple testing, we also found that the NOTCH2rs10923931T and RBMS1rs7593730 SNPs associated with the risk of developing PCa (p = 8.49 × 10-4 and 0.004). Interestingly, we found that the protective effect attributed to the HFN1B locus could be mediated by the SULT1A1 protein (p = 0.00030), an arylsulfotransferase that catalyzes the sulfate conjugation of many hormones, neurotransmitters, drugs, and xenobiotic compounds. In addition to these results, eQTL analysis revealed that the HNF1Brs7501939, HNF1Brs757210, HNF1Brs4430796, NOTCH2rs10923931, and RBMS1rs7593730 SNPs influence the risk of PCa through the modulation of mRNA levels of their respective genes in whole blood and/or liver. These results confirm that functional TD2-related variants influence the risk of developing PCa, but also highlight the need of additional experiments to validate our functional results in a tumoral tissue context.
Collapse
Affiliation(s)
- José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
| | - Ricardo Collado
- Medical Oncology Department, Hospital de San Pedro Alcántara, 10003 Cáceres, Spain; (R.C.); (M.I.B.-F.); (J.O.-R.)
| | - Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
| | - Fernando Gálvez-Montosa
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - Inmaculada Robles-Fernández
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
| | - Verónica Arenas-Rodríguez
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Blanca Cano-Gutiérrez
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Olivier Bakker
- Department of Genetics, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | | | - Francisco José García-Verdejo
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - José Antonio López López
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - Jesús Olivares-Ruiz
- Medical Oncology Department, Hospital de San Pedro Alcántara, 10003 Cáceres, Spain; (R.C.); (M.I.B.-F.); (J.O.-R.)
| | | | | | | | - Yang Li
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Manuel Jurado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
- Department of Medicine, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Jose Antonio Lorente
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Department of Legal Medicine, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Pedro Sánchez-Rovira
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.-M.); (F.J.G.-V.); (J.A.L.L.); (P.S.-R.)
| | - María Jesús Álvarez-Cubero
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (J.M.S.-M.); (A.J.C.-S.); (I.R.-F.); (V.A.-R.); (M.J.); (J.A.L.); (M.J.Á.-C.)
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain;
- Instituto de Investigación Biosanataria IBs. Granada, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Correspondence: ; Tel.: +34-95871-5500 (ext. 126); Fax: +34-9-5863-7071
| |
Collapse
|
7
|
Menshikov M, Zubkova E, Stafeev I, Parfyonova Y. Autophagy, Mesenchymal Stem Cell Differentiation, and Secretion. Biomedicines 2021; 9:biomedicines9091178. [PMID: 34572364 PMCID: PMC8467641 DOI: 10.3390/biomedicines9091178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent cells capable to differentiate into adipogenic, osteogenic, and chondrogenic directions, possessing immunomodulatory activity and a capability to stimulate angiogenesis. A scope of these features and capabilities makes MSC a significant factor of tissue homeostasis and repair. Among factors determining the fate of MSC, a prominent place belongs to autophagy, which is activated under different conditions including cell starvation, inflammation, oxidative stress, and some others. In addition to supporting cell homeostasis by elimination of protein aggregates, and non-functional and damaged proteins, autophagy is a necessary factor of change in cell phenotype on the process of cell differentiation. In present review, some mechanisms providing participation of autophagy in cell differentiation are discussed
Collapse
|
8
|
Sarah EH, El Omri N, Ibrahimi A, El Jaoudi R. Metabolic and genetic studies of glimepiride and metformin and their association with type 2 diabetes. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
9
|
Sex-specific associations of TCF7L2 variants with fasting glucose, type 2 diabetes and coronary heart disease among Turkish adults. Anatol J Cardiol 2020; 24:326-333. [PMID: 33122478 PMCID: PMC7724383 DOI: 10.14744/anatoljcardiol.2020.57736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Objective: TCF7L2 is a repressor and transactivator of genes, and its variants are strongly associated with diabetes. This study aimed to evaluate the sex-specific relationship between the most common TCF7L2 gene variants (-98368G>T, rs12255372 and -47833C>T, rs7903146) with diabetes and coronary heart disease in Turkish Adult Risk Factor (TARF) Study. Methods: Single nucleotide variants (SNVs) have been genotyped using the TaqMan allelic discrimination assays in 2,024 (51.3% in women, age: 55±11.8) Turkish adults participating in the TARF study. Statistical analyses were used to investigate the association of genotypes with clinical and biochemical measurements. Results: Among the TARF study participants, 11.7%, 24.3%, 14.1%, and 38.3% had diabetes, hypertension, coronary heart disease (CHD), and obesity, respectively. The frequencies of T allele for -47833C>T and -98368G>T in Turkish adults were determined to be 0.35 and 0.33, respectively. -47833C>T was significantly associated with higher fasting glucose concentrations in all participants, especially in men. Both SNVs were significantly associated with diabetes and CHD in all participants (p<0.05). When study population was stratified according to sex, -98368G>T was associated with diabetes in women (p=0.041) and -47833C>T was associated with diabetes and CHD in men (p=0.018 and p=0.032, respectively). Also, both SNVs and the diplotypes of common haplotype (H1) remained strongly associated with type 2 diabetes after risk factors were adjusted (p<0.05). Conclusion: T allele homozygosity of two SNVs as well as the diplotype H1-/H1- reflects risk of diabetes primarily in men. Enhanced CHD risk is determined by the presence of diplotype H1-/H1- among nondiabetic participants.
Collapse
|
10
|
Kaur N, Bhatti GK, Kaur S, Bhadada SK, Singh S, Bhatti JS. Transcription factor 7-like 2 gene, rs12255372 (G/T) variant and susceptibility to type 2 diabetes mellitus in North Indians. GENE REPORTS 2020; 19:100595. [DOI: 10.1016/j.genrep.2020.100595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
11
|
Sirdah MM, Reading NS. Genetic predisposition in type 2 diabetes: A promising approach toward a personalized management of diabetes. Clin Genet 2020; 98:525-547. [PMID: 32385895 DOI: 10.1111/cge.13772] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus, also known simply as diabetes, has been described as a chronic and complex endocrine metabolic disorder that is a leading cause of death across the globe. It is considered a key public health problem worldwide and one of four important non-communicable diseases prioritized for intervention through world health campaigns by various international foundations. Among its four categories, Type 2 diabetes (T2D) is the commonest form of diabetes accounting for over 90% of worldwide cases. Unlike monogenic inherited disorders that are passed on in a simple pattern, T2D is a multifactorial disease with a complex etiology, where a mixture of genetic and environmental factors are strong candidates for the development of the clinical condition and pathology. The genetic factors are believed to be key predisposing determinants in individual susceptibility to T2D. Therefore, identifying the predisposing genetic variants could be a crucial step in T2D management as it may ameliorate the clinical condition and preclude complications. Through an understanding the unique genetic and environmental factors that influence the development of this chronic disease individuals can benefit from personalized approaches to treatment. We searched the literature published in three electronic databases: PubMed, Scopus and ISI Web of Science for the current status of T2D and its associated genetic risk variants and discus promising approaches toward a personalized management of this chronic, non-communicable disorder.
Collapse
Affiliation(s)
- Mahmoud M Sirdah
- Division of Hematology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA.,Biology Department, Al Azhar University-Gaza, Gaza, Palestine
| | - N Scott Reading
- Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah, USA.,Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
12
|
Khan SM, El Karte N, El Hajj Chehadeh S, Hassoun A, Afandi B, Tay GK, Alsafar H. Association between type 2 diabetes mellitus & TCF7L2 gene variants in the Emirati population: Genetics of diabetes in the United Arab Emirates. Am J Hum Biol 2020; 33:e23434. [PMID: 32445548 DOI: 10.1002/ajhb.23434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 04/22/2020] [Accepted: 05/04/2020] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) has a multifactorial etiology involving a complex interplay between genes and the environment. The prevalence of T2DM among the countries of the Gulf Corporation Council (GCC), including the United Arab Emirates (UAE), ranks among the top 15 in the world. A number of studies have shown an increase in T2DM risk for the "TT" genotype at the rs4506565 and rs12255372 Single Nucleotide Polymorphisms (SNP) of the TCF7L2 gene. However, the association between TCF7L2 and T2DM still needs to be investigated in the UAE population. Therefore, this study analyzed the potential associations with rs4506565 and rs12255372 in UAE subjects. METHODS For this case-control study, T2DM patients (n = 890) and healthy subjects (n = 686) were genotyped using a Taqman Real-Time PCR assay. Statistical analysis was performed with the resulting data using the R (version 3.3.1) and STATA (version 13) software packages. RESULTS The rs12255372 SNP was significantly associated with T2DM (OR = 1.16, 95% CI = 1.00-1.34; P = .042). However, no significant association was found for the rs4506565 SNP (P = .120). After gender stratification, a significant association was found for both SNPs in males (Prs4506565 = .009 and Prs12255372 = .021). Interestingly, we found the interaction between the SNP rs4506565 with gender alone (P = .032) and in conjunction with BMI and age (P = .036) confers associations with T2DM. CONCLUSIONS These findings suggest that the genetic variants of the TCF7L2 gene are associated with an increased susceptibility to T2DM, especially in Emirati males. Our study also highlights the impact of biological and environmental risk factors including age, BMI, and gender on the genetic susceptibility to T2DM.
Collapse
Affiliation(s)
- Saad M Khan
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nora El Karte
- Esipe Créteil, Ingénierie Spécialisée en Biomédical et Santé, Université Paris-Est Créteil, Créteil, France
| | - Sarah El Hajj Chehadeh
- Center of Biotechnology, Khalifa University of Science, Technology & Research, Abu Dhabi, United Arab Emirates
| | - Ahmed Hassoun
- Dubai Diabetes Centre, Dubai Health Authority, Dubai, United Arab Emirates
| | - Bachar Afandi
- Endocrine Diabetes Center, Tawam Hospital, SEHA, Al-Ain, United Arab Emirates
| | - Guan K Tay
- Center of Biotechnology, Khalifa University of Science, Technology & Research, Abu Dhabi, United Arab Emirates.,Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Nedlands, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Habiba Alsafar
- Center of Biotechnology, Khalifa University of Science, Technology & Research, Abu Dhabi, United Arab Emirates.,Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Collage of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Boucher P, Matz RL, Terrand J. atherosclerosis: gone with the Wnt? Atherosclerosis 2020; 301:15-22. [PMID: 32289618 DOI: 10.1016/j.atherosclerosis.2020.03.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/19/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis, a pathology affecting large and medium-sized arteries, is the major cause of cardiovascular morbidity/mortality in industrialized countries. During atherosclerosis, cells accumulate large amounts of cholesterol through the uptake of modified low-density lipoprotein particles to form foam cells. This accumulation forms the basis for the development of the disease and for a large spectrum of other diseases in various organs. Massive research efforts have yielded valuable information about the underlying molecular mechanisms of atherosclerosis. In particular, newer discoveries on the early stage of lesion formation, cholesterol accumulation, reverse cholesterol transport, and local inflammation in the vascular wall have opened unanticipated horizons of understanding and raised novel questions and therapeutic opportunities. In this review, we focus on Wnt signaling, which has received little attention so far, yet affects lysosomal function and signalling pathways that limit cholesterol accumulation. This occurs in different tissues and cell types, including smooth muscle cells, endothelial cells and macrophages in the arterial wall, and thus profoundly impacts on atherosclerotic disease development and progression.
Collapse
Affiliation(s)
- Philippe Boucher
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France.
| | - Rachel L Matz
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| | - Jérôme Terrand
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| |
Collapse
|
14
|
Nasykhova YA, Barbitoff YA, Serebryakova EA, Katserov DS, Glotov AS. Recent advances and perspectives in next generation sequencing application to the genetic research of type 2 diabetes. World J Diabetes 2019; 10:376-395. [PMID: 31363385 PMCID: PMC6656706 DOI: 10.4239/wjd.v10.i7.376] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/23/2019] [Accepted: 06/11/2019] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes (T2D) mellitus is a common complex disease that currently affects more than 400 million people worldwide and has become a global health problem. High-throughput sequencing technologies such as whole-genome and whole-exome sequencing approaches have provided numerous new insights into the molecular bases of T2D. Recent advances in the application of sequencing technologies to T2D research include, but are not limited to: (1) Fine mapping of causal rare and common genetic variants; (2) Identification of confident gene-level associations; (3) Identification of novel candidate genes by specific scoring approaches; (4) Interrogation of disease-relevant genes and pathways by transcriptional profiling and epigenome mapping techniques; and (5) Investigation of microbial community alterations in patients with T2D. In this work we review these advances in application of next-generation sequencing methods for elucidation of T2D pathogenesis, as well as progress and challenges in implementation of this new knowledge about T2D genetics in diagnosis, prevention, and treatment of the disease.
Collapse
Affiliation(s)
- Yulia A Nasykhova
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
| | - Yury A Barbitoff
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Bioinformatics Institute, St. Petersburg 194021, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Elena A Serebryakova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
- Department of Genetics, City Hospital No. 40, St. Petersburg 197706, Russia
| | - Dmitry S Katserov
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad 236016, Russia
| | - Andrey S Glotov
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
- Department of Genetics, City Hospital No. 40, St. Petersburg 197706, Russia
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad 236016, Russia
| |
Collapse
|
15
|
Geoghegan G, Simcox J, Seldin MM, Parnell TJ, Stubben C, Just S, Begaye L, Lusis AJ, Villanueva CJ. Targeted deletion of Tcf7l2 in adipocytes promotes adipocyte hypertrophy and impaired glucose metabolism. Mol Metab 2019; 24:44-63. [PMID: 30948248 PMCID: PMC6531814 DOI: 10.1016/j.molmet.2019.03.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/02/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Activation of the Wnt-signaling pathway is known to inhibit differentiation in adipocytes. However, there is a gap in our understanding of the transcriptional network regulated by components of the Wnt-signaling pathway during adipogenesis and in adipocytes during postnatal life. The key intracellular effectors of the Wnt-signaling pathway occur through TCF transcription factors such as TCF7L2 (transcription factor-7-like 2). Several genetic variants in proximity to TCF7L2 have been linked to type 2 diabetes through genome-wide association studies in various human populations. Our work aims to functionally characterize the adipocyte specific gene program regulated by TCF7L2 and understand how this program regulates metabolism. METHODS We generated Tcf7l2F/F mice and assessed TCF7L2 function in isolated adipocytes and adipose specific knockout mice. ChIP-sequencing and RNA-sequencing was performed on the isolated adipocytes with control and TCF7L2 knockout cells. Adipose specific TCF7L2 knockout mice were challenged with high fat diet and assessed for body weight, glucose tolerance, and lipolysis. RESULTS Here we report that TCF7L2 regulates adipocyte size, endocrine function, and glucose metabolism. Tcf7l2 is highly expressed in white adipose tissue, and its expression is suppressed in genetic and diet-induced models of obesity. Genome-wide distribution of TCF7L2 binding and gene expression analysis in adipocytes suggests that TCF7L2 directly regulates genes implicated in cellular metabolism and cell cycle control. When challenged with a high-fat diet, conditional deletion of TCF7L2 in adipocytes led to impaired glucose tolerance, impaired insulin sensitivity, promoted weight gain, and increased adipose tissue mass. This was accompanied by reduced expression of triglyceride hydrolase, reduced fasting-induced free fatty acid release, and adipocyte hypertrophy in subcutaneous adipose tissue. CONCLUSIONS Together our studies support that TCF7L2 is a central transcriptional regulator of the adipocyte metabolic program by directly regulating the expression of genes involved in lipid and glucose metabolism.
Collapse
Affiliation(s)
- Gisela Geoghegan
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Judith Simcox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Marcus M Seldin
- Department of Human Genetics/Medicine, University of California, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Timothy J Parnell
- Bioinformatics Shared Resources, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Chris Stubben
- Bioinformatics Shared Resources, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Steven Just
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Lori Begaye
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Aldons J Lusis
- Department of Human Genetics/Medicine, University of California, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Claudio J Villanueva
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
16
|
Kalantari S, Sharafshah A, Keshavarz P, Davoudi A, Habibipour R. Single and multi-locus association study of TCF7L2 gene variants with susceptibility to type 2 diabetes mellitus in an Iranian population. Gene 2019; 696:88-94. [PMID: 30776466 DOI: 10.1016/j.gene.2019.01.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/04/2019] [Accepted: 01/22/2019] [Indexed: 01/03/2023]
Abstract
Prior studies indicated that some of transcription factor 7-like 2 (TCF7L2) gene variants such as rs7903146, rs12255372 and rs11255372 are constantly associated with Type 2 diabetes mellitus (T2DM) in various populations and ethnic groups. The purpose of this study was to assess the association between TCF7L2 variants (rs7903146, rs11196205, and rs11255372) and T2DM by TaqMan assay. Statistical analysis was performed through SNPAlyze and SPSS. Significant associations of rs7903146 (P = 1.9 × 10-7), and rs11255372 (P = 2.98 × 10-10) both under a dominant model were found. Based on allele frequency, there was a significant difference between the two study groups at rs7903146 and rs12255372 variants (P = 6.8 × 10-10, and P = 9.3 × 10-11, respectively). Two haplotypes including Hap-1 (C-G-G) and Hap-2 (T-G-T) indicated a significant difference between the two study groups (P = 1.174 × 10-9and P = 7.432 × 109 respectively). In conclusion, rs7903146 and rs12255372 were significantly associated with T2DM in the specified Northern Iranian population.
Collapse
Affiliation(s)
- Saeed Kalantari
- Department of Endocrinology, Guilan University of Medical Sciences, Rasht, Iran
| | - Alireza Sharafshah
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvaneh Keshavarz
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Arash Davoudi
- Division of Cytogenetic, Medical Genetic Laboratory of Dr. Keshavarz, Rasht, Iran
| | - Razie Habibipour
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
17
|
Overview of genomics and post-genomics research on type 2 diabetes mellitus: Future perspectives and a framework for further studies. J Biosci 2019. [DOI: 10.1007/s12038-018-9818-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
18
|
Srinivasan S, Kaur V, Chamarthi B, Littleton KR, Chen L, Manning AK, Merino J, Thomas MK, Hudson M, Goldfine A, Florez JC. TCF7L2 Genetic Variation Augments Incretin Resistance and Influences Response to a Sulfonylurea and Metformin: The Study to Understand the Genetics of the Acute Response to Metformin and Glipizide in Humans (SUGAR-MGH). Diabetes Care 2018; 41:554-561. [PMID: 29326107 PMCID: PMC5829963 DOI: 10.2337/dc17-1386] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/07/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The rs7903146 T allele in transcription factor 7 like 2 (TCF7L2) is strongly associated with type 2 diabetes (T2D), but the mechanisms for increased risk remain unclear. We evaluated the physiologic and hormonal effects of TCF7L2 genotype before and after interventions that influence glucose physiology. RESEARCH DESIGN AND METHODS We genotyped rs7903146 in 608 individuals without diabetes and recorded biochemical data before and after 1) one dose of glipizide (5 mg) on visit 1 and 2) a 75-g oral glucose tolerance test (OGTT) performed after administration of metformin 500 mg twice daily over 2 days. Incretin levels were measured in 150 of the 608 participants. RESULTS TT risk-allele homozygotes had 1.6 mg/dL higher baseline fasting glucose levels and 2.5 pg/mL lower glucagon levels per T allele than carriers of other genotypes at baseline. In a subset of participants, the T allele was associated with higher basal glucagon-like peptide 1 (GLP-1) levels at visit 1 (β = 1.52, P = 0.02 and β = 0.96, P = 0.002 for total and active GLP-1, respectively), and across all points of the OGTT after metformin administration. Regarding drug response, the T allele was associated with a shorter time (β = -7.00, P = 0.03) and a steeper slope (β = 0.23, P = 0.04) to trough glucose levels after glipizide administration, and lower visit 2 fasting glucose level adjusted for visit 1 fasting glucose level (β = -1.02, P = 0.04) and a greater decline in glucose level between visits (β = -1.61, P = 0.047) after metformin administration. CONCLUSIONS Our findings demonstrate that common variation at TCF7L2 influences acute responses to both glipizide and metformin in people without diabetes and highlight altered incretin signaling as a potential mechanism by which TCF7L2 variation increases T2D risk.
Collapse
Affiliation(s)
- Shylaja Srinivasan
- Pediatric Endocrine Unit, Massachusetts General Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Varinderpal Kaur
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Bindu Chamarthi
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA.,Department of Medicine, Harvard Medical School, Boston, MA
| | - Katherine R Littleton
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Ling Chen
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Alisa K Manning
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA.,Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| | - Jordi Merino
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | | | - Margo Hudson
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA.,Department of Medicine, Harvard Medical School, Boston, MA
| | - Allison Goldfine
- Department of Medicine, Harvard Medical School, Boston, MA.,Joslin Diabetes Center, Boston, MA
| | - Jose C Florez
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA .,Department of Medicine, Harvard Medical School, Boston, MA.,Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
19
|
A common variant within the HNF1B gene is associated with overall survival of multiple myeloma patients: results from the IMMEnSE consortium and meta-analysis. Oncotarget 2018; 7:59029-59048. [PMID: 27437873 PMCID: PMC5312293 DOI: 10.18632/oncotarget.10665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/19/2016] [Indexed: 01/01/2023] Open
Abstract
Diabetogenic single nucleotide polymorphisms (SNPs) have recently been associated with multiple myeloma (MM) risk but their impact on overall survival (OS) of MM patients has not been analysed yet. In order to investigate the impact of 58 GWAS-identified variants for type 2 diabetes (T2D) on OS of patients with MM, we analysed genotyping data of 936 MM patients collected by the International Multiple Myeloma rESEarch (IMMENSE) consortium and an independent set of 700 MM patients recruited by the University Clinic of Heidelberg. A meta-analysis of the cox regression results of the two sets showed that rs7501939 located in the HNF1B gene negatively impacted OS (HRRec= 1.44, 95% CI = 1.18-1.76, P = 0.0001). The meta-analysis also showed a noteworthy gender-specific association of the SLC30A8rs13266634 SNP with OS. The presence of each additional copy of the minor allele at rs13266634 was associated with poor OS in men whereas no association was seen in women (HRMen-Add = 1.32, 95% CI 1.13-1.54, P = 0.0003). In conclusion, these data suggest that the HNF1Brs7501939 SNP confers poor OS in patients with MM and that a SNP in SLC30A8 affect OS in men.
Collapse
|
20
|
Kodama S, Fujihara K, Ishiguro H, Horikawa C, Ohara N, Yachi Y, Tanaka S, Shimano H, Kato K, Hanyu O, Sone H. Quantitative Relationship Between Cumulative Risk Alleles Based on Genome-Wide Association Studies and Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis. J Epidemiol 2017; 28:3-18. [PMID: 29093303 PMCID: PMC5742374 DOI: 10.2188/jea.je20160151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many epidemiological studies have assessed the genetic risk of having undiagnosed or of developing type 2 diabetes mellitus (T2DM) using several single nucleotide polymorphisms (SNPs) based on findings of genome-wide association studies (GWAS). However, the quantitative association of cumulative risk alleles (RAs) of such SNPs with T2DM risk has been unclear. The aim of this meta-analysis is to review the strength of the association between cumulative RAs and T2DM risk. Systematic literature searches were conducted for cross-sectional or longitudinal studies that examined odds ratios (ORs) for T2DM in relation to genetic profiles. Logarithm of the estimated OR (log OR) of T2DM for 1 increment in RAs carried (1-ΔRA) in each study was pooled using a random-effects model. There were 46 eligible studies that included 74,880 cases among 249,365 participants. In 32 studies with a cross-sectional design, the pooled OR for T2DM morbidity for 1-ΔRA was 1.16 (95% confidence interval [CI], 1.13–1.19). In 15 studies that had a longitudinal design, the OR for incident T2DM was 1.10 (95% CI, 1.08–1.13). There was large heterogeneity in the magnitude of log OR (P < 0.001 for both cross-sectional studies and longitudinal studies). The top 10 commonly used genes significantly explained the variance in the log OR (P = 0.04 for cross-sectional studies; P = 0.006 for longitudinal studies). The current meta-analysis indicated that carrying 1-ΔRA in T2DM-associated SNPs was associated with a modest risk of prevalent or incident T2DM, although the heterogeneity in the used genes among studies requires us to interpret the results with caution.
Collapse
Affiliation(s)
- Satoru Kodama
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Niigata University Graduate School of Medical and Dental Sciences
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Hajime Ishiguro
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Chika Horikawa
- Department of Health and Nutrition, Faculty of Human Life Studies, University of Niigata Prefecture
| | - Nobumasa Ohara
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Yoko Yachi
- Department of Administrative Dietetics, Faculty of Health and Nutrition, Yamanashi Gakuin University
| | - Shiro Tanaka
- Department of Clinical Trial, Design & Management, Translational Research Center, Kyoto University Hospital
| | - Hitoshi Shimano
- Department of Internal Medicine, University of Tsukuba Institute of Clinical Medicine
| | - Kiminori Kato
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Niigata University Graduate School of Medical and Dental Sciences
| | - Osamu Hanyu
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| |
Collapse
|
21
|
Dey R, Schmidt EM, Abecasis GR, Lee S. A Fast and Accurate Algorithm to Test for Binary Phenotypes and Its Application to PheWAS. Am J Hum Genet 2017; 101:37-49. [PMID: 28602423 DOI: 10.1016/j.ajhg.2017.05.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022] Open
Abstract
The availability of electronic health record (EHR)-based phenotypes allows for genome-wide association analyses in thousands of traits and has great potential to enable identification of genetic variants associated with clinical phenotypes. We can interpret the phenome-wide association study (PheWAS) result for a single genetic variant by observing its association across a landscape of phenotypes. Because a PheWAS can test thousands of binary phenotypes, and most of them have unbalanced or often extremely unbalanced case-control ratios (1:10 or 1:600, respectively), existing methods cannot provide an accurate and scalable way to test for associations. Here, we propose a computationally fast score-test-based method that estimates the distribution of the test statistic by using the saddlepoint approximation. Our method is much (∼100 times) faster than the state-of-the-art Firth's test. It can also adjust for covariates and control type I error rates even when the case-control ratio is extremely unbalanced. Through application to PheWAS data from the Michigan Genomics Initiative, we show that the proposed method can control type I error rates while replicating previously known association signals even for traits with a very small number of cases and a large number of controls.
Collapse
Affiliation(s)
- Rounak Dey
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ellen M Schmidt
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Goncalo R Abecasis
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seunggeun Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Song J, Yang Y, Mauvais-Jarvis F, Wang YP, Niu T. KCNJ11, ABCC8 and TCF7L2 polymorphisms and the response to sulfonylurea treatment in patients with type 2 diabetes: a bioinformatics assessment. BMC MEDICAL GENETICS 2017; 18:64. [PMID: 28587604 PMCID: PMC5461698 DOI: 10.1186/s12881-017-0422-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 05/11/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Type 2 diabetes (T2D) is a worldwide epidemic with considerable health and economic consequences. Sulfonylureas are widely used drugs for the treatment of patients with T2D. KCNJ11 and ABCC8 encode the Kir6.2 (pore-forming subunit) and SUR1 (regulatory subunit that binds to sulfonylurea) of pancreatic β cell KATP channel respectively with a critical role in insulin secretion and glucose homeostasis. TCF7L2 encodes a transcription factor expressed in pancreatic β cells that regulates insulin production and processing. Because mutations of these genes could affect insulin secretion stimulated by sulfonylureas, the aim of this study is to assess associations between molecular variants of KCNJ11, ABCC8 and TCF7L2 genes and response to sulfonylurea treatment and to predict their potential functional effects. METHODS Based on a comprehensive literature search, we found 13 pharmacogenetic studies showing that single nucleotide polymorphisms (SNPs) located in KCNJ11: rs5219 (E23K), ABCC8: rs757110 (A1369S), rs1799854 (intron 15, exon 16 -3C/T), rs1799859 (R1273R), and TCF7L2: rs7903146 (intron 4) were significantly associated with responses to sulfonylureas. For in silico bioinformatics analysis, SIFT, PolyPhen-2, PANTHER, MutPred, and SNPs3D were applied for functional predictions of 36 coding (KCNJ11: 10, ABCC8: 24, and TCF7L2: 2; all are missense), and HaploReg v4.1, RegulomeDB, and Ensembl's VEP were used to predict functions of 7 non-coding (KCNJ11: 1, ABCC8: 1, and TCF7L2: 5) SNPs, respectively. RESULTS Based on various in silico tools, 8 KCNJ11 missense SNPs, 23 ABCC8 missense SNPs, and 2 TCF7L2 missense SNPs could affect protein functions. Of them, previous studies showed that mutant alleles of 4 KCNJ11 missense SNPs and 5 ABCC8 missense SNPs can be successfully rescued by sulfonylurea treatments. Further, 3 TCF7L2 non-coding SNPs (rs7903146, rs11196205 and rs12255372), can change motif(s) based on HaploReg v4.1 and are predicted as risk factors by Ensembl's VEP. CONCLUSIONS Our study indicates that a personalized medicine approach by tailoring sulfonylurea therapy of T2D patients according to their genotypes of KCNJ11, ABCC8, and TCF7L2 could attain an optimal treatment efficacy.
Collapse
Affiliation(s)
- Jingwen Song
- Department of Global Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112 USA
| | - Yunzhong Yang
- Department of Global Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112 USA
| | - Franck Mauvais-Jarvis
- Division of Endocrinology and Metabolism, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112 USA
| | - Yu-Ping Wang
- Department of Biomedical Engineering, Tulane University School of Science and Engineering, New Orleans, LA 70118 USA
| | - Tianhua Niu
- Department of Global Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112 USA
| |
Collapse
|
23
|
Chang S, Wang Z, Wu L, Lu X, Shangguan S, Xin Y, Li L, Wang L. Association between TCF7L2 polymorphisms and gestational diabetes mellitus: A meta-analysis. J Diabetes Investig 2017; 8:560-570. [PMID: 28002648 PMCID: PMC5497039 DOI: 10.1111/jdi.12612] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/08/2016] [Accepted: 12/15/2016] [Indexed: 12/13/2022] Open
Abstract
AIMS/INTRODUCTION Studies have been carried out to evaluate the correlation between TCF7L2 genetic polymorphisms and gestational diabetes mellitus (GDM) risk. However, the conclusions from these studies are incomplete, because partial single nucleotide polymorphisms (SNPs) were analyzed. We carried out a meta-analysis aimed to systematically evaluate TCF7L2 gene polymorphisms and GDM susceptibility in all population and racial/ethnic subgroups to afford a foundation for future research. MATERIALS AND METHODS Published studies censoring TCF7L2 variants and GDM risk were captured from the EMBASE, PubMed, CNKI and Wanfang databases. The meta-analysis was processed using software of RevMan 5.2 and Stata13. The relationship between TCF7L2 polymorphism and GDM occurrence was evaluated by pooled odds ratios. Stratified analysis based on race/ethnicity was also carried out. The allele-specific odds ratios and 95% confidence intervals were counted, and based on homogeneity evaluated using the I2 -test, fixed- or random-effects pooled measures were selected. RESULTS A total of 22 studies were covered, capturing eight TCF7L2 SNPs and involving 5,573 cases and 13,266 controls. Six of eight SNPs showed significant relationships with GDM occurrence, of which the SNPs rs7903146, rs12255372 and rs7901695 were the most powerful. Stratified analysis by race/ethnicity showed discrepant results in these three SNPs. In Caucasians and other races, all these SNPs were found to have a significant association with GDM risk, but in Asians, only SNP rs7903146 showed a significant association. CONCLUSIONS Six of eight SNPs were found to have significant associations between TCF7L2 variants and GDM risk in the overall population, with the most powerful in SNPs being rs7903146, rs12255372 and rs7901695, but the contribution of these SNPs to GDM risk were variable among different racial/ethnic groups.
Collapse
Affiliation(s)
- Shaoyan Chang
- Beijing Key Laboratory, Capital Institute of Pediatrics, Beijing, China
| | - Zhen Wang
- Beijing Key Laboratory, Capital Institute of Pediatrics, Beijing, China
| | - Lihua Wu
- Beijing Key Laboratory, Capital Institute of Pediatrics, Beijing, China
| | - Xiaolin Lu
- Beijing Key Laboratory, Capital Institute of Pediatrics, Beijing, China
| | | | - Yu Xin
- Beijing Key Laboratory, Capital Institute of Pediatrics, Beijing, China
| | - Li Li
- Neonatology Department, Capital Institute of Pediatrics, Beijing, China
| | - Li Wang
- Beijing Key Laboratory, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
24
|
Chidambaram M, Liju S, Saboo B, Sathyavani K, Viswanathan V, Pankratz N, Gross M, Mohan V, Radha V. Replication of genome-wide association signals in Asian Indians with early-onset type 2 diabetes. Acta Diabetol 2016; 53:915-923. [PMID: 27488727 DOI: 10.1007/s00592-016-0889-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 07/12/2016] [Indexed: 01/22/2023]
Abstract
AIMS To evaluate the association of 87 genetic variants previously associated with type 2 diabetes mellitus (T2DM) in genome-wide association studies of populations of European ancestry in an Asian Indian population with early-onset type 2 diabetes mellitus (EOT2DM). METHODS The study groups comprised of 877 type 2 diabetes individuals, 436 individuals with EOT2DM (age at diagnosis below 35 years), 441 individuals with older T2DM (diagnosis at 35 years or greater) and controls with normal glucose tolerance (NGT) (n = 400 younger than 35 years; n = 438 older than 35 years). The participants were genotyped for 87 SNPs from 44 genes and 27 intergenic loci. Associations were tested using logistic regression. RESULTS All the variants in TCF7L2 and CDKN2A/2B showed study-wide significance (p < 1.4 × 10-4) with T2DM, but only rs7903146, rs12243326, rs12255372 of TCF7L2 and rs7020996 of CDKN2A/2B showed study-wide significance (p < 1.4 × 10-4) with EOT2DM in this population. In addition, an intergenic SNP on chromosome 1 (rs10493685) was also shown to be study-wide significant (p = 7.1 × 10-6). Several additional SNPs previously associated with T2DM reached borderline significance in this study, but may have been limited by relatively low sample numbers. Various other SNPs of T2DM were not associated with EOT2DM. CONCLUSIONS Some of the variants in TCF7L2 and CDKN2A/2B associated with T2DM are associated with EOT2DM as well. An intergenic SNP on chromosome 1p31 showed association only with early-onset T2DM in this Asian Indian population. The lack of association with many other SNPs of T2DM may be a reflection of the lack of power of the study, sample size, differences in the frequencies of genetic polymorphisms in different ethnic groups, effect sizes, as well as ancestral differences in pattern of LD between the genetic variants involved in early- and late-onset T2DM.
Collapse
Affiliation(s)
- Manickam Chidambaram
- Madras Diabetes Research Foundation, 4, Conran Smith Road, Gopalapuram, Chennai, 600 086, India
- Division of Cardiovascular Research, Sidra Medical and Research Center, Doha, Qatar
| | - Samuel Liju
- Madras Diabetes Research Foundation, 4, Conran Smith Road, Gopalapuram, Chennai, 600 086, India
| | - Banshi Saboo
- Diabetologist and Endocrine and Metabolic Physician at Diabetes Care and Hormone Clinic, Ahmedabad, Gujarat, India
| | - Kumpatla Sathyavani
- M.V. Hospital for Diabetes and Prof. M. Viswanathan Diabetes Research Centre, Chennai, Tamil Nadu, India
| | - Vijay Viswanathan
- M.V. Hospital for Diabetes and Prof. M. Viswanathan Diabetes Research Centre, Chennai, Tamil Nadu, India
| | - Nathan Pankratz
- Department of Laboratory Medicine Pathology, Medical School University of Minnesota, Minneapolis, MN, USA
| | - Myron Gross
- Department of Laboratory Medicine Pathology, Medical School University of Minnesota, Minneapolis, MN, USA
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation, 4, Conran Smith Road, Gopalapuram, Chennai, 600 086, India
- Dr. Mohan's Diabetes Specialities Centre, WHO Collaborating Centre for Non-Communicable Diseases Prevention and Control, IDF Centre of Education, Chennai, India
| | - Venkatesan Radha
- Madras Diabetes Research Foundation, 4, Conran Smith Road, Gopalapuram, Chennai, 600 086, India.
| |
Collapse
|
25
|
Rizvi S, Raza ST, Rahman Q, Mahdi F. Role of GNB3, NET, KCNJ11, TCF7L2 and GRL genes single nucleotide polymorphism in the risk prediction of type 2 diabetes mellitus. 3 Biotech 2016; 6:255. [PMID: 28330327 PMCID: PMC5135703 DOI: 10.1007/s13205-016-0572-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 11/19/2016] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes (T2DM) is a polygenic metabolic disorder characterized by hyperglycemia occurring as a result of impaired insulin secretion or insulin resistance. Various environmental and genetic factors interact and increase the risk of T2DM and its complications. Among the various genetic factors associated with T2DM, single nucleotide polymorphism in different candidate genes have been studied intensively and the resulting genetic variants have been found to have either positive or negative association with T2DM thereby increasing or decreasing the risk of T2DM, respectively. In this review, we will focus on Guanine nucleotide-binding protein subunit beta 3 (GNB3), Norepinephrine Transporter (NET), Potassium Channel gene (KCNJ11), Transcription Factor 7-Like 2 (TCF7L2) and Glucocorticoid receptor (GRL) genes and their association with T2DM studied in different ethnic groups. The products of these genes are involved in the biochemical pathway leading to T2DM. Polymorphisms in these genes have been intensively studied in individuals of different ethnic origins. Results show that genetic variants of TCF7L2 and KCNJ11 genes have potential to emerge as a risk biomarker for T2DM whereas results of GNB3, GRL and NET genes have been controversial when studied in individuals of different ethnicities. We have tried to summarize the results generated globally in context to the selected genes which could possibly help researchers working in this field and would eventually help in understanding the mechanistic pathways of T2DM leading early diagnosis and prevention.
Collapse
|
26
|
Dhawan D, Padh H. Genetic variations in TCF7L2 influence therapeutic response to sulfonylureas in Indian diabetics. Diabetes Res Clin Pract 2016; 121:35-40. [PMID: 27639123 DOI: 10.1016/j.diabres.2016.08.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 08/17/2016] [Accepted: 08/25/2016] [Indexed: 12/25/2022]
Abstract
Sulfonylureas are widely used to treat type 2 diabetes, with considerable inter-individual variation in the hypoglycaemic response to sulfonylureas. Genetic variants in the gene encoding for transcription factor-7-like 2 (TCF7L2) have been associated with type 2 diabetes. This study aimed to study the effect of variations in TCF7L2 on therapeutic response to sulfonylureas in Type 2 diabetes mellitus patients. The effect of TCF7L2 rs12255372, rs7903146 and rs4506565 genotypes on glycaemic response was observed in 250 diabetic patients treated with sulfonylureas and sulfonylureas along with metformin. The genotyping tests were done by allele-specific multiplex PCR. Glycated haemoglobin (HbA1c) levels were used as phenotypic marker. 60% of sulfonylurea users did not achieve a target HbA1c levels of ⩽6.5% (48mmol/mol) (which denotes good control in diabetics). Genotype influenced response to sulfonylureas, with more treatment failure in the TT homozygotes in case of rs12255372 and rs4506565. The GG genotype at rs12255372 favourably influences treatment success with sulfonylurea therapy in patients with type 2 diabetes (p⩽0.05). At rs12255372, 70.5% GT or TT genotype failed to achieve therapeutic target, an absolute difference of 19% compared to GG homozygotes. Our preliminary data show that genetic variation at rs12255372 has a direct correlation with therapeutic success with sulfonylureas in type 2 diabetes, hence paving the way for better treatment outcomes in diabetics.
Collapse
Affiliation(s)
- Dipali Dhawan
- B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej-Gandhinagar Highway, Thaltej, Ahmedabad 380 054, Gujarat, India.
| | - Harish Padh
- B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Thaltej-Gandhinagar Highway, Thaltej, Ahmedabad 380 054, Gujarat, India
| |
Collapse
|
27
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
28
|
Pharmacogenomics in type 2 diabetes: oral antidiabetic drugs. THE PHARMACOGENOMICS JOURNAL 2016; 16:399-410. [DOI: 10.1038/tpj.2016.54] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/08/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023]
|
29
|
Jin T. Current Understanding on Role of the Wnt Signaling Pathway Effector TCF7L2 in Glucose Homeostasis. Endocr Rev 2016; 37:254-77. [PMID: 27159876 DOI: 10.1210/er.2015-1146] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The role of the Wnt signaling pathway in metabolic homeostasis has drawn our intensive attention, especially after the genome-wide association study discovery that certain polymorphisms of its key effector TCF7L2 are strongly associated with the susceptibility to type 2 diabetes. For a decade, great efforts have been made in determining the function of TCF7L2 in various metabolic organs, which have generated both considerable achievements and disputes. In this review, I will briefly introduce the canonical Wnt signaling pathway, focusing on its effector β-catenin/TCF, including emphasizing the bidirectional feature of TCFs and β-catenin post-translational modifications. I will then summarize the observations on the association between TCF7L2 polymorphisms and type 2 diabetes risk. The main content, however, is on the intensive functional exploration of the metabolic role of TCF7L2, including the disputes generated on determining its role in the pancreas and liver with various transgenic mouse lines. Finally, I will discuss those achievements and disputes and present my future perspectives.
Collapse
Affiliation(s)
- Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
30
|
Replication Study in a Japanese Population of Six Susceptibility Loci for Type 2 Diabetes Originally Identified by a Transethnic Meta-Analysis of Genome-Wide Association Studies. PLoS One 2016; 11:e0154093. [PMID: 27115357 PMCID: PMC4845992 DOI: 10.1371/journal.pone.0154093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 04/08/2016] [Indexed: 01/21/2023] Open
Abstract
AIM We performed a replication study in a Japanese population to evaluate the association between type 2 diabetes and six susceptibility loci (TMEM154, SSR1, FAF1, POU5F1, ARL15, and MPHOSPH9) originally identified by a transethnic meta-analysis of genome-wide association studies (GWAS) in 2014. METHODS We genotyped 7,620 Japanese participants (5,817 type 2 diabetes patients and 1,803 controls) for each of the single nucleotide polymorphisms (SNPs) using a multiplex polymerase chain reaction invader assay. The association of each SNP locus with the disease was evaluated using logistic regression analysis. RESULTS Of the six SNPs examined in this study, four (rs6813195 near TMEM154, rs17106184 in FAF1, rs3130501 in POU5F1 and rs4275659 near MPHOSPH9) had the same direction of effect as in the original reports, but two (rs9505118 in SSR1 and rs702634 in ARL15) had the opposite direction of effect. Among these loci, rs3130501 and rs4275659 were nominally associated with type 2 diabetes (rs3130501; p = 0.017, odds ratio [OR] = 1.113, 95% confidence interval [CI] 1.019-1.215, rs4275659; p = 0.012, OR = 1.127, 95% CI 1.026-1.238, adjusted for sex, age and body mass index), but we did not observe a significant association with type 2 diabetes for any of the six evaluated SNP loci in our Japanese population. CONCLUSIONS Our results indicate that effects of the six SNP loci identified in the transethnic GWAS meta-analysis are not major among the Japanese, although SNPs in POU5F1 and MPHOSPH9 loci may have some effect on susceptibility to type 2 diabetes in this population.
Collapse
|
31
|
Kodama S, Fujihara K, Ishiguro H, Horikawa C, Ohara N, Yachi Y, Tanaka S, Shimano H, Kato K, Hanyu O, Sone H. Meta-analytic research on the relationship between cumulative risk alleles and risk of type 2 diabetes mellitus. Diabetes Metab Res Rev 2016; 32:178-86. [PMID: 26265102 DOI: 10.1002/dmrr.2680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Our aim is to examine the dose-response association between cumulative genetic risk and actual risk of type 2 diabetes mellitus (T2DM) and the influence of adjustment for covariates on T2DM risk through a comprehensive meta-analysis of observational studies. METHODS Electronic literature search using EMBASE and MEDLINE (from 2003 to 2014) was conducted for cross-sectional or longitudinal studies that presented the odds ratio (OR) for T2DM in each group with categories based on the total number of risk alleles (RAs) carried (RAtotal ) using at least two single-nucleotide polymorphisms. Spline regression model was used to determine the shape of the relationship between the difference from the referent group of each study in RAtotal (ΔRAtotal ) and the natural logarithms of ORs (log OR) for T2DM. RESULTS Sixty-five eligible studies that included 68 267 cases among 182 603 participants were analysed. In both crude and adjusted ORs, defined by adjusting the risk for at least two confounders among age, gender and body mass index, the slope of the log OR for T2DM became less steep as the ΔRAtotal increased. In the analysis limited to 14 cross-sectional and four longitudinal studies presenting both crude and adjusted ORs, regression curves of both ORs in relation to ΔRAtotal were almost identical. CONCLUSION Using only single-nucleotide polymorphisms for T2DM screening was of limited value. However, when genotypic T2DM risk was considered independently from risk in relation to covariates, it was suggested that genetic profiles might have a supplementary role related to conventional T2DM risk factors in identifying individuals at high risk of T2DM. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Satoru Kodama
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Diseases, Niigata University Faculty of Medicine, Niigata, Japan
| | - Kazuya Fujihara
- Department of Internal Medicine, University of Tsukuba Institute of Clinical Medicine, Ibaraki, Japan
| | - Hajime Ishiguro
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Chika Horikawa
- Department of Health and Nutrition, Faculty of Human Life Studies, University of Niigata Prefecture, Niigata, Japan
| | - Nobumasa Ohara
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Diseases, Niigata University Faculty of Medicine, Niigata, Japan
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Yoko Yachi
- Department of Administrative Dietetics, Faculty of Health and Nutrition, Yamanashi Gakuin University, Yamanashi, Japan
| | - Shiro Tanaka
- Department of Clinical Trial, Design and Management, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine, University of Tsukuba Institute of Clinical Medicine, Ibaraki, Japan
| | - Kiminori Kato
- Department of Laboratory Medicine and Clinical Epidemiology for Prevention of Noncommunicable Diseases, Niigata University Faculty of Medicine, Niigata, Japan
| | - Osamu Hanyu
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| |
Collapse
|
32
|
Rezaei M, Palizban A, Zamani-doabi S, Shojaee M. Transcription factor 7-like 2 (TCF7L2) Gene Polymorphism rs7903146 is Associated with Lipid Profile and Risk of Cardiovascular Disease in Metabolic Syndrome Subjects. ACTA ACUST UNITED AC 2016. [DOI: 10.15412/j.jbtw.01050703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Pourahmadi M, Erfanian S, Moradzadeh M, Jahromi AS. Non-Association between rs7903146 and rs12255372 Polymorphisms in Transcription Factor 7-Like 2 Gene and Type 2 Diabetes Mellitus in Jahrom City, Iran. Diabetes Metab J 2015; 39:512-7. [PMID: 26616591 PMCID: PMC4696988 DOI: 10.4093/dmj.2015.39.6.512] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Transcription factor 7-like 2 (TCF7L2) is a transcription factor in the Wnt signaling pathway. High levels of TCF7L2 have been reported in most human tissues, including the heart, lung, brain, liver, kidney, placenta, adipose tissues, and pancreatic β-cells. The purpose of this study was to assess the association between TCF7L2 polymorphisms (rs12255372 and rs7903146) and type 2 diabetes mellitus in the city of Jahrom, Iran. METHODS This case-control study was conducted with 200 patients referred to Diabetes Clinics and 200 healthy subjects in Jahrom City. Biochemical characteristics were first determined. TCF7L2 rs1255372 and rs7903146 polymorphisms were then genotyped using the polymerase chain reaction-restriction fragment length polymorphism method. RESULTS T-allele frequencies of both single nucleotide polymorphisms (SNPs) were significantly higher in diabetic patients than in normal glucose-tolerant subjects (rs12255372: 20.3% vs. 14.5%; rs7903146: 28.5% vs. 22.25%). The rs12255372 (G/T) polymorphism analysis showed an odds ratio of 0.473 (95% confidence interval [CI], 0.170 to 1.314; P=0.151) for the TT genotype and 0.646 (95% CI, 0.410 to 1.019; P=0.060) for the TG genotype, compared with the GG genotype. The rs7903146 (C/T) polymorphism odds ratios for TT and TC genotypes were 0.564 (95% CI, 0.280 to 1.135; P=0.109) and 0.751 (95% CI, 0.487 to 1.157; P=0.194) compared with the CC genotype, respectively. CONCLUSION The rs12255372 and rs7903146 SNPs of the TCF7L2 gene were not associated with insulin resistance in the evaluated population.
Collapse
Affiliation(s)
- Mohammad Pourahmadi
- Department of Anatomy, Jahrom University of Medical Sciences, Jahrom, Iran
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Saiedeh Erfanian
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Malihe Moradzadeh
- Department of New Sciences and Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
34
|
Ding Y, Hu Z, Yuan S, Xie P, Liu Q. Association between transcription factor 7-like 2 rs7903146 polymorphism and diabetic retinopathy in type 2 diabetes mellitus: A meta-analysis. Diab Vasc Dis Res 2015; 12:436-44. [PMID: 26316572 DOI: 10.1177/1479164115598274] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
As one of the vascular complications of type 2 diabetes mellitus, the incidence of diabetes retinopathy is greatly increasing worldwide. Both genetic and environmental factors are involved in the pathologies. A meta-analysis was conducted to assess the association between transcription factor 7-like 2 polymorphism (rs7903146) and type 2 diabetic retinopathy. Published literature from PubMed, Web of Science and China National Knowledge Infrastructure were retrieved. Pooled odds ratios with 95% confidence intervals were calculated to estimate the strength of the association. Eight studies including 6422 participants were included in the final meta-analysis. Our analysis provides substantial evidence that the rs7903146 variant is significantly associated with the risk of diabetic retinopathy in Caucasian populations while not in East Asian populations. The variant of rs7903146 appeared more likely to be a promising genetic biomarker of diabetic retinopathy in Caucasians.
Collapse
Affiliation(s)
- Yuzhi Ding
- The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Zizhong Hu
- The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Ping Xie
- The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qinghuai Liu
- The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Allahdini M, Kamalidehghan B, Akbari L, Azadfar P, Rahmani A, Ahmadipour F, Meng GY, Masserrat A, Houshmand M. Prevalence of the rs7903146C>T polymorphism in TCF7L2 gene for prediction of type 2 diabetes risk among Iranians of different ethnicities. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5835-41. [PMID: 26604685 PMCID: PMC4629960 DOI: 10.2147/dddt.s82485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Pharmacogenetics is the study of genetic polymorphisms affecting responses to drug therapy. The common rs7903146 (C>T) polymorphism of the TCF7L2 gene has recently been associated with type 2 diabetes (T2D). In this study, prevalence of the rs7903146 (C>T) polymorphism in the TCF7L2 gene for prediction of T2D risk was examined in an Iranian population of different ethnicities. Methods The prevalence of rs7903146 (C>T) and the predicted phenotypes, including extensive metabolizers, intermediate metabolizers, and poor metabolizers were investigated in blood samples of 300 unrelated healthy individuals in an Iranian population, including Fars, Turk, Lure, and Kurd, using polymerase chain reaction restriction fragment length polymorphism and direct genomic DNA sequencing. Results The homozygous wild-type (C/C), heterozygous (C/T), and homozygous (T/T) allelic frequencies of rs7903146 (C>T) in the TCF7L2 gene were 29% (extensive metabolizers), 66.34% (intermediate metabolizers), and 4.66% (poor metabolizers), respectively. The C/C, C/T, and T/T genotypic frequencies of the rs7903146 (C>T) allele were significantly different (P<0.01) among Iranians of different ethnicities. The frequency of the homozygous T/T variant of the rs7903146 (C>T) allele was significantly low in the Lure (P<0.01) and high in the Fars (P<0.001) ethnicities. Additionally, the frequency of the T/T variant of the rs7903146 (C>T) allele in the South of Iran was the highest (P<0.04), while the East of Iran had the lowest frequency (P<0.01). Conclusion The prediction of rs7903146 (C>T) is required in drug research and routine treatment, where the information would be helpful for clinicians to optimize therapy and adverse drug reactions and predict drug response in individuals at risk of T2D.
Collapse
Affiliation(s)
- Mojgan Allahdini
- Department of Molecular Biology, Ahar Branch Islamic Azad University, Ahar, Iran
| | - Behnam Kamalidehghan
- Pharmacy Department, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Leila Akbari
- Department of Biology, Sciences and Research Branch, Azad University, Tehran, Iran
| | - Parisa Azadfar
- Department of Biology, Sciences and Research Branch, Azad University, Tehran, Iran
| | - Ali Rahmani
- Department of Molecular Biology, Ahar Branch Islamic Azad University, Ahar, Iran
| | - Fatemeh Ahmadipour
- Pharmacy Department, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Goh Yong Meng
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Malaysia
| | | | - Massoud Houshmand
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
36
|
Ríos R, Lupiañez CB, Campa D, Martino A, Martínez-López J, Martínez-Bueno M, Varkonyi J, García-Sanz R, Jamroziak K, Dumontet C, Cayuela AJ, Wętek M, Landi S, Rossi AM, Lesueur F, Reis RM, Moreno V, Marques H, Jurczyszyn A, Andersen V, Vogel U, Buda G, Orciuolo E, Jacobsen SEH, Petrini M, Vangsted AJ, Gemignani F, Canzian F, Jurado M, Sainz J. Type 2 diabetes-related variants influence the risk of developing multiple myeloma: results from the IMMEnSE consortium. Endocr Relat Cancer 2015; 22:545-59. [PMID: 26099684 DOI: 10.1530/erc-15-0029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/15/2015] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes (T2D) has been suggested to be a risk factor for multiple myeloma (MM), but the relationship between the two traits is still not well understood. The aims of this study were to evaluate whether 58 genome-wide-association-studies (GWAS)-identified common variants for T2D influence the risk of developing MM and to determine whether predictive models built with these variants might help to predict the disease risk. We conducted a case-control study including 1420 MM patients and 1858 controls ascertained through the International Multiple Myeloma (IMMEnSE) consortium. Subjects carrying the KCNQ1rs2237892T allele or the CDKN2A-2Brs2383208G/G, IGF1rs35767T/T and MADDrs7944584T/T genotypes had a significantly increased risk of MM (odds ratio (OR)=1.32-2.13) whereas those carrying the KCNJ11rs5215C, KCNJ11rs5219T and THADArs7578597C alleles or the FTOrs8050136A/A and LTArs1041981C/C genotypes showed a significantly decreased risk of developing the disease (OR=0.76-0.85). Interestingly, a prediction model including those T2D-related variants associated with the risk of MM showed a significantly improved discriminatory ability to predict the disease when compared to a model without genetic information (area under the curve (AUC)=0.645 vs AUC=0.629; P=4.05×10(-) (06)). A gender-stratified analysis also revealed a significant gender effect modification for ADAM30rs2641348 and NOTCH2rs10923931 variants (Pinteraction=0.001 and 0.0004, respectively). Men carrying the ADAM30rs2641348C and NOTCH2rs10923931T alleles had a significantly decreased risk of MM whereas an opposite but not significant effect was observed in women (ORM=0.71 and ORM=0.66 vs ORW=1.22 and ORW=1.15, respectively). These results suggest that TD2-related variants may influence the risk of developing MM and their genotyping might help to improve MM risk prediction models.
Collapse
Affiliation(s)
- Rafael Ríos
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Carmen Belén Lupiañez
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Daniele Campa
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Alessandro Martino
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Joaquin Martínez-López
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Manuel Martínez-Bueno
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Judit Varkonyi
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Ramón García-Sanz
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Krzysztof Jamroziak
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Charles Dumontet
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Andrés Jerez Cayuela
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Marzena Wętek
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Stephano Landi
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Anna Maria Rossi
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Fabienne Lesueur
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Rui Manuel Reis
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Victor Moreno
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Herlander Marques
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Artur Jurczyszyn
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Vibeke Andersen
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Ulla Vogel
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Gabriele Buda
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Enrico Orciuolo
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Svend E H Jacobsen
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Mario Petrini
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Annette J Vangsted
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Federica Gemignani
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Federico Canzian
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark
| | - Manuel Jurado
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| | - Juan Sainz
- Genomic Oncology AreaGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, Granada, 18016 Granada, SpainHematology DepartmentVirgen de las Nieves University Hospital, Granada, SpainGenomic Epidemiology GroupGerman Cancer Research Center (DKFZ), Heidelberg, GermanyDepartment of BiologyUniversity of Pisa, Pisa, ItalyDepartment of HematologyHospital Universitario Doce de Octubre, Madrid, SpainArea of Genomic MedicineGENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, SpainSemmelweis UniversityBudapest, HungaryHaematology DepartmentUniversity Hospital of Salamanca and IBSAL, Salamanca, SpainMedical University of LodzLodz, PolandINSERM UMR 1052/CNRS 5286Université Claude Bernard Lyon I, Lyon, FranceMorales Meseguer General University HospitalMurcia, SpainHaematoloy ClinikHolly Cross Cancer Center, Kielce, PolandINSERMU900, Genetic Epidemiology of Cancers team, Institut Curie, Mines ParisTech, Paris, FranceLife and Health Sciences Research Institute (ICVS)School of Health Sciences, University of Minho, Braga, PortugalICVS/3B's - PT Government Associate LaboratoryBraga/Guimarães, PortugalMolecular Oncology Research CenterBarretos Cancer Hospital, Barretos, BrazilIDIBELL - Catalan Institute of OncologyUniversity of Barcelona, Barcelona 08907, SpainDepartment of HematologyCracow University Hospital, Cracow, PolandOrgan CenterHospital of Southern Jutland, DK-6200 Aabenraa, DenmarkFaculty of Health SciencesInstitute of Regional Health Research, University of Southern Denmark, DK-5000 Odense C, DenmarkUO HematologyDepartment of Internal and Experimental Medicine, University of Pisa, Pisa, ItalyClinic of Biochemistry and ImmunologyLaboratory Center, Hospital of Southern Jutland, Aabenraa, DenmarkDepartment of HaematologyRigshospitalet and Roskilde Hospital, Copenhagen University, Copenhagen, Denmark Genomic Oncology AreaGENYO, Cen
| |
Collapse
|
37
|
de Melo SF, Frigeri HR, dos Santos-Weiss ICR, Réa RR, de Souza EM, Alberton D, Gomes de Moraes Rego F, Picheth G. Polymorphisms in FTO and TCF7L2 genes of Euro-Brazilian women with gestational diabetes. Clin Biochem 2015; 48:1064-7. [PMID: 26102344 DOI: 10.1016/j.clinbiochem.2015.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/14/2015] [Accepted: 06/15/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate the association between fat mass and obesity-associated (FTO) gene polymorphisms rs8050136C>A and rs9939609T>A, and transcription factor 7-like 2 (TCF7L2) gene polymorphisms rs12255372G>T and rs7903146C>T, in a sample group of pregnant Euro-Brazilian women with or without gestational diabetes mellitus (GDM). METHODS Subjects were classified as either healthy pregnant control (n=200) or GDM (n=200) according to the 2010 criteria of the American Diabetes Association. The polymorphisms were genotyped using fluorescent probes (TaqMan®). RESULTS All groups were in the Hardy-Weinberg equilibrium. The genotype and allele frequencies of the examined polymorphisms did not exhibit significant difference (P>0.05) between the groups. In the healthy and GDM pregnant women groups, the A-allele frequencies (95% CI) of FTO polymorphisms rs8050136 and rs9939609 were 39% (34-44%); 38% (33-43%) and 40% (35-45%); 41% (36-46%), respectively; and the T-allele frequencies of TCF7L2 polymorphisms rs12255372 and rs7903146 were 30% (26-35%), 32% (27-37%) and 29% (25-34%), 36% (31-41%), respectively. CONCLUSION The examined polymorphisms were not associated with GDM in the Euro-Brazilian population studied.
Collapse
Affiliation(s)
| | - Henrique Ravanhol Frigeri
- Post-Graduate Program in Pharmaceutical Science, Federal University of Parana, Brazil; Health and Biosciences School, Pontifical Catholic University of Parana, Curitiba, Parana, Brazil
| | | | - Rosângela Roginski Réa
- Endocrinology and Metabolism Service (SEMPR), Clinical Hospital, Federal University of Parana, Brazil
| | | | - Dayane Alberton
- Post-Graduate Program in Pharmaceutical Science, Federal University of Parana, Brazil
| | | | - Geraldo Picheth
- Post-Graduate Program in Pharmaceutical Science, Federal University of Parana, Brazil.
| |
Collapse
|
38
|
Nanfa D, Sobngwi E, Atogho-Tiedeu B, Noubiap JJN, Donfack OS, Mofo EPM, Guewo-Fokeng M, Nguimmo Metsadjio A, Ndonwi Ngwa E, Pokam Fosso P, Djahmeni E, Djokam-Dadjeu R, Evehe MS, Aminkeng F, Mbacham WF, Mbanya JC. Association between the TCF7L2 rs12255372 (G/T) gene polymorphism and type 2 diabetes mellitus in a Cameroonian population: a pilot study. Clin Transl Med 2015; 4:17. [PMID: 25995831 PMCID: PMC4434239 DOI: 10.1186/s40169-015-0058-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/01/2015] [Indexed: 01/22/2023] Open
Abstract
Background To study the relationship between the rs12255372 (G/T) polymorphism of the transcription factor 7-like 2 (TCF7L2) and type 2 diabetes mellitus (T2DM) in a Cameroonian population. Methods This case–control study included 60 T2DM patients and 60 healthy normoglycemic controls, all unrelated and of Cameroonian origin, aged above 40 years (range 40–87). The Restriction Fragment Length Polymorphism - Polymerase Chain Reaction (RFLP-PCR) was used for genotyping. Results The T allele frequency was significantly higher in the diabetic group (0.44) than in the control group (0.17). This allele was significantly associated to a greater risk of developing T2DM as compared to the G allele (OR = 3.92, 95% CI 2.04 – 7.67, p < 0.0001). The codominant (additive) model explained best the risk of developing the disease, as the TT genotype was significantly associated to T2DM when compared to the GG genotype (OR = 4.45, 95% CI 1.64 – 12.83, p = 0.0014). By logistic regression adjusted for age, this OR was 4.33 (95% CI: 1.57 – 11.92, p = 0.005). Conclusion Our findings suggest that the rs12255372 (G/T) polymorphism of the TCF7L2 gene is an important risk factor for T2DM in the Cameroonian population.
Collapse
Affiliation(s)
- Dieudonne Nanfa
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Eugene Sobngwi
- Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon ; Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon ; National Obesity Center, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Barbara Atogho-Tiedeu
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean Jacques N Noubiap
- Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa ; Medical Diagnostic Center, Yaoundé, Cameroon
| | - Olivier Sontsa Donfack
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Edith Pascale Mato Mofo
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Magellan Guewo-Fokeng
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | | | - Elvis Ndonwi Ngwa
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Priscille Pokam Fosso
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Eric Djahmeni
- Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - Rosine Djokam-Dadjeu
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Marie-Solange Evehe
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Folefac Aminkeng
- The Canadian Pharmacogenomics Network for Drug Safety (CPNDS), Center for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Wilfred F Mbacham
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Public Health Research Biotechnologies, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean Claude Mbanya
- Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon ; Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon ; National Obesity Center, Yaoundé Central Hospital, Yaoundé, Cameroon
| |
Collapse
|
39
|
Guewo-Fokeng M, Sobngwi E, Atogho-Tiedeu B, Donfack OS, Noubiap JJN, Ngwa EN, Mato-Mofo EP, Fosso PP, Djahmeni E, Djokam-Dadjeu R, Evehe MS, Aminkeng F, Mbacham WF, Mbanya JC. Contribution of the TCF7L2 rs7903146 (C/T) gene polymorphism to the susceptibility to type 2 diabetes mellitus in Cameroon. J Diabetes Metab Disord 2015; 14:26. [PMID: 25897419 PMCID: PMC4403887 DOI: 10.1186/s40200-015-0148-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/08/2015] [Indexed: 01/08/2023]
Abstract
Background Data on the genetic variants for type 2 diabetes mellitus (T2DM) in sub-Saharan African populations are very scarce. This study aimed to investigate the association of transcription factor 7-like (TCF7L2) with T2DM in a Cameroonian population and explore possible genotype-phenotype correlation. Methods This is a case–control study involving 37 T2DM patients and 37 non-diabetic volunteers of Cameroonian ethnicity aged 40 years old and above. We collected clinical and biological data to determine phenotypic traits. TCF7L2 was analyzed by genotyping for rs7903146 (C/T) using PCR-RFLP. Biochemical analyses were performed using a spectrophotometer with Chronolab kits. Statistical analyses were carried out using IBM SPSS, PS and Quanto. Results TCF7L2 was associated with T2DM in this Cameroonian population (p = 0.013 for alleles, and p = 0.013 for genotypes). The risk allele was C (9.5% patients vs. 0% healthy controls, OR = 16.56) and the protective allele was T (90.5% patients vs. 100.0% healthy controls, OR = 0.06). The risk genotype was C/T (18.9% patients vs. 0% healthy controls, OR = 18.44), while the protective genotype was T/T (81.1% patients vs. 100.0% healthy controls, OR = 0.054). The statistical power was 99.99%. TCF7L2 was not preferentially associated with a specific disease phenotype. Conclusion TCF7L2 is associated with T2DM in this Cameroonian population. The association is not dependent on a specific T2DM phenotype. Clinical genetic testing for TCF7L2 can help to predict the occurrence of T2DM in Cameroon.
Collapse
Affiliation(s)
- Magellan Guewo-Fokeng
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Eugene Sobngwi
- Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon ; Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon ; National Obesity Center, Yaoundé Central Hospital, University of Yaoundé 1, Yaoundé, Cameroon
| | - Barbara Atogho-Tiedeu
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Olivier Sontsa Donfack
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean Jacques N Noubiap
- Department of Medicine, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa ; Medical Diagnostic Center, Yaoundé, Cameroon
| | - Elvis Ndonwi Ngwa
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Edith Pascale Mato-Mofo
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Priscille Pokam Fosso
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Eric Djahmeni
- National Obesity Center, Yaoundé Central Hospital, University of Yaoundé 1, Yaoundé, Cameroon
| | - Rosine Djokam-Dadjeu
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Marie-Solange Evehe
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Folefac Aminkeng
- The Canadian Pharmacogenomics Network for Drug Safety (CPNDS), Center for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Wilfred F Mbacham
- Department of Biochemistry, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon ; Laboratory for Public Health Research Biotechnologies, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon
| | - Jean Claude Mbanya
- Laboratory for Molecular Medicine and Metabolism, Biotechnology Center, University of Yaoundé I, Yaoundé, Cameroon ; Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon ; National Obesity Center, Yaoundé Central Hospital, University of Yaoundé 1, Yaoundé, Cameroon
| |
Collapse
|
40
|
Genetic influences on the association between fetal growth and susceptibility to type 2 diabetes. J Dev Orig Health Dis 2014; 1:96-105. [PMID: 25143063 DOI: 10.1017/s2040174410000127] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The fetal insulin hypothesis proposes that low birth weight and susceptibility to type 2 diabetes (T2D) could both be two phenotypes of the same genotype. Insulin is a key growth factor in utero, and T2D is characterized by insulin resistance and/or beta-cell dysfunction. Therefore, genetic variants impacting on insulin secretion and action are likely to alter both fetal growth and susceptibility to T2D. There are three lines of evidence in support of this hypothesis. (1) Studies of rare monogenic diabetes have shown mutations in a single gene, such as GCK or KCNJ11, can cause diabetes by reducing insulin secretion, and these mutations are also associated with reduced birth weight. (2) Epidemiological studies have indicated that children born to fathers with diabetes are born smaller. As the father cannot influence the intrauterine environment, this association is likely to reflect genes inherited by the fetus from the father. (3) The most compelling evidence comes from recent genome-wide association studies. Variants in the CDKAL1 and HHEX-IDE genes that predispose to diabetes, if present in the fetus, are associated with reduced birth weight. These data provide evidence for a genetic contribution to the association between low birth weight and susceptibility to T2D. This genetic background is important to take into consideration when investigating the impact of environmental determinants and developing strategies for intervention and prevention.
Collapse
|
41
|
Pradas-Juni M, Nicod N, Fernández-Rebollo E, Gomis R. Differential transcriptional and posttranslational transcription factor 7-like regulation among nondiabetic individuals and type 2 diabetic patients. Mol Endocrinol 2014; 28:1558-70. [PMID: 25058603 DOI: 10.1210/me.2014-1065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Human genetic studies have revealed that the T minor allele of single nucleotide polymorphism rs7903146 in the transcription factor 7-like 2 (TCF7L2) gene is strongly associated with an increased risk of diabetes by 30%-40%. Molecular and clinical studies are of great importance for understanding how this unique variation in TCF7L2 influences type 2 diabetes (T2D) onset and progression. At the molecular level, some studies have been performed in diabetic mice and pancreatic islets from healthy human donors. Whereas TCF7L2 mRNA levels are up-regulated in islets, protein levels are down-regulated. We performed studies on TCF7L2 splicing, mRNA expression, and protein levels in immortalized human lymphocytes from nondiabetic individuals and T2D patients carrying the C/C or the at-risk T/T genotype. Our results show differential expression of TCF7L2 splice variants between nondiabetic and T2D patients carrying the at-risk genotype, as well as differences in protein levels. Therefore, we investigated the regulation of splice variants, and our results propose that splicing of exon 4 is under control of the serine-arginine-rich factor transformer 2 β (TRA2B). Finally, we studied the endoplasmic reticulum stress pathways, looking for a posttranslational explanation. We saw a shift in the activation of these pathways between nondiabetic individuals and T2D patients carrying the at-risk genotype. These results suggest that, in human immortalized lymphocytes carrying the at-risk T/T genotype, first the differential expression of TCF7L2 splice variants implies a regulation, at least for exon 4, by TRA2B and second, the differential protein levels between both T/T carriers point to a different activation of endoplasmic reticulum stress pathways.
Collapse
Affiliation(s)
- M Pradas-Juni
- Diabetes and Obesity Research Laboratory (M.P.-J., N.N., E.F-R., R.G.), Institut d'Investigacions Biomèdiques August Pi i Sunyer, 08036 Barcelona, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (M.P.-J., N.N., E.F.-R., R.G.), 08017 Barcelona, Spain; Department of Medicine (R.G.), University of Barcelona, 08036 Barcelona, Spain; and Hospital Clínic de Barcelona (R.G.), 08036 Barcelona, Spain
| | | | | | | |
Collapse
|
42
|
DeMenna J, Puppala S, Chittoor G, Schneider J, Kim JY, Shaibi GQ, Mandarino LJ, Duggirala R, Coletta DK. Association of common genetic variants with diabetes and metabolic syndrome related traits in the Arizona Insulin Resistance registry: a focus on Mexican American families in the Southwest. Hum Hered 2014; 78:47-58. [PMID: 25060389 DOI: 10.1159/000363411] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 05/06/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND/AIMS The increased occurrence of type 2 diabetes and its clinical correlates is a global public health issue, and there are continued efforts to find its genetic determinant across ethnically diverse populations. The aims of this study were to determine the heritability of diabetes and metabolic syndrome phenotypes in the Arizona Insulin Resistance (AIR) registry and to perform an association analysis of common single nucleotide polymorphisms (SNPs) identified by GWAS with these traits. All study participants were Mexican Americans from the AIR registry. METHODS Metabolic, anthropometric, demographic and medical history information was obtained on the 667 individuals enrolled in the registry. RESULTS The heritability estimates were moderate to high in magnitude and significant, indicating that the AIR registry is well suited for the identification of genetic factors contributing to diabetes and the metabolic syndrome. From the 30 GWAS genes selected (some genes were represented by multiple SNPs), 20 SNPs exhibited associations with one or more of the diabetes related traits with nominal significance (p ≤ 0.05). In addition, 25 SNPs were nominally significantly associated with one or more of the metabolic phenotypes tested (p ≤ 0.05). Most notably, 5 SNPs from 5 genes [body mass index (BMI), hip circumference: rs3751812/FTO; fasting plasma glucose, hemoglobin A1c: rs4607517/GCK; very-low-density lipoprotein: rs10830963/MTNR1B; BMI: rs13266634/SLC30A8, and total cholesterol, low-density lipoprotein: rs7578597/THADA] were significantly associated with obesity, glycemic, and lipid phenotypes when using the multiple testing significance threshold of 0.0015. CONCLUSION These findings extend previous work on Mexican Americans to suggest that metabolic disease is strongly influenced by genetic background in this high-risk population.
Collapse
Affiliation(s)
- Jacob DeMenna
- School of Life Sciences, Arizona State University, Tempe, Ariz., USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Impact of TCF7L2 single nucleotide polymorphisms on hydrochlorothiazide-induced diabetes. Pharmacogenet Genomics 2014; 23:697-705. [PMID: 24128935 DOI: 10.1097/fpc.0000000000000012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Thiazide diuretics have been associated with increased risk for new onset diabetes (NOD), but pharmacogenetic markers of thiazide-induced NOD are not well studied. Single nucleotide polymorphisms (SNPs) in the transcription factor 7-like 2 gene (TCF7L2) represent the strongest and most reproducible genetic associations with diabetes. We investigated the association of tag SNPs in TCF7L2 with thiazide-induced NOD. METHODS We identified cases that developed NOD and age, sex, and race/ethnicity-matched controls from the INternational VErapamil SR-Trandolapril STudy (INVEST). INVEST compared cardiovascular outcomes between two antihypertensive treatment strategies in ethnically diverse patients with hypertension and coronary artery disease. We genotyped 101 TCF7L2 tag SNPs and used logistic regression to test for pharmacogenetic (SNP×hydrochlorothiazide treatment) interactions. Permuted interaction P values were corrected with the PACT test and adjusted for diabetes-related variables. RESULTS In INVEST whites, we observed three TCF7L2 SNPs with significant SNP×treatment interactions for NOD. The strongest pharmacogenetic interaction was observed for rs7917983 [synergy index 3.37 (95% CI 1.72-6.59), P=5.0×10, PACT=0.03], which was associated with increased NOD risk in hydrochlorothiazide-treated patients [odds ratio 1.53 (1.04-2.25), P=0.03] and decreased NOD risk in non hydrochlorothiazide-treated patients [odds ratio 0.48 (0.27-0.86), P=0.02]. The TCF7L2 SNP rs4506565, previously associated with diabetes, showed a similar, significant pharmacogenetic association. CONCLUSION Our results suggest that hydrochlorothiazide treatment is an environmental risk factor that increases diabetes risk beyond that attributed to TCF7L2 variation in white, hypertensive patients. Further study and replication of our results is needed to confirm pharmacogenetic influences of TCF7L2 SNPs on thiazide-induced NOD.
Collapse
|
44
|
Genetics of type 2 diabetes: insights into the pathogenesis and its clinical application. BIOMED RESEARCH INTERNATIONAL 2014; 2014:926713. [PMID: 24864266 PMCID: PMC4016836 DOI: 10.1155/2014/926713] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 03/22/2014] [Indexed: 02/06/2023]
Abstract
With rapidly increasing prevalence, diabetes has become one of the major causes of mortality worldwide. According to the latest studies, genetic information makes substantial contributions towards the prediction of diabetes risk and individualized antidiabetic treatment. To date, approximately 70 susceptibility genes have been identified as being associated with type 2 diabetes (T2D) at a genome-wide significant level (P < 5 × 10−8). However, all the genetic loci identified so far account for only about 10% of the overall heritability of T2D. In addition, how these novel susceptibility loci correlate with the pathophysiology of the disease remains largely unknown. This review covers the major genetic studies on the risk of T2D based on ethnicity and briefly discusses the potential mechanisms and clinical utility of the genetic information underlying T2D.
Collapse
|
45
|
Abstract
Few Type 2 diabetes loci are considered confirmed and replicated across multiple populations. Some genes that have become accepted as contributors to diabetes risk include: calpain 10, peroxisome proliferator-activated receptor-gamma, ATP-sensitive inwardly rectifying potassium channel subunit Kir6.2, hepatocyte nuclear factor 4alpha and hepatic transcription factor 1. While numerous reports of new diabetes loci enter the literature on a regular basis, this review focuses on selected novel associations reported within the last 12 months. In particular, we highlight recent reports of associations between Type 2 diabetes and the transcription factor 7-like 2 gene, associations with micro-opioid receptor and supressor of cytokine signaling 2 genes, and expression and functional analyses of adipokines vaspin and retinol binding protein 4. These new results provide insights into possible mechanisms influencing disease susceptibility and thus new diagnostic and therapeutic opportunities for Type 2 diabetes.
Collapse
Affiliation(s)
- Michèle M Sale
- Internal Medicine, Wake Forest University School of Medicine, Center for Human Genomics, Medical Center Blvd, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
46
|
Ramos RB, Wiltgen D, Spritzer PM. Polymorphisms of TCF7L2 gene in South Brazilian women with polycystic ovary syndrome: a cross-sectional study. Eur J Endocrinol 2013; 169:569-76. [PMID: 23935130 DOI: 10.1530/eje-13-0105] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE To assess whether TCF7L2 single nucleotide polymorphisms rs7903146 C/T and rs11196236 C/T are associated with polycystic ovary syndrome (PCOS) in South Brazilian women. DESIGN Cross-sectional study. METHODS Two hundred PCOS patients and 102 non-hirsute, ovulatory controls were genotyped by real-time PCR. Haplotypes were constructed from the combination of both polymorphisms. Frequencies were inferred using the PHASE 2.1.1 software. RESULTS AND CONCLUSIONS The distribution of rs7903146 (PCOS, 54.4% CC; 28.5% CT; 17.1% TT; controls, 51.0% CC; 37.0% CT; 12.0% TT) and rs11196236 (PCOS, 4.3% CC; 33.5% CT; 62.2% TT; controls, 3.2% CC; 35.5% CT; 61.3% TT) was similar between the groups. rs7903146 and rs11196236 were not in linkage disequilibrium (|D'|=0.34; r(2)=0.07). PCOS participants were younger, with higher age-adjusted BMI, waist circumference, blood pressure, triglycerides, insulin, homeostasis model assessment index to estimate insulin resistance and total testosterone, and lower HDL-C and sex hormone binding globulin vs controls. In PCOS, no differences between genotypes and haplotypes were found for clinical and metabolic variables. However, for each T (rs7903146) and T (rs11196236) allele added to the haplotypes, a variation of 5.87 cm in waist (P trend=0.01), 10.7 mg/dl in total cholesterol (P trend=0.03), and 10.3 mg/dl in LDL-C (P trend=0.01) was recorded. TCF7L2 variants are probably not implicated in PCOS development in South Brazilian women.
Collapse
Affiliation(s)
- Ramon Bossardi Ramos
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, 90035-003 Porto Alegre, RS, Brazil
| | | | | |
Collapse
|
47
|
Xu M, Bi Y, Cui B, Hong J, Wang W, Ning G. The new perspectives on genetic studies of type 2 diabetes and thyroid diseases. Curr Genomics 2013; 14:33-48. [PMID: 23997649 PMCID: PMC3580778 DOI: 10.2174/138920213804999138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 11/16/2012] [Accepted: 11/19/2012] [Indexed: 12/18/2022] Open
Abstract
Recently, genome-wide association studies (GWAS) have led to the discovery of hundreds of susceptibility loci that are associated with complex metabolic diseases, such as type 2 diabetes and hyperthyroidism. The majority of the susceptibility loci are common across different races or populations; while some of them show ethnicity-specific distribution. Though the abundant novel susceptibility loci identified by GWAS have provided insight into biology through the discovery of new genes or pathways that were previously not known, most of them are in introns and the associated variants cumulatively explain only a small fraction of total heritability. Here we reviewed the genetic studies on the metabolic disorders, mainly type 2 diabetes and hyperthyroidism, including candidate genes-based findings and more recently the GWAS discovery; we also included the clinical relevance of these novel loci and the gene-environmental interactions. Finally, we discussed the future direction about the genetic study on the exploring of the pathogenesis of the metabolic diseases.
Collapse
Affiliation(s)
- Min Xu
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, E-Institute of Shanghai Universities, Shanghai, China
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Type 2 diabetes (T2D) is the result of interaction between environmental factors and a strong hereditary component. We review the heritability of T2D as well as the history of genetic and genomic research in this area. Very few T2D risk genes were identified using candidate gene and linkage-based studies, but the advent of genome-wide association studies has led to the identification of multiple genes, including several that were not previously known to play any role in T2D. Highly replicated genes, for example TCF7L2, KCNQ1 and KCNJ11, are discussed in greater detail. Taken together, the genetic loci discovered to date explain only a small proportion of the observed heritability. We discuss possible explanations for this “missing heritability”, including the role of rare variants, gene-environment interactions and epigenetics. The clinical utility of current findings and avenues of future research are also discussed.
Collapse
|
49
|
Pang D, Smith A, Humphries S. Functional analysis of TCF7L2 genetic variants associated with type 2 diabetes. Nutr Metab Cardiovasc Dis 2013; 23:550-556. [PMID: 22402060 PMCID: PMC3778915 DOI: 10.1016/j.numecd.2011.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 11/30/2011] [Accepted: 12/28/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Common non-coding variations within the TCF7L2 locus have a strong influence on type 2 diabetes (T2D) susceptibility through uncharacterised mechanisms. An islet-specific functional polymorphism has been identified, although this does not explain the association between genotype and gene expression in other cell types. This study sought to identify these other functional TCF7L2 variants. METHODS AND RESULTS Alternative splicing and gene expression from TCF7L2 was examined from peripheral blood mononuclear cells from 100 healthy Caucasians using two T2D-associated SNPs, rs7903146 and rs12255372. Electrophoretic mobility shift assays and luciferase reporter assays were performed with these SNPs and those in strong LD to determine potential SNP functionality. Individuals homozygous for rs7903146 and rs12255372 T2D risk alleles (TT/TT) expressed 2.6-fold greater levels of TCF7L2 mRNA compared to individuals homozygous for the non-risk alleles (CC/GG, p = 0.006), although differentially spliced TCF7L2 transcripts did not differ by T2D risk-associated genotype. From SNPs identified to be in strong LD with the T2D-associated SNPs, rs7903146 and rs12255372, five (rs4132670, rs4506565, rs7903146, rs7901695, rs17747324) demonstrated allele-specific binding in electrophoretic mobility shift assays (EMSA). In luciferase reporter assays, rs4132670 exhibited 1.3-fold higher levels of enhancer activity in the Huh7 cell line (p = 3.8 × 10(-5)) and 2-fold higher levels in a WiDr colon carcinoma cell line (p = 0.008). CONCLUSIONS These results suggest that rs4132670, located in a region of chromatin accessibility, is a non-tissue-specific candidate functional SNP that has the potential to play a role in TCF7L2 gene expression and T2D risk.
Collapse
|
50
|
Tönjes A, Kovacs P. SGLT2: a potential target for the pharmacogenetics of Type 2 diabetes? Pharmacogenomics 2013; 14:825-33. [DOI: 10.2217/pgs.13.61] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The kidney has attracted the attention of diabetologists as an organ involved in the regulation of glucose homeostasis not only by gluconeogenesis, but also by renal glucose excretion. Sodium–glucose cotransporters (SGLTs), particularly SGLT2, are responsible for reabsorption of up to 99% of the filtered glucose. SGLT2 is coded by the SLC5A2 gene, which maps on chromosome 16. Pharmacological reduction of tubular glucose reabsorption results in improved glycemic control in Type 2 diabetic patients. Since the SGLTs reabsorb most of the filtered glucose (90%), it is not surprising that mutations in SLC5A2 cause familial renal glucosuria. A recent study pointed out a possible role of common genetic variation in SLC5A2 in the control of glucose homeostasis. SLC5A2 polymorphisms might therefore represent potential candidates for pharmacogenomic studies targeting the impact of these variants on the efficacy of antidiabetic treatment that is based on inhibition of SGLT2 activity.
Collapse
Affiliation(s)
- Anke Tönjes
- University of Leipzig, Medical Department, Liebigstraße 21, 04103 Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig, Germany
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig, Germany
- University of Leipzig, Medical Department, Liebigstraße 21, 04103 Leipzig, Germany
| |
Collapse
|