1
|
Díaz-García JD, Leyva-Leyva M, Sánchez-Aguillón F, de León-Bautista MP, Fuentes-Venegas A, Torres-Viloria A, Tenorio-Aguirre EK, Morales-Lázaro SL, Olivo-Díaz A, González-Ramírez R. Association Study of CACNA1D, KCNJ11, KCNQ1, and CACNA1E Single-Nucleotide Polymorphisms with Type 2 Diabetes Mellitus. Int J Mol Sci 2024; 25:9196. [PMID: 39273144 PMCID: PMC11395491 DOI: 10.3390/ijms25179196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex chronic disease characterized by decreased insulin secretion and the development of insulin resistance. Previous genome-wide association studies demonstrated that single-nucleotide polymorphisms (SNPs) present in genes coding for ion channels involved in insulin secretion increase the risk of developing this disease. We determined the association of 16 SNPs found in CACNA1D, KCNQ1, KCNJ11, and CACNA1E genes and the increased probability of developing T2DM. In this work, we performed a case-control study in 301 Mexican adults, including 201 cases with diabetes and 100 controls without diabetes. Our findings indicate a moderate association between T2DM and the C allele, and the C/C genotype of rs312480 within CACNA1D. The CAG haplotype surprisingly showed a protective effect, whereas the CAC and CGG haplotypes have a strong association with T2DM. The C allele and C/C genotype of rs5219 were significantly associated with diabetes. Also, an association was observed between diabetes and the A allele and the A/A genotype of rs3753737 and rs175338 in CACNA1E. The TGG and CGA haplotypes were also found to be significantly associated. The findings of this study indicate that the SNPs examined could serve as a potential diagnostic tool and contribute to the susceptibility of the Mexican population to this disease.
Collapse
Affiliation(s)
- Juan Daniel Díaz-García
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Margarita Leyva-Leyva
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Fabiola Sánchez-Aguillón
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Mercedes Piedad de León-Bautista
- Escuela de Medicina, Universidad Vasco de Quiroga, Morelia 58090, Mexico;
- Laboratorio de Enfermedades Infecciosas y Genómica (INEX LAB), Morelia 58280, Mexico
| | - Abel Fuentes-Venegas
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Alfredo Torres-Viloria
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Erika Karina Tenorio-Aguirre
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Sara Luz Morales-Lázaro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
- Centro de Investigación Sobre el Envejecimiento, CINVESTAV, Mexico City 14330, Mexico
| | - Angélica Olivo-Díaz
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Ricardo González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
- Centro de Investigación Sobre el Envejecimiento, CINVESTAV, Mexico City 14330, Mexico
| |
Collapse
|
2
|
Adadey SM, Mensah JA, Acquah KS, Abugri J, Osei-Yeboah R. Early-onset diabetes in Africa: A mini-review of the current genetic profile. Eur J Med Genet 2023; 66:104887. [PMID: 37995864 DOI: 10.1016/j.ejmg.2023.104887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Early-onset diabetes is poorly diagnosed partly due to its heterogeneity and variable presentations. Although several genes have been associated with the disease, these genes are not well studied in Africa. We sought to identify the major neonatal, early childhood, juvenile, or early-onset diabetes genes in Africa; and evaluate the available molecular methods used for investigating these gene variants. A literature search was conducted on PubMed, Scopus, Africa-Wide Information, and Web of Science databases. The retrieved records were screened and analyzed to identify genetic variants associated with early-onset diabetes. Although 319 records were retrieved, 32 were considered for the current review. Most of these records (22/32) were from North Africa. The disease condition was genetically heterogenous with most cases possessing unique gene variants. We identified 22 genes associated with early-onset diabetes, 9 of which had variants (n = 19) classified as pathogenic or likely pathogenic (PLP). Among the PLP variants, IER3IP1: p.(Leu78Pro) was the variant with the highest number of cases. There was limited data from West Africa, hence the contribution of genetic variability to early-onset diabetes in Africa could not be comprehensively evaluated. It is worth mentioning that most studies were focused on natural products as antidiabetics and only a few studies reported on the genetics of the disease. ABCC8 and KCNJ11 were implicated as major contributors to early-onset diabetes gene networks. Gene ontology analysis of the network associated ion channels, impaired glucose tolerance, and decreased insulin secretions to the disease. Our review highlights 9 genes from which PLP variants have been identified and can be considered for the development of an African diagnostic panel. There is a gap in early-onset diabetes genetic research from sub-Saharan Africa which is much needed to develop a comprehensive, efficient, and cost-effective genetic panel that will be useful in clinical practice on the continent and among the African diasporas.
Collapse
Affiliation(s)
- Samuel Mawuli Adadey
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana; School of Medicine and Health Science, University for Development Studies, Tamale, Ghana.
| | | | - Kojo Sekyi Acquah
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA.
| | - James Abugri
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C.K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana.
| | - Richard Osei-Yeboah
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
3
|
Ferreira G, Santander A, Cardozo R, Chavarría L, Domínguez L, Mujica N, Benítez M, Sastre S, Sobrevia L, Nicolson GL. Nutrigenomics of inward rectifier potassium channels. Biochim Biophys Acta Mol Basis Dis 2023:166803. [PMID: 37406972 DOI: 10.1016/j.bbadis.2023.166803] [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: 03/20/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
Inwardly rectifying potassium (Kir) channels play a key role in maintaining the resting membrane potential and supporting potassium homeostasis. There are many variants of Kir channels, which are usually tetramers in which the main subunit has two trans-membrane helices attached to two N- and C-terminal cytoplasmic tails with a pore-forming loop in between that contains the selectivity filter. These channels have domains that are strongly modulated by molecules present in nutrients found in different diets, such as phosphoinositols, polyamines and Mg2+. These molecules can impact these channels directly or indirectly, either allosterically by modulation of enzymes or via the regulation of channel expression. A particular type of these channels is coupled to cell metabolism and inhibited by ATP (KATP channels, essential for insulin release and for the pathogenesis of metabolic diseases like diabetes mellitus). Genomic changes in Kir channels have a significant impact on metabolism, such as conditioning the nutrients and electrolytes that an individual can take. Thus, the nutrigenomics of ion channels is an important emerging field in which we are attempting to understand how nutrients and diets can affect the activity and expression of ion channels and how genomic changes in such channels may be the basis for pathological conditions that limit nutrition and electrolyte intake. In this contribution we briefly review Kir channels, discuss their nutrigenomics, characterize how different components in the diet affect their function and expression, and suggest how their genomic changes lead to pathological phenotypes that affect diet and electrolyte intake.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay.
| | - Axel Santander
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay
| | - Romina Cardozo
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay
| | - Luisina Chavarría
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay
| | - Lucía Domínguez
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay
| | - Nicolás Mujica
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay
| | - Milagros Benítez
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay
| | - Santiago Sastre
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, CP 11800, Universidad de la Republica, Montevideo, Uruguay; Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo CP 11800, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), Sao Paulo State University (UNESP), Brazil; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, 4029, Queensland, Australia; Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León, Mexico
| | - Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
4
|
Bayer S, Reik A, von Hesler L, Hauner H, Holzapfel C. Association between Genotype and the Glycemic Response to an Oral Glucose Tolerance Test: A Systematic Review. Nutrients 2023; 15:nu15071695. [PMID: 37049537 PMCID: PMC10096950 DOI: 10.3390/nu15071695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
The inter-individual variability of metabolic response to foods may be partly due to genetic variation. This systematic review aims to assess the associations between genetic variants and glucose response to an oral glucose tolerance test (OGTT). Three databases (PubMed, Web of Science, Embase) were searched for keywords in the field of genetics, OGTT, and metabolic response (PROSPERO: CRD42021231203). Inclusion criteria were available data on single nucleotide polymorphisms (SNPs) and glucose area under the curve (gAUC) in a healthy study cohort. In total, 33,219 records were identified, of which 139 reports met the inclusion criteria. This narrative synthesis focused on 49 reports describing gene loci for which several reports were available. An association between SNPs and the gAUC was described for 13 gene loci with 53 different SNPs. Three gene loci were mostly investigated: transcription factor 7 like 2 (TCF7L2), peroxisome proliferator-activated receptor gamma (PPARγ), and potassium inwardly rectifying channel subfamily J member 11 (KCNJ11). In most reports, the associations were not significant or single findings were not replicated. No robust evidence for an association between SNPs and gAUC after an OGTT in healthy persons was found across the identified studies. Future studies should investigate the effect of polygenic risk scores on postprandial glucose levels.
Collapse
Affiliation(s)
- Sandra Bayer
- Institute for Nutritional Medicine, School of Medicine, University Hospital “Klinikum Rechts der Isar”, Technical University of Munich, 80992 Munich, Germany
| | - Anna Reik
- Institute for Nutritional Medicine, School of Medicine, University Hospital “Klinikum Rechts der Isar”, Technical University of Munich, 80992 Munich, Germany
| | - Lena von Hesler
- Institute for Nutritional Medicine, School of Medicine, University Hospital “Klinikum Rechts der Isar”, Technical University of Munich, 80992 Munich, Germany
| | - Hans Hauner
- Institute for Nutritional Medicine, School of Medicine, University Hospital “Klinikum Rechts der Isar”, Technical University of Munich, 80992 Munich, Germany
- Else Kröner-Fresenius-Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Christina Holzapfel
- Institute for Nutritional Medicine, School of Medicine, University Hospital “Klinikum Rechts der Isar”, Technical University of Munich, 80992 Munich, Germany
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, 36037 Fulda, Germany
- Correspondence:
| |
Collapse
|
5
|
From Metabolic Syndrome to Type 2 Diabetes in Youth. CHILDREN 2023; 10:children10030516. [PMID: 36980074 PMCID: PMC10047215 DOI: 10.3390/children10030516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/26/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023]
Abstract
In the frame of metabolic syndrome, type 2 diabetes emerges along a continuum of the risk from the clustering of all its components, namely visceral obesity, high blood pressure and lipids, and impaired glucose homeostasis. Insulin resistance is the hallmark common to all the components and, in theory, is a reversible condition. Nevertheless, the load that this condition can exert on the β-cell function at the pubertal transition is such as to determine its rapid and irreversible deterioration leading to plain diabetes. The aim of this review is to highlight, in the context of metabolic syndrome, age-specific risk factors that lead to type 2 diabetes onset in youth; resume age specific screening and diagnostic criteria; and anticipate potential for treatment. Visceral obesity and altered lipid metabolism are robust grounds for the development of the disease. Genetic differences in susceptibility to hampered β-cell function in the setting of obesity and insulin resistance largely explain why some adolescents with obesity do develop diabetes at a young age and some others do not. Lifestyle intervention with a healthy diet and physical activity remains the pillar of the type 2 diabetes treatment in youth. As to the pharmacological management, metformin and insulin have failed to rescue β-cell function and to ensure long-lasting glycemic control in youth. A new era might start with the approval for use in pediatric age of drugs largely prescribed in adults, such as dipeptidyl peptidase-4 and sodium-dependent glucose transport inhibitors, and of new weight-lowering drugs in the pipeline such as single and multiple agonists of the glucagon-like peptide 1 receptor. The latter drugs can have tremendous impact on the natural history of the disease. By treating diabetes, they will reduce the burden of all the metabolic abnormalities belonging to the syndrome while causing a tremendous weight loss hitherto never seen before.
Collapse
|
6
|
Abstract
Ubiquitously expressed throughout the body, ATP-sensitive potassium (KATP) channels couple cellular metabolism to electrical activity in multiple tissues; their unique assembly as four Kir6 pore-forming subunits and four sulfonylurea receptor (SUR) subunits has resulted in a large armory of selective channel opener and inhibitor drugs. The spectrum of monogenic pathologies that result from gain- or loss-of-function mutations in these channels, and the potential for therapeutic correction of these pathologies, is now clear. However, while available drugs can be effective treatments for specific pathologies, cross-reactivity with the other Kir6 or SUR subfamily members can result in drug-induced versions of each pathology and may limit therapeutic usefulness. This review discusses the background to KATP channel physiology, pathology, and pharmacology and considers the potential for more specific or effective therapeutic agents.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
7
|
O'Sullivan JW, Ashley EA, Elliott PM. Polygenic risk scores for the prediction of cardiometabolic disease. Eur Heart J 2023; 44:89-99. [PMID: 36478054 DOI: 10.1093/eurheartj/ehac648] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 08/28/2022] [Accepted: 10/27/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiometabolic diseases contribute more to global morbidity and mortality than any other group of disorders. Polygenic risk scores (PRSs), the weighted summation of individually small-effect genetic variants, represent an advance in our ability to predict the development and complications of cardiometabolic diseases. This article reviews the evidence supporting the use of PRS in seven common cardiometabolic diseases: coronary artery disease (CAD), stroke, hypertension, heart failure and cardiomyopathies, obesity, atrial fibrillation (AF), and type 2 diabetes mellitus (T2DM). Data suggest that PRS for CAD, AF, and T2DM consistently improves prediction when incorporated into existing clinical risk tools. In other areas such as ischaemic stroke and hypertension, clinical application appears premature but emerging evidence suggests that the study of larger and more diverse populations coupled with more granular phenotyping will propel the translation of PRS into practical clinical prediction tools.
Collapse
Affiliation(s)
- Jack W O'Sullivan
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Euan A Ashley
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Perry M Elliott
- UCL Institute of Cardiovascular Science, Gower Street, London WC1E 6BT, UK
- St. Bartholomew's Hospital, W Smithfield, London EC1A 7BE, UK
| |
Collapse
|
8
|
Connally NJ, Nazeen S, Lee D, Shi H, Stamatoyannopoulos J, Chun S, Cotsapas C, Cassa CA, Sunyaev SR. The missing link between genetic association and regulatory function. eLife 2022; 11:e74970. [PMID: 36515579 PMCID: PMC9842386 DOI: 10.7554/elife.74970] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
The genetic basis of most traits is highly polygenic and dominated by non-coding alleles. It is widely assumed that such alleles exert small regulatory effects on the expression of cis-linked genes. However, despite the availability of gene expression and epigenomic datasets, few variant-to-gene links have emerged. It is unclear whether these sparse results are due to limitations in available data and methods, or to deficiencies in the underlying assumed model. To better distinguish between these possibilities, we identified 220 gene-trait pairs in which protein-coding variants influence a complex trait or its Mendelian cognate. Despite the presence of expression quantitative trait loci near most GWAS associations, by applying a gene-based approach we found limited evidence that the baseline expression of trait-related genes explains GWAS associations, whether using colocalization methods (8% of genes implicated), transcription-wide association (2% of genes implicated), or a combination of regulatory annotations and distance (4% of genes implicated). These results contradict the hypothesis that most complex trait-associated variants coincide with homeostatic expression QTLs, suggesting that better models are needed. The field must confront this deficit and pursue this 'missing regulation.'
Collapse
Affiliation(s)
- Noah J Connally
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Sumaiya Nazeen
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Department of Neurology, Harvard Medical SchoolBostonUnited States
| | - Daniel Lee
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Huwenbo Shi
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
- Department of Epidemiology, Harvard T.H. Chan School of Public HealthBostonUnited States
| | | | - Sung Chun
- Division of Pulmonary Medicine, Boston Children’s HospitalBostonUnited States
| | - Chris Cotsapas
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
- Department of Neurology, Yale Medical SchoolNew HavenUnited States
- Department of Genetics, Yale Medical SchoolNew HavenUnited States
| | - Christopher A Cassa
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Shamil R Sunyaev
- Department of Biomedical Informatics, Harvard Medical SchoolBostonUnited States
- Brigham and Women’s Hospital, Division of Genetics, Harvard Medical SchoolBostonUnited States
- Program in Medical and Population Genetics, Broad Institute of MIT and HarvardCambridgeUnited States
| |
Collapse
|
9
|
Moazzam-Jazi M, Najd-Hassan-Bonab L, Masjoudi S, Tohidi M, Hedayati M, Azizi F, Daneshpour MS. Risk of type 2 diabetes and KCNJ11 gene polymorphisms: a nested case-control study and meta-analysis. Sci Rep 2022; 12:20709. [PMID: 36456687 PMCID: PMC9715540 DOI: 10.1038/s41598-022-24931-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Due to the central role in insulin secretion, the potassium inwardly-rectifying channel subfamily J member 11 (KCNJ11) gene is one of the essential genes for type 2 diabetes (T2D) predisposition. However, the relevance of this gene to T2D development is not consistent among diverse populations. In the current study, we aim to capture the possible association of common KCNJ11 variants across Iranian adults, followed by a meta-analysis. We found that the tested variants of KCNJ11 have not contributed to T2D incidence in Iranian adults, consistent with similar insulin secretion levels among individuals with different genotypes. The integration of our results with 72 eligible published case-control studies (41,372 cases and 47,570 controls) as a meta-analysis demonstrated rs5219 and rs5215 are significantly associated with the increased T2D susceptibility under different genetic models. Nevertheless, the stratified analysis according to ethnicity showed rs5219 is involved in the T2D risk among disparate populations, including American, East Asian, European, and Greater Middle Eastern, but not South Asian. Additionally, the meta-regression analysis demonstrated that the sample size of both case and control groups was significantly associated with the magnitude of pooled genetic effect size. The present study can expand our knowledge about the KCNJ11 common variant's contributions to T2D incidence, which is valuable for designing SNP-based panels for potential clinical applications in precision medicine. It also highlights the importance of similar sample sizes for avoiding high heterogeneity and conducting a more precise meta-analysis.
Collapse
Affiliation(s)
- Maryam Moazzam-Jazi
- Cellular, and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Najd-Hassan-Bonab
- Cellular, and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajedeh Masjoudi
- Cellular, and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Tohidi
- Prevention of Metabolic Disorder Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular, and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Laakso M, Fernandes Silva L. Genetics of Type 2 Diabetes: Past, Present, and Future. Nutrients 2022; 14:nu14153201. [PMID: 35956377 PMCID: PMC9370092 DOI: 10.3390/nu14153201] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 02/01/2023] Open
Abstract
Diabetes has reached epidemic proportions worldwide. Currently, approximately 537 million adults (20–79 years) have diabetes, and the total number of people with diabetes is continuously increasing. Diabetes includes several subtypes. About 80% of all cases of diabetes are type 2 diabetes (T2D). T2D is a polygenic disease with an inheritance ranging from 30 to 70%. Genetic and environment/lifestyle factors, especially obesity and sedentary lifestyle, increase the risk of T2D. In this review, we discuss how studies on the genetics of diabetes started, how they expanded when genome-wide association studies and exome and whole-genome sequencing became available, and the current challenges in genetic studies of diabetes. T2D is heterogeneous with respect to clinical presentation, disease course, and response to treatment, and has several subgroups which differ in pathophysiology and risk of micro- and macrovascular complications. Currently, genetic studies of T2D focus on these subgroups to find the best diagnoses and treatments for these patients according to the principles of precision medicine.
Collapse
Affiliation(s)
- Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, 70210 Kuopio, Finland
- Correspondence: ; Tel.: +358-40-672-3338
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| |
Collapse
|
11
|
Nichols CG, York NW, Remedi MS. ATP-Sensitive Potassium Channels in Hyperinsulinism and Type 2 Diabetes: Inconvenient Paradox or New Paradigm? Diabetes 2022; 71:367-375. [PMID: 35196393 PMCID: PMC8893938 DOI: 10.2337/db21-0755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/28/2021] [Indexed: 11/13/2022]
Abstract
Secretion of insulin from pancreatic β-cells is complex, but physiological glucose-dependent secretion is dominated by electrical activity, in turn controlled by ATP-sensitive potassium (KATP) channel activity. Accordingly, loss-of-function mutations of the KATP channel Kir6.2 (KCNJ11) or SUR1 (ABCC8) subunit increase electrical excitability and secretion, resulting in congenital hyperinsulinism (CHI), whereas gain-of-function mutations cause underexcitability and undersecretion, resulting in neonatal diabetes mellitus (NDM). Thus, diazoxide, which activates KATP channels, and sulfonylureas, which inhibit KATP channels, have dramatically improved therapies for CHI and NDM, respectively. However, key findings do not fit within this simple paradigm: mice with complete absence of β-cell KATP activity are not hyperinsulinemic; instead, they are paradoxically glucose intolerant and prone to diabetes, as are older human CHI patients. Critically, despite these advances, there has been little insight into any role of KATP channel activity changes in the development of type 2 diabetes (T2D). Intriguingly, the CHI progression from hypersecretion to undersecretion actually mirrors the classical response to insulin resistance in the progression of T2D. In seeking to explain the progression of CHI, multiple lines of evidence lead us to propose that underlying mechanisms are also similar and that development of T2D may involve loss of KATP activity.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Nathaniel W York
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Maria S Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Division of Endocrinology Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
12
|
Venkatachalapathy P, Padhilahouse S, Sellappan M, Subramanian T, Kurian SJ, Miraj SS, Rao M, Raut AA, Kanwar RK, Singh J, Khadanga S, Mondithoka S, Munisamy M. Pharmacogenomics and Personalized Medicine in Type 2 Diabetes Mellitus: Potential Implications for Clinical Practice. Pharmgenomics Pers Med 2021; 14:1441-1455. [PMID: 34803393 PMCID: PMC8598203 DOI: 10.2147/pgpm.s329787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/04/2021] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is the most common form of diabetes, and is rising in incidence with widespread prevalence. Multiple gene variants are associated with glucose homeostasis, complex T2DM pathogenesis, and its complications. Exploring more effective therapeutic strategies for patients with diabetes is crucial. Pharmacogenomics has made precision medicine possible by allowing for individualized drug therapy based on a patient's genetic and genomic information. T2DM is treated with various classes of oral hypoglycemic agents, such as biguanides, sulfonylureas, thiazolidinediones, meglitinides, DPP4 inhibitors, SGLT2 inhibitors, α-glucosidase inhibitors, and GLP1 analogues, which exhibit various pharmacogenetic variants. Although genomic interventions in monogenic diabetes have been implemented in clinical practice, they are still in the early stages for complex polygenic disorders, such as T2DM. Precision DM medicine has the potential to be effective in personalized therapy for those suffering from various forms of DM, such as T2DM. With recent developments in genetic techniques, the application of candidate-gene studies, large-scale genotyping investigations, genome-wide association studies, and "multiomics" studies has begun to produce results that may lead to changes in clinical practice. Enhanced knowledge of the genetic architecture of T2DM presents a bigger translational potential. This review summarizes the genetics and pathophysiology of T2DM, candidate-gene approaches, genome-wide association studies, personalized medicine, clinical relevance of pharmacogenetic variants associated with oral hypoglycemic agents, and paths toward personalized diabetology.
Collapse
Affiliation(s)
| | - Sruthi Padhilahouse
- Department of Pharmacy Practice, Karpagam College of Pharmacy, Coimbatore, Tamilnadu, India
| | - Mohan Sellappan
- Department of Pharmacy Practice, Karpagam College of Pharmacy, Coimbatore, Tamilnadu, India
| | | | - Shilia Jacob Kurian
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sonal Sekhar Miraj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ashwin Ashok Raut
- Translational Medicine Centre, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Rupinder Kaur Kanwar
- Translational Medicine Centre, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Jitendra Singh
- Translational Medicine Centre, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Sagar Khadanga
- Department of General Medicine, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Sukumar Mondithoka
- Department of General Medicine, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Murali Munisamy
- Translational Medicine Centre, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| |
Collapse
|
13
|
Sachse G, Haythorne E, Hill T, Proks P, Joynson R, Terrón-Expósito R, Bentley L, Tucker SJ, Cox RD, Ashcroft FM. The KCNJ11-E23K Gene Variant Hastens Diabetes Progression by Impairing Glucose-Induced Insulin Secretion. Diabetes 2021; 70:1145-1156. [PMID: 33568422 DOI: 10.2337/db20-0691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 02/05/2021] [Indexed: 11/13/2022]
Abstract
The ATP-sensitive K+ (KATP) channel controls blood glucose levels by coupling glucose metabolism to insulin secretion in pancreatic β-cells. E23K, a common polymorphism in the pore-forming KATP channel subunit (KCNJ11) gene, has been linked to increased risk of type 2 diabetes. Understanding the risk-allele-specific pathogenesis has the potential to improve personalized diabetes treatment, but the underlying mechanism has remained elusive. Using a genetically engineered mouse model, we now show that the K23 variant impairs glucose-induced insulin secretion and increases diabetes risk when combined with a high-fat diet (HFD) and obesity. KATP-channels in β-cells with two K23 risk alleles (KK) showed decreased ATP inhibition, and the threshold for glucose-stimulated insulin secretion from KK islets was increased. Consequently, the insulin response to glucose and glycemic control was impaired in KK mice fed a standard diet. On an HFD, the effects of the KK genotype were exacerbated, accelerating diet-induced diabetes progression and causing β-cell failure. We conclude that the K23 variant increases diabetes risk by impairing insulin secretion at threshold glucose levels, thus accelerating loss of β-cell function in the early stages of diabetes progression.
Collapse
Affiliation(s)
- Gregor Sachse
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K.
| | - Elizabeth Haythorne
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Thomas Hill
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Peter Proks
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
- Department of Physics, University of Oxford, Oxford, U.K
| | - Russell Joynson
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, U.K
| | - Raul Terrón-Expósito
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| | - Liz Bentley
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, U.K
| | | | - Roger D Cox
- Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Oxfordshire, U.K
| | - Frances M Ashcroft
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, U.K
| |
Collapse
|
14
|
Kim M, Huda MN, O'Connor A, Albright J, Durbin-Johnson B, Bennett BJ. Hepatic transcriptional profile reveals the role of diet and genetic backgrounds on metabolic traits in female progenitor strains of the Collaborative Cross. Physiol Genomics 2021; 53:173-192. [PMID: 33818129 PMCID: PMC8424536 DOI: 10.1152/physiolgenomics.00140.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/28/2022] Open
Abstract
Mice have provided critical mechanistic understandings of clinical traits underlying metabolic syndrome (MetSyn) and susceptibility to MetSyn in mice is known to vary among inbred strains. We investigated the diet- and strain-dependent effects on metabolic traits in the eight Collaborative Cross (CC) founder strains (A/J, C57BL/6J, 129S1/SvImJ, NOD/ShiLtJ, NZO/HILtJ, CAST/EiJ, PWK/PhJ, and WSB/EiJ). Liver transcriptomics analysis showed that both atherogenic diet and host genetics have profound effects on the liver transcriptome, which may be related to differences in metabolic traits observed between strains. We found strain differences in circulating trimethylamine N-oxide (TMAO) concentration and liver triglyceride content, both of which are traits associated with metabolic diseases. Using a network approach, we identified a module of transcripts associated with TMAO and liver triglyceride content, which was enriched in functional pathways. Interrogation of the module related to metabolic traits identified NADPH oxidase 4 (Nox4), a gene for a key enzyme in the production of reactive oxygen species, which showed a strong association with plasma TMAO and liver triglyceride. Interestingly, Nox4 was identified as the highest expressed in the C57BL/6J and NZO/HILtJ strains and the lowest expressed in the CAST/EiJ strain. Based on these results, we suggest that there may be genetic variation in the contribution of Nox4 to the regulation of plasma TMAO and liver triglyceride content. In summary, we show that liver transcriptomic analysis identified diet- or strain-specific pathways for metabolic traits in the Collaborative Cross (CC) founder strains.
Collapse
Affiliation(s)
- Myungsuk Kim
- Department of Nutrition, University of California, Davis, California
- USDA-ARS-Western Human Nutrition Research Center, Davis, California
| | - M Nazmul Huda
- Department of Nutrition, University of California, Davis, California
- USDA-ARS-Western Human Nutrition Research Center, Davis, California
| | - Annalouise O'Connor
- Nutrition Research Institute, University of North Carolina, Kannapolis, North Carolina
| | - Jody Albright
- Nutrition Research Institute, University of North Carolina, Kannapolis, North Carolina
| | | | - Brian J Bennett
- Department of Nutrition, University of California, Davis, California
- USDA-ARS-Western Human Nutrition Research Center, Davis, California
| |
Collapse
|
15
|
Denyer AL, Catchpole B, Davison LJ. Genetics of canine diabetes mellitus part 2: Current understanding and future directions. Vet J 2021; 270:105612. [PMID: 33641811 DOI: 10.1016/j.tvjl.2021.105612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023]
Abstract
Part 1 of this 2-part review outlined the importance of disease classification in diabetes genetic studies, as well as the ways in which genetic variants may contribute to risk of a complex disease within an individual, or within a particular group of individuals. Part 2, presented here, describes in more detail our current understanding of the genetics of canine diabetes mellitus compared to our knowledge of the human disease. Ongoing work to improve our knowledge, using new technologies, is also introduced.
Collapse
Affiliation(s)
- Alice L Denyer
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - Brian Catchpole
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - Lucy J Davison
- Department of Clinical Sciences and Services, Royal Veterinary College, Hatfield, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | |
Collapse
|
16
|
Abstract
With change in global concern toward food quality over food quantity, consumer concern and choice of healthy food has become a matter of prime importance. It gave rise to concept of “personalized or precision nutrition”. The theory behind personalization of nutrition is supported by multiple factors including advances in food analytics, nutrition based diseases and public health programs, increasing use of information technology in nutrition science, concept of gene-diet interaction and growing consumer capacity or concern by better and healthy foods. The advances in “omics” tools and related analytical techniques have resulted into tremendous scope of their application in nutrition science. As a consequence, a better understanding of underlying interaction between diet and individual is expected with addressing of key challenges for successful implementation of this science. In this chapter, the above aspects are discussed to get an insight into driving factors for increasing concern in personalized nutrition.
Collapse
|
17
|
Goenka V, Borkar T, Desai A, Das RK. Therapeutic potential of mesenchymal stem cells in treating both types of diabetes mellitus and associated diseases. J Diabetes Metab Disord 2020; 19:1979-1993. [PMID: 33520872 PMCID: PMC7843693 DOI: 10.1007/s40200-020-00647-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/24/2020] [Indexed: 10/23/2022]
Abstract
Diabetes mellitus is a common lifestyle disease which can be classified into type 1 diabetes mellitus and type 2 diabetes mellitus. While both result in hyperglycemia due to lack of insulin action and further associated chronic ailments, there is a marked distinction in the cause for each type due to which both require a different prophylaxis. As observed, type 1 diabetes is caused due to the autoimmune action of the body resulting in the destruction of pancreatic islet cells. On the other hand, type 2 diabetes is caused either due to insulin resistance of target cells or lack of insulin production as per physiological requirements. Attempts to cure the disease have been made by bringing drastic changes in the patients' lifestyle; parenteral administration of insulin; prescription of drugs such as biguanides, meglitinides, and amylin; pancreatic transplantation; and immunotherapy. While these attempts cause a certain degree of relief to the patient, none of these can cure diabetes mellitus. However, a new treatment strategy led by the discovery of mesenchymal stem cells and their unique immunomodulatory and multipotent properties has inspired therapies to treat diabetes by essentially reversing the conditions causing the disease. The current review aims to enumerate the role of various mesenchymal stem cells and the different approaches to treat both types of diabetes and its associated diseases as well.
Collapse
Affiliation(s)
- Vidul Goenka
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - Tanhai Borkar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - Aska Desai
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu India
| | - Raunak Kumar Das
- Centre for Biomaterials, Cellular and Molecular Theranostics, Vellore Institute of Technology, Vellore, Tamil Nadu India
| |
Collapse
|
18
|
Naseri R, Navabi SJ, Samimi Z, Mishra AP, Nigam M, Chandra H, Olatunde A, Tijjani H, Morais-Urano RP, Farzaei MH. Targeting Glycoproteins as a therapeutic strategy for diabetes mellitus and its complications. Daru 2020; 28:333-358. [PMID: 32006343 PMCID: PMC7095136 DOI: 10.1007/s40199-020-00327-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Glycoproteins are organic compounds formed from proteins and carbohydrates, which are found in many parts of the living systems including the cell membranes. Furthermore, impaired metabolism of glycoprotein components plays the main role in the pathogenesis of diabetes mellitus. The aim of this study is to investigate the influence of glycoprotein levels in the treatment of diabetes mellitus. METHODS All relevant papers in the English language were compiled by searching electronic databases, including Scopus, PubMed and Cochrane library. The keywords of glycoprotein, diabetes mellitus, glycan, glycosylation, and inhibitor were searched until January 2019. RESULTS Glycoproteins are pivotal elements in the regulation of cell proliferation, growth, maturation and signaling pathways. Moreover, they are involved in drug binding, drug transportation, efflux of chemicals and stability of therapeutic proteins. These functions, structure, composition, linkages, biosynthesis, significance and biological effects are discussed as related to their use as a therapeutic strategy for the treatment of diabetes mellitus and its complications. CONCLUSIONS The findings revealed several chemical and natural compounds have significant beneficial effects on glycoprotein metabolism. The comprehension of glycoprotein structure and functions are very essential and inevitable to enhance the knowledge of glycoengineering for glycoprotein-based therapeutics as may be required for the treatment of diabetes mellitus and its associated complications. Graphical abstract.
Collapse
Affiliation(s)
- Rozita Naseri
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Jafar Navabi
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abhay Prakash Mishra
- Department of Pharmaceutical Chemistry, Hemwati Nandan Bahuguna Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand, 246174, India.
| | - Manisha Nigam
- Department of Biochemistry, Hemwati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Harish Chandra
- Department of Microbiology, Gurukul Kangri Vishwavidhyalya, Haridwar, Uttarakhand, 249404, India
| | - Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauchi, Nigeria
| | - Habibu Tijjani
- Natural Product Research Laboratory, Department of Biochemistry, Bauchi State University, Gadau, Nigeria
| | - Raquel P Morais-Urano
- Instituto de Química de São Carlos, Universidade de São Paulo, 13560-970, São Carlos, SP, Brasil
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Wang X, Wei C, Zhang Z, Liu D, Guo Y, Sun G, Wang Y, Li H, Tian Y, Kang X, Han R, Li Z. Association of growth traits with a structural variation downstream of the KCNJ11 gene: a large population-based study in chickens. Br Poult Sci 2020; 61:320-327. [PMID: 32008360 DOI: 10.1080/00071668.2020.1724878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
1. The potassium voltage-gated channel subfamily J member 11 gene (KCNJ11) is involved in the insulin secretion pathway. Studies have shown that mutation in this gene is associated with muscle weakness. The objective of the present study was to establish the association between KCNJ11 gene polymorphism and chicken growth performance and to analyse its expression pattern. 2. A novel 163-bp insertion/deletion (indel) polymorphism was identified in the region downstream of the KCNJ11 gene in 2330 individuals from ten populations by polymerase chain reaction (PCR). An F2 resource population was used to investigate the genetic effects of the chicken KCNJ11 gene. Association analysis showed that the indel was significantly associated with chicken growth traits and that the phenotypic value of the ins-ins (II) genotype is higher than that of the ins-del (ID) and del-del (DD) genotypes. 3. Gene expression for different genotypes showed that birds carrying the II allele had a higher expression level than the DD genotypes. Analysis of tissue and spatiotemporal expression patterns indicated that the KCNJ11 gene was highly expressed in muscle tissues, with the highest levels in muscle tissue at one week of age, and that a 10% crude protein diet reduced the expression of this gene, average daily gain and muscle fibre diameter. 4. The results suggested that this novel 163-bp indel has the potential to become a new target for marker-assisted selection.
Collapse
Affiliation(s)
- X Wang
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - C Wei
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - Z Zhang
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - D Liu
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - Y Guo
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - G Sun
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - Y Wang
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - H Li
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - Y Tian
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - X Kang
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - R Han
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| | - Z Li
- Department of Animal Breeding and Genetics, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China.,Henan Innovative Engineering Research Center of Poultry Germplasm Resource, No.15 Longzihu University Area, Zhengdong New District, College of Animal Science and Veterinary Medicine, Henan Agricultural University , Zhengzhou, China
| |
Collapse
|
20
|
Khatami F, Mohajeri-Tehrani MR, Tavangar SM. The Importance of Precision Medicine in Type 2 Diabetes Mellitus (T2DM): From Pharmacogenetic and Pharmacoepigenetic Aspects. Endocr Metab Immune Disord Drug Targets 2020; 19:719-731. [PMID: 31122183 DOI: 10.2174/1871530319666190228102212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/18/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Type 2 Diabetes Mellitus (T2DM) is a worldwide disorder as the most important challenges of health-care systems. Controlling the normal glycaemia greatly profit long-term prognosis and gives explanation for early, effective, constant, and safe intervention. MATERIAL AND METHODS Finding the main genetic and epigenetic profile of T2DM and the exact molecular targets of T2DM medications can shed light on its personalized management. The comprehensive information of T2DM was earned through the genome-wide association study (GWAS) studies. In the current review, we represent the most important candidate genes of T2DM like CAPN10, TCF7L2, PPAR-γ, IRSs, KCNJ11, WFS1, and HNF homeoboxes. Different genetic variations of a candidate gene can predict the efficacy of T2DM personalized strategy medication. RESULTS SLCs and AMPK variations are considered for metformin, CYP2C9, KATP channel, CDKAL1, CDKN2A/2B and KCNQ1 for sulphonylureas, OATP1B, and KCNQ1 for repaglinide and the last but not the least ADIPOQ, PPAR-γ, SLC, CYP2C8, and SLCO1B1 for thiazolidinediones response prediction. CONCLUSION Taken everything into consideration, there is an extreme need to determine the genetic status of T2DM patients in some known genetic region before planning the medication strategies.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad R Mohajeri-Tehrani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed M Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
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
|
22
|
Barroso I, McCarthy MI. The Genetic Basis of Metabolic Disease. Cell 2019; 177:146-161. [PMID: 30901536 PMCID: PMC6432945 DOI: 10.1016/j.cell.2019.02.024] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/11/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
Recent developments in genetics and genomics are providing a detailed and systematic characterization of the genetic underpinnings of common metabolic diseases and traits, highlighting the inherent complexity within systems for homeostatic control and the many ways in which that control can fail. The genetic architecture underlying these common metabolic phenotypes is complex, with each trait influenced by hundreds of loci spanning a range of allele frequencies and effect sizes. Here, we review the growing appreciation of this complexity and how this has fostered the implementation of genome-scale approaches that deliver robust mechanistic inference and unveil new strategies for translational exploitation.
Collapse
Affiliation(s)
- Inês Barroso
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| |
Collapse
|
23
|
Mannino GC, Andreozzi F, Sesti G. Pharmacogenetics of type 2 diabetes mellitus, the route toward tailored medicine. Diabetes Metab Res Rev 2019; 35:e3109. [PMID: 30515958 PMCID: PMC6590177 DOI: 10.1002/dmrr.3109] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic disease that has reached the levels of a global epidemic. In order to achieve optimal glucose control, it is often necessary to rely on combination therapy of multiple drugs or insulin because uncontrolled glucose levels result in T2DM progression and enhanced risk of complications and mortality. Several antihyperglycemic agents have been developed over time, and T2DM pharmacotherapy should be prescribed based on suitability for the individual patient's characteristics. Pharmacogenetics is the branch of genetics that investigates how our genome influences individual responses to drugs, therapeutic outcomes, and incidence of adverse effects. In this review, we evaluated the pharmacogenetic evidences currently available in the literature, and we identified the top informative genetic variants associated with response to the most common anti-diabetic drugs: metformin, DPP-4 inhibitors/GLP1R agonists, thiazolidinediones, and sulfonylureas/meglitinides. Overall, we found 40 polymorphisms for each drug class in a total of 71 loci, and we examined the possibility of encouraging genetic screening of these variants/loci in order to critically implement decision-making about the therapeutic approach through precision medicine strategies. It is possible then to anticipate that when the clinical practice will take advantage of the genetic information of the diabetic patients, this will provide a useful resource for the prevention of T2DM progression, enabling the identification of the precise drug that is most likely to be effective and safe for each patient and the reduction of the economic impact on a global scale.
Collapse
Affiliation(s)
- Gaia Chiara Mannino
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Francesco Andreozzi
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Giorgio Sesti
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| |
Collapse
|
24
|
Viji D, Aswathi P, Pricilla Charmine P, Akram Husain R, Noorul Ameen S, Ahmed SS, Ramakrishnan V. Genetic association of ABCC8 rs757110 polymorphism with Type 2 Diabetes Mellitus risk: A case-control study in South India and a meta-analysis. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
25
|
Onaolapo AY, Onaolapo OJ. Circadian dysrhythmia-linked diabetes mellitus: Examining melatonin’s roles in prophylaxis and management. World J Diabetes 2018; 9:99-114. [PMID: 30079146 PMCID: PMC6068738 DOI: 10.4239/wjd.v9.i7.99] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is a chronic, life-threatening metabolic disorder that occurs worldwide. Despite an increase in the knowledge of the risk factors that are associated with diabetes mellitus, its worldwide prevalence has continued to rise; thus, necessitating more research into its aetiology. Recent researches are beginning to link a dysregulation of the circadian rhythm to impairment of intermediary metabolism; with evidences that circadian rhythm dysfunction might play an important role in the aetiology, course or prognosis of some cases of diabetes mellitus. These evidences thereby suggest possible relationships between the circadian rhythm regulator melatonin, and diabetes mellitus. In this review, we discuss the roles of the circadian rhythm in the regulation of the metabolism of carbohydrates and other macronutrients; with emphasis on the importance of melatonin and the impacts of its deficiency on carbohydrate homeostasis. Also, the possibility of using melatonin and its analogs for the “prophylaxis” or management of diabetes mellitus is also considered.
Collapse
Affiliation(s)
- Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho 210211, Oyo State, Nigeria
| | - Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo 230263, Osun State, Nigeria
| |
Collapse
|
26
|
Wheeler E, Marenne G, Barroso I. Genetic aetiology of glycaemic traits: approaches and insights. Hum Mol Genet 2017; 26:R172-R184. [PMID: 28977447 PMCID: PMC5886471 DOI: 10.1093/hmg/ddx293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 07/18/2017] [Accepted: 07/21/2017] [Indexed: 12/17/2022] Open
Abstract
Glycaemic traits such as fasting and post-challenge glucose and insulin measures, as well as glycated haemoglobin (HbA1c), are used to diagnose and monitor diabetes. These traits are risk factors for cardiovascular disease even below the diabetic threshold, and their study can additionally yield insights into the pathophysiology of type 2 diabetes. To date, a diverse set of genetic approaches have led to the discovery of over 97 loci influencing glycaemic traits. In this review, we will focus on recent advances in the genetic aetiology of glycaemic traits, and the resulting biological insights. We will provide a brief overview of results ranging from common, to low- and rare-frequency variant-trait association studies, studies leveraging the diversity across populations, and studies harnessing the power of genetic and genomic approaches to gain insights into the biological underpinnings of these traits.
Collapse
Affiliation(s)
- Eleanor Wheeler
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Gaëlle Marenne
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Inês Barroso
- Department of Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
27
|
Thomsen SK, Gloyn AL. Human genetics as a model for target validation: finding new therapies for diabetes. Diabetologia 2017; 60:960-970. [PMID: 28447115 PMCID: PMC5423999 DOI: 10.1007/s00125-017-4270-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/14/2017] [Indexed: 01/01/2023]
Abstract
Type 2 diabetes is a global epidemic with major effects on healthcare expenditure and quality of life. Currently available treatments are inadequate for the prevention of comorbidities, yet progress towards new therapies remains slow. A major barrier is the insufficiency of traditional preclinical models for predicting drug efficacy and safety. Human genetics offers a complementary model to assess causal mechanisms for target validation. Genetic perturbations are 'experiments of nature' that provide a uniquely relevant window into the long-term effects of modulating specific targets. Here, we show that genetic discoveries over the past decades have accurately predicted (now known) therapeutic mechanisms for type 2 diabetes. These findings highlight the potential for use of human genetic variation for prospective target validation, and establish a framework for future applications. Studies into rare, monogenic forms of diabetes have also provided proof-of-principle for precision medicine, and the applicability of this paradigm to complex disease is discussed. Finally, we highlight some of the limitations that are relevant to the use of genome-wide association studies (GWAS) in the search for new therapies for diabetes. A key outstanding challenge is the translation of GWAS signals into disease biology and we outline possible solutions for tackling this experimental bottleneck.
Collapse
Affiliation(s)
- Soren K Thomsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford, OX3 7LE, UK.
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
- National Institute of Health Research Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
28
|
Yang Y, Chan L. Monogenic Diabetes: What It Teaches Us on the Common Forms of Type 1 and Type 2 Diabetes. Endocr Rev 2016; 37:190-222. [PMID: 27035557 PMCID: PMC4890265 DOI: 10.1210/er.2015-1116] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To date, more than 30 genes have been linked to monogenic diabetes. Candidate gene and genome-wide association studies have identified > 50 susceptibility loci for common type 1 diabetes (T1D) and approximately 100 susceptibility loci for type 2 diabetes (T2D). About 1-5% of all cases of diabetes result from single-gene mutations and are called monogenic diabetes. Here, we review the pathophysiological basis of the role of monogenic diabetes genes that have also been found to be associated with common T1D and/or T2D. Variants of approximately one-third of monogenic diabetes genes are associated with T2D, but not T1D. Two of the T2D-associated monogenic diabetes genes-potassium inward-rectifying channel, subfamily J, member 11 (KCNJ11), which controls glucose-stimulated insulin secretion in the β-cell; and peroxisome proliferator-activated receptor γ (PPARG), which impacts multiple tissue targets in relation to inflammation and insulin sensitivity-have been developed as major antidiabetic drug targets. Another monogenic diabetes gene, the preproinsulin gene (INS), is unique in that INS mutations can cause hyperinsulinemia, hyperproinsulinemia, neonatal diabetes mellitus, one type of maturity-onset diabetes of the young (MODY10), and autoantibody-negative T1D. Dominant heterozygous INS mutations are the second most common cause of permanent neonatal diabetes. Moreover, INS gene variants are strongly associated with common T1D (type 1a), but inconsistently with T2D. Variants of the monogenic diabetes gene Gli-similar 3 (GLIS3) are associated with both T1D and T2D. GLIS3 is a key transcription factor in insulin production and β-cell differentiation during embryonic development, which perturbation forms the basis of monogenic diabetes as well as its association with T1D. GLIS3 is also required for compensatory β-cell proliferation in adults; impairment of this function predisposes to T2D. Thus, monogenic forms of diabetes are invaluable "human models" that have contributed to our understanding of the pathophysiological basis of common T1D and T2D.
Collapse
Affiliation(s)
- Yisheng Yang
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Lawrence Chan
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
29
|
Abstract
KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.
Collapse
Affiliation(s)
- Monique N Foster
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
30
|
Nair AK, Baier LJ. Complex Genetics of Type 2 Diabetes and Effect Size: What have We Learned from Isolated Populations? Rev Diabet Stud 2016; 12:299-319. [PMID: 27111117 DOI: 10.1900/rds.2015.12.299] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genetic studies in large outbred populations have documented a complex, highly polygenic basis for type 2 diabetes (T2D). Most of the variants currently known to be associated with T2D risk have been identified in large studies that included tens of thousands of individuals who are representative of a single major ethnic group such as European, Asian, or African. However, most of these variants have only modest effects on the risk for T2D; identification of definitive 'causal variant' or 'causative loci' is typically lacking. Studies in isolated populations offer several advantages over outbred populations despite being, on average, much smaller in sample size. For example, reduced genetic variability, enrichment of rare variants, and a more uniform environment and lifestyle, which are hallmarks of isolated populations, can reduce the complexity of identifying disease-associated genes. To date, studies in isolated populations have provided valuable insight into the genetic basis of T2D by providing both a deeper understanding of previously identified T2D-associated variants (e.g. demonstrating that variants in KCNQ1 have a strong parent-of-origin effect) or providing novel variants (e.g. ABCC8 in Pima Indians, TBC1D4 in the Greenlandic population, HNF1A in Canadian Oji-Cree). This review summarizes advancements in genetic studies of T2D in outbred and isolated populations, and provides information on whether the difference in the prevalence of T2D in different populations (Pima Indians vs. non-Hispanic Whites and non-Hispanic Whites vs. non-Hispanic Blacks) can be explained by the difference in risk allele frequencies of established T2D variants.
Collapse
Affiliation(s)
- Anup K Nair
- Diabetes Molecular Genetics Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85004, USA
| | - Leslie J Baier
- Diabetes Molecular Genetics Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85004, USA
| |
Collapse
|
31
|
Modeling K,ATP--dependent excitability in pancreatic islets. Biophys J 2015; 107:2016-26. [PMID: 25418087 DOI: 10.1016/j.bpj.2014.09.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/22/2014] [Accepted: 09/30/2014] [Indexed: 11/23/2022] Open
Abstract
In pancreatic ?-cells, K,ATP channels respond to changes in glucose to regulate cell excitability and insulin release. Confirming a high sensitivity of electrical activity to K,ATP activity, mutations that cause gain of K,ATP function cause neonatal diabetes. Our aim was to quantitatively assess the contribution of K,ATP current to the regulation of glucose-dependent bursting by reproducing experimentally observed changes in excitability when K,ATP conductance is altered by genetic manipulation. A recent detailed computational model of single cell pancreatic ?-cell excitability reproduces the ?-cell response to varying glucose concentrations. However, initial simulations showed that the model underrepresents the significance of K,ATP activity and was unable to reproduce K,ATP conductance-dependent changes in excitability. By altering the ATP and glucose dependence of the L-type Ca(2+) channel and the Na-K ATPase to better fit experiment, appropriate dependence of excitability on K,ATP conductance was reproduced. Because experiments were conducted in islets, which contain cell-to-cell variability, we extended the model from a single cell to a three-dimensional model (10×10×10 cell) islet with 1000 cells. For each cell, the conductance of the major currents was allowed to vary as was the gap junction conductance between cells. This showed that single cell glucose-dependent behavior was then highly variable, but was uniform in coupled islets. The study highlights the importance of parameterization of detailed models of ?-cell excitability and suggests future experiments that will lead to improved characterization of ?-cell excitability and the control of insulin secretion.
Collapse
|
32
|
George PS, Tavendale R, Palmer CNA, McCrimmon RJ. Diazoxide improves hormonal counterregulatory responses to acute hypoglycemia in long-standing type 1 diabetes. Diabetes 2015; 64:2234-41. [PMID: 25591873 DOI: 10.2337/db14-1539] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/07/2015] [Indexed: 11/13/2022]
Abstract
Individuals with long-standing type 1 diabetes (T1D) are at increased risk of severe hypoglycemia secondary to impairments in normal glucose counterregulatory responses (CRRs). Strategies to prevent hypoglycemia are often ineffective, highlighting the need for novel therapies. ATP-sensitive potassium (KATP) channels within the hypothalamus are thought to be integral to hypoglycemia detection and initiation of CRRs; however, to date this has not been confirmed in human subjects. In this study, we examined whether the KATP channel-activator diazoxide was able to amplify the CRR to hypoglycemia in T1D subjects with long-duration diabetes. A randomized, double-blind, placebo-controlled cross-over trial using a stepped hyperinsulinemic hypoglycemia clamp was performed in 12 T1D subjects with prior ingestion of diazoxide (7 mg/kg) or placebo. Diazoxide resulted in a 37% increase in plasma levels of epinephrine and a 44% increase in plasma norepinephrine during hypoglycemia compared with placebo. In addition, a subgroup analysis revealed that the response to oral diazoxide was blunted in participants with E23K polymorphism in the KATP channel. This study has therefore shown for the first time the potential utility of KATP channel activators to improve CRRs to hypoglycemia in individuals with T1D and, moreover, that it may be possible to stratify therapeutic approaches by genotype.
Collapse
Affiliation(s)
- Priya S George
- Division of Diabetes and Cardiovascular Medicine, Medical Research Institute, Ninewells Hospital and Medical School, Dundee, Scotland, U.K.
| | - Roger Tavendale
- The Pat McPherson Centre for Pharmacogenomics & Pharmacogenetics, Medical Research Institute, Ninewells Hospital and Medical School, Dundee, Scotland, U.K
| | - Colin N A Palmer
- The Pat McPherson Centre for Pharmacogenomics & Pharmacogenetics, Medical Research Institute, Ninewells Hospital and Medical School, Dundee, Scotland, U.K
| | - Rory J McCrimmon
- Division of Diabetes and Cardiovascular Medicine, Medical Research Institute, Ninewells Hospital and Medical School, Dundee, Scotland, U.K
| |
Collapse
|
33
|
Sokolova EA, Bondar IA, Shabelnikova OY, Pyankova OV, Filipenko ML. Replication of KCNJ11 (p.E23K) and ABCC8 (p.S1369A) Association in Russian Diabetes Mellitus 2 Type Cohort and Meta-Analysis. PLoS One 2015; 10:e0124662. [PMID: 25955821 PMCID: PMC4425644 DOI: 10.1371/journal.pone.0124662] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/17/2015] [Indexed: 12/26/2022] Open
Abstract
The genes ABCC8 and KCNJ11 have received intense focus in type 2 diabetes mellitus (T2DM) research over the past two decades. It has been hypothesized that the p.E23K (KCNJ11) mutation in the 11p15.1 region may play an important role in the development of T2DM. In 2009, Hamming et al. found that the p.1369A (ABCC8) variant may be a causal factor in the disease; therefore, in this study we performed a meta-analysis to evaluate the association between these single nucleotide polymorphisms (SNPs), including our original data on the Siberian population (1384 T2DM and 414 controls). We found rs5219 and rs757110 were not associated with T2DM in this population, and that there was linkage disequilibrium in Siberians (D’=0.766, r2= 0.5633). In addition, the haplotype rs757110[T]-rs5219[C] (p.23K/p.S1369) was associated with T2DM (OR = 1.52, 95% CI: 1.04-2.24). We included 44 original studies published by June 2014 in a meta-analysis of the p.E23K association with T2DM. The total OR was 1.14 (95% CI: 1.11-1.17) for p.E23K for a total sample size of 137,298. For p.S1369A, a meta-analysis was conducted on a total of 10 studies with a total sample size of 14,136 and pooled OR of 1.14 [95% CI (1.08-1.19); p = 2 x 10-6]. Our calculations identified causal genetic variation within the ABCC8/KCNJ11 region for T2DM with an OR of approximately 1.15 in Caucasians and Asians. Moreover, the OR value was not dependent on the frequency of p.E23K or p.S1369A in the populations.
Collapse
Affiliation(s)
- Ekaterina Alekseevna Sokolova
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Division, Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Irina Arkadievna Bondar
- Novosibirsk State Regional Hospital, Regional Diabetes center, Novosibirsk, Russia
- Novosibirsk State Medical University, Novosibirsk, Russia
| | - Olesya Yurievna Shabelnikova
- Novosibirsk State Regional Hospital, Regional Diabetes center, Novosibirsk, Russia
- Novosibirsk State Medical University, Novosibirsk, Russia
| | - Olga Vladimirovna Pyankova
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Division, Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Maxim Leonidovich Filipenko
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Siberian Division, Russian Academy of Sciences, Novosibirsk, Russia
- Kazan Federal University, Kazan, Russia
- * E-mail:
| |
Collapse
|
34
|
Prasad RB, Groop L. Genetics of type 2 diabetes-pitfalls and possibilities. Genes (Basel) 2015; 6:87-123. [PMID: 25774817 PMCID: PMC4377835 DOI: 10.3390/genes6010087] [Citation(s) in RCA: 290] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/28/2015] [Accepted: 02/27/2015] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex disease that is caused by a complex interplay between genetic, epigenetic and environmental factors. While the major environmental factors, diet and activity level, are well known, identification of the genetic factors has been a challenge. However, recent years have seen an explosion of genetic variants in risk and protection of T2D due to the technical development that has allowed genome-wide association studies and next-generation sequencing. Today, more than 120 variants have been convincingly replicated for association with T2D and many more with diabetes-related traits. Still, these variants only explain a small proportion of the total heritability of T2D. In this review, we address the possibilities to elucidate the genetic landscape of T2D as well as discuss pitfalls with current strategies to identify the elusive unknown heritability including the possibility that our definition of diabetes and its subgroups is imprecise and thereby makes the identification of genetic causes difficult.
Collapse
Affiliation(s)
- Rashmi B Prasad
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Lund University, CRC, Skåne University Hospital SUS, SE-205 02 Malmö, Sweden.
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Lund University, CRC, Skåne University Hospital SUS, SE-205 02 Malmö, Sweden.
- Finnish Institute of Molecular Medicine (FIMM), Helsinki University, Helsinki 00014, Finland.
| |
Collapse
|
35
|
Abstract
Type 2 diabetes (T2D) had long been referred to as the "geneticist's nightmare." Genome-wide association studies have fully confirmed the polygenic nature of T2D, demonstrating the role of many genes in T2D risk. The increasingly busier picture of T2D genetics is quite difficult to understand for the diabetes research community, which can create misunderstandings with geneticists, and can eventually limit both basic research and translational outcomes of these genetic discoveries. The present review wishes to lift the fog around genetics of T2D with the hope that it will foster integrated diabetes modeling approaches from genetic defects to personalized medicine.
Collapse
Affiliation(s)
- Amélie Bonnefond
- CNRS-UMR8199, Lille Pasteur Institute, Lille 59000, France; Lille University, Lille 59000, France; European Genomic Institute for Diabetes (EGID), Lille 59000, France
| | - Philippe Froguel
- CNRS-UMR8199, Lille Pasteur Institute, Lille 59000, France; Lille University, Lille 59000, France; European Genomic Institute for Diabetes (EGID), Lille 59000, France; Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, London W12 0NN, UK.
| |
Collapse
|
36
|
The E23K and A190A variations of the KCNJ11 gene are associated with early-onset type 2 diabetes and blood pressure in the Chinese population. Mol Cell Biochem 2015; 404:133-41. [PMID: 25725792 DOI: 10.1007/s11010-015-2373-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/23/2015] [Indexed: 01/19/2023]
Abstract
Conflicting associations between define (KCNJ11) variations and susceptibility to late-onset (>40 years old) type 2 diabetes mellitus (T2DM) have been reported in different ethnic groups. We investigated whether the E23K (G→A, rs5219) or A190A (C→T, rs5218) variations in KCNJ11 are associated with early-onset T2DM and blood pressure in the Chinese population. Case-control study of 175 unrelated Chinese patients with early-onset T2DM (age of onset <40 years old) who receive (ins+, n = 57) or do not receive insulin (ins-, n = 118), and 182 non-diabetic control subjects. PCR-direct sequencing was performed to genotype E23K and A190A; the genotypic frequencies and associations with clinical characteristics were analyzed. The genotypic frequencies of E23K-GA+AA were higher and A190A-TT was lower in the early-onset T2DM group, especially the T2D-ins+ group, compared to the non-diabetic control group (p < 0.01 or 0.05, respectively). In non-diabetic subjects, E23K-AA carriers had significantly higher 2 h plasma glucose and lower 2 h insulin than E23K-GG carriers (both p < 0.05). A190A-TT or E23K-GG carriers had higher systolic blood pressure (SBP) than CC or AA carriers in the non-diabetic control and T2DM groups (both p < 0.05). In the T2DM ins+ group, E23K-AA carriers had lower onset age and duration of diabetes and higher BMI than GG carriers, and A190A-TT carriers had higher SBP than CC carriers (all p < 0.05). The E23K-GA or AA genotypes may increase the susceptibility to early-onset T2DM, while A190A-TT may protect against early-onset T2DM. On the other hand the A190A-TT or E23K-GG genotypes may increase the risk of hypertension in the Chinese population.
Collapse
|
37
|
Nikitin AG, Potapov VA, Brovkin AN, Lavrikova EY, Khodyrev DS, Shamhalova MS, Smetanina SA, Suplotova LN, Shestakova MV, Nosikov VV, Averyanov AV. Association of FTO, KCNJ11, SLC30A8, and CDKN2B polymorphisms with type 2 diabetes mellitus. Mol Biol 2015. [DOI: 10.1134/s0026893315010112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
38
|
Bonfanti DH, Alcazar LP, Arakaki PA, Martins LT, Agustini BC, de Moraes Rego FG, Frigeri HR. ATP-dependent potassium channels and type 2 diabetes mellitus. Clin Biochem 2015; 48:476-82. [PMID: 25583094 DOI: 10.1016/j.clinbiochem.2014.12.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 12/24/2022]
Abstract
Diabetes mellitus is a public health problem, which affects a millions worldwide. Most diabetes cases are classified as type 2 diabetes mellitus, which is highly associated with obesity. Type 2 diabetes is considered a multifactorial disorder, with both environmental and genetic factors contributing to its development. An important issue linked with diabetes development is the failure of the insulin releasing mechanism involving abnormal activity of the ATP-dependent potassium channel, KATP. This channel is a transmembrane protein encoded by the KCNJ11 and ABCC8 genes. Furthermore, polymorphisms in these genes have been linked to type 2 diabetes because of the role of KATP in insulin release. While several genetic variations have been reported to be associated with this disease, the E23K polymorphism is most commonly associated with this pathology, as well as to obesity. Here, we review the molecular genetics of the potassium channel and discusses its most described polymorphisms and their associations with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Dianne Heloisa Bonfanti
- Health and Biosciences School, Pontifical Catholic University of Parana, Curitiba, Parana, Brazil
| | - Larissa Pontes Alcazar
- Health and Biosciences School, Pontifical Catholic University of Parana, Curitiba, Parana, Brazil
| | - Priscila Akemi Arakaki
- Health and Biosciences School, Pontifical Catholic University of Parana, Curitiba, Parana, Brazil
| | - Laysa Toschi Martins
- Health and Biosciences School, Pontifical Catholic University of Parana, Curitiba, Parana, Brazil
| | - Bruna Carla Agustini
- Health and Biosciences School, Pontifical Catholic University of Parana, Curitiba, Parana, Brazil
| | | | | |
Collapse
|
39
|
Rastegari A, Rabbani M, Sadeghi HM, Imani EF, Hasanzadeh A, Moazen F. Association of KCNJ11 (E23K) gene polymorphism with susceptibility to type 2 diabetes in Iranian patients. Adv Biomed Res 2015; 4:1. [PMID: 25625107 PMCID: PMC4300590 DOI: 10.4103/2277-9175.148256] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 01/21/2014] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a multifactorial disease with susceptibility of several genes that are related to T2D. Insulin secretion pathway starts with potassium channels in pancreatic beta cells. KCNJ11 gene encodes ATP-sensitive potassium channel subunits. Some studies suggested that KCNJ11 (E23K) mutation increases the risk of T2D. Therefore, present study was designed to investigate the association between E23K polymorphism of KCNJ11 gene and type 2 diabetes mellitus (T2DM) in the Iranian population. MATERIALS AND METHODS The type of study was case-control and 40 unrelated subjects, including 20 healthy controls and 20 diabetic patients were recruited (diagnosed based on American Diabetes Association criteria). Blood samples were used for isolation of genomic deoxyribonucleic acid (DNA). Having extracted the genomic DNA from human blood leukocytes by means of High Pure PCR Template Preparation Kit, PCR-restriction fragment length polymorphism method was used to detect KCNJ11 E23K gene polymorphism. BanII restriction enzyme was used for digestion. Data were analyzed using Chi-square or Fisher exact test or independent t-test, as appropriate. P < 0.05 was considered. RESULTS We found that the carrier homozygous for KK genotype are susceptible to T2D (0.049) and in patients the frequency of K allele was higher than control subjects (0.048). CONCLUSION The present study suggests that KCNJ11 (E23K) gene polymorphism is associated with T2DM.
Collapse
Affiliation(s)
- Ali Rastegari
- Department of Pharmaceutical Biotechnology, University of Medical Sciences, Isfahan, Iran
| | - Mohammad Rabbani
- Department of Pharmaceutical Biotechnology, University of Medical Sciences, Isfahan, Iran
| | | | - Elham Faghih Imani
- Department of Internal Medicine, University of Medical Sciences, Isfahan, Iran
| | - Akbar Hasanzadeh
- Department of Biostatistics and Epidemiology, University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Moazen
- Department of Pharmaceutical Biotechnology, University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
40
|
Esplin ED, Oei L, Snyder MP. Personalized sequencing and the future of medicine: discovery, diagnosis and defeat of disease. Pharmacogenomics 2014; 15:1771-1790. [PMID: 25493570 DOI: 10.2217/pgs.14.117] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The potential for personalized sequencing to individually optimize medical treatment in diseases such as cancer and for pharmacogenomic application is just beginning to be realized, and the utility of sequencing healthy individuals for managing health is also being explored. The data produced requires additional advancements in interpretation of variants of unknown significance to maximize clinical benefit. Nevertheless, personalized sequencing, only recently applied to clinical medicine, has already been broadly applied to the discovery and study of disease. It is poised to enable the earlier and more accurate diagnosis of disease risk and occurrence, guide prevention and individualized intervention as well as facilitate monitoring of healthy and treated patients, and play a role in the prevention and recurrence of future disease. This article documents the advancing capacity of personalized sequencing, reviews its impact on disease-oriented scientific discovery and anticipates its role in the future of medicine.
Collapse
Affiliation(s)
- Edward D Esplin
- 300 Pasteur Drive, Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
41
|
Population specific impact of genetic variants in KCNJ11 gene to type 2 diabetes: a case-control and meta-analysis study. PLoS One 2014; 9:e107021. [PMID: 25247988 PMCID: PMC4172481 DOI: 10.1371/journal.pone.0107021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 08/04/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Potassium inwardly rectifying channel, subfamily J, member 11 (KCNJ11) gene have a key role in insulin secretion and is of substantial interest as a candidate gene for type 2 diabetes (T2D). The current work was performed to delineate the genetic influence of KCNJ11 polymorphisms on risk of T2D in South Indian population through case-control association study along with systematic review and meta-analysis. METHODS A case-control study of 400 T2D cases and controls of South Indian origin were performed to analyze the association of KCNJ11 polymorphisms (rs5219, rs5215, rs41282930, rs1800467) and copy number variations (CNV) on the risk of T2D. In addition a systematic review and meta-analysis for KCNJ11 rs5219 was conducted in 3,831 cases and 3,543 controls from 5 published reports from South-Asian population by searching various databases. Odds ratio with 95% confidence interval (CI) was used to assess the association strength. Cochran's Q, I2 statistics were used to study heterogeneity between the eligible studies. RESULTS KCNJ11 rs5215, C-G-C-C haplotype and two loci analysis (rs5219 vs rs1800467) showed a significant association with T2D but CNV analysis did not show significant variation between T2D cases and control subjects. Lower age of disease onset (P = 0.04) and higher body mass index (BMI) (P = 0.04) were associated with rs5219 TT genotype in T2D patients. The meta-analysis of KCNJ11 rs5219 on South Asian population showed no association on susceptibility to T2D with an overall pooled OR = 0.98, 95% CI = 0.83-1.16. Stratification analysis showed East Asian population and global population were associated with T2D when compared to South Asians. CONCLUSION KCNJ11 rs5219 is not independently associated with T2D in South-Indian population and our meta-analysis suggests that KCNJ11 polymorphism (rs5219) is associated with risk of T2D in East Asian population and global population but this outcome could not be replicated in South Asian sub groups.
Collapse
|
42
|
Sarikonda G, Pettus J, Phatak S, Sachithanantham S, Miller JF, Wesley JD, Cadag E, Chae J, Ganesan L, Mallios R, Edelman S, Peters B, von Herrath M. CD8 T-cell reactivity to islet antigens is unique to type 1 while CD4 T-cell reactivity exists in both type 1 and type 2 diabetes. J Autoimmun 2014; 50:77-82. [PMID: 24387802 DOI: 10.1016/j.jaut.2013.12.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/06/2013] [Accepted: 12/08/2013] [Indexed: 01/19/2023]
Abstract
Previous cross-sectional analyses demonstrated that CD8(+) and CD4(+) T-cell reactivity to islet-specific antigens was more prevalent in T1D subjects than in healthy donors (HD). Here, we examined T1D-associated epitope-specific CD4(+) T-cell cytokine production and autoreactive CD8(+) T-cell frequency on a monthly basis for one year in 10 HD, 33 subjects with T1D, and 15 subjects with T2D. Autoreactive CD4(+) T-cells from both T1D and T2D subjects produced more IFN-γ when stimulated than cells from HD. In contrast, higher frequencies of islet antigen-specific CD8(+) T-cells were detected only in T1D. These observations support the hypothesis that general beta-cell stress drives autoreactive CD4(+) T-cell activity while islet over-expression of MHC class I commonly seen in T1D mediates amplification of CD8(+) T-cells and more rapid beta-cell loss. In conclusion, CD4(+) T-cell autoreactivity appears to be present in both T1D and T2D while autoreactive CD8(+) T-cells are unique to T1D. Thus, autoreactive CD8(+) cells may serve as a more T1D-specific biomarker.
Collapse
Affiliation(s)
| | - Jeremy Pettus
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; University of California San Diego, San Diego, CA, USA
| | - Sonal Phatak
- University of California San Diego, San Diego, CA, USA
| | | | | | | | | | - Ji Chae
- University of California San Diego, San Diego, CA, USA
| | | | - Ronna Mallios
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Steve Edelman
- University of California San Diego, San Diego, CA, USA
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Matthias von Herrath
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA; Novo Nordisk Type 1 Diabetes R & D Center, Seattle, WA, USA.
| |
Collapse
|
43
|
Qiu L, Na R, Xu R, Wang S, Sheng H, Wu W, Qu Y. Quantitative assessment of the effect of KCNJ11 gene polymorphism on the risk of type 2 diabetes. PLoS One 2014; 9:e93961. [PMID: 24710510 PMCID: PMC3977990 DOI: 10.1371/journal.pone.0093961] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 02/19/2014] [Indexed: 12/31/2022] Open
Abstract
To clarify the role of potassium inwardly-rectifying-channel, subfamily-J, member 11 (KCNJ11) variation in susceptibility to type 2 diabetes (T2D), we performed a systematic meta-analysis to investigate the association between the KCNJ11 E23K polymorphism (rs5219) and the T2D in different genetic models. Databases including PubMed, Medline, EMBASE, and ISI Web of Science were searched to identify relevant studies. A total of 48 published studies involving 56,349 T2D cases, 81,800 controls, and 483 family trios were included in this meta-analysis. Overall, the E23K polymorphism was significantly associated with increased T2D risk with per-allele odds ratio (OR) of 1.12 (95% CI: 1.09-1.16; P<10-5). The summary OR for T2D was 1.09 (95% CI: 1.03-1.14; P<10-5), and 1.26 (95% CI: 1.17-1.35; P<10-5), for heterozygous and homozygous, respectively. Similar results were also detected under dominant and recessive genetic models. When stratified by ethnicity, significantly increased risks were found for the polymorphism in Caucasians and East Asians. However, no such associations were detected among Indian and other ethnic populations. Significant associations were also observed in the stratified analyses according to different mean BMI of cases and sample size. Although significant between study heterogeneity was identified, meta-regression analysis suggested that the BMI of controls significantly correlated with the magnitude of the genetic effect. The current meta-analysis demonstrated that a modest but statistically significant effect of the 23K allele of rs5219 polymorphism in susceptibility to T2D. But the contribution of its genetic variants to the epidemic of T2D in Indian and other ethnic populations appears to be relatively low.
Collapse
Affiliation(s)
- Ling Qiu
- Department of Geriatrics, Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Risu Na
- Department of Endocrinology, Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Rong Xu
- Department of Geriatrics, Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Siyang Wang
- Department of Geriatrics, Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Hongguang Sheng
- Department of Endocrinology, Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Wanling Wu
- Department of Endocrinology, The Ninth People's Hospital Attach to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Qu
- Department of Geriatrics, Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
44
|
Groop L, Pociot F. Genetics of diabetes--are we missing the genes or the disease? Mol Cell Endocrinol 2014; 382:726-739. [PMID: 23587769 DOI: 10.1016/j.mce.2013.04.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 01/25/2013] [Accepted: 04/02/2013] [Indexed: 12/20/2022]
Abstract
Diabetes is a group of metabolic diseases characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both. The chronic hyperglycemia of diabetes is associated with long-term damage, dysfunction, and failure of different organs, especially the eyes, kidneys, nerves, heart, and blood vessels. Several pathogenic processes are involved in the development of diabetes. These range from autoimmune destruction of the beta-cells of the pancreas with consequent insulin deficiency to abnormalities that result in resistance to insulin action (American Diabetes Association, 2011). The vast majority of cases of diabetes fall into two broad categories. In type 1 diabetes (T1D), the cause is an absolute deficiency of insulin secretion, whereas in type 2 diabetes (T2D), the cause is a combination of resistance to insulin action and an inadequate compensatory insulin secretory response. However, the subdivision into two main categories represents a simplification of the real situation, and research during the recent years has shown that the disease is much more heterogeneous than a simple subdivision into two major subtypes assumes. Worldwide prevalence figures estimate that there are 280 million diabetic patients in 2011 and more than 500 million in 2030 (http://www.diabetesatlas.org/). In Europe, about 6-8% of the population suffer from diabetes, of them about 90% has T2D and 10% T1D, thereby making T2D to the fastest increasing disease in Europe and worldwide. This epidemic has been ascribed to a collision between the genes and the environment. While our knowledge about the genes is clearly better for T1D than for T2D given the strong contribution of variation in the HLA region to the risk of T1D, the opposite is the case for T2D, where our knowledge about the environmental triggers (obesity, lack of exercise) is much better than the understanding of the underlying genetic causes. This lack of knowledge about the underlying genetic causes of diabetes is often referred to as missing heritability (Manolio et al., 2009) which exceeds 80% for T2D but less than 25% for T1D. In the following review, we will discuss potential sources of this missing heritability which also includes the possibility that our definition of diabetes and its subgroups is imprecise and thereby making the identification of genetic causes difficult.
Collapse
Affiliation(s)
- Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, University Hospital Skåne, Malmö, Sweden; Glostrup Research Institute, Glostrup University Hospital, Glostrup, Denmark.
| | - Flemming Pociot
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University, University Hospital Skåne, Malmö, Sweden; Glostrup Research Institute, Glostrup University Hospital, Glostrup, Denmark
| |
Collapse
|
45
|
Bondar' IA, Shabel'nikova OY. Genetic framework of type 2 diabetes mellitus. DIABETES MELLITUS 2013. [DOI: 10.14341/dm2013411-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
More than 100 genes associated with the risk of type 2 diabetes mellitus (T2DM) are now established. Most of them affect insulin secretion, adipogenesis and insulin resistance, but the exact molecular mechanisms determining their involvement in the pathogenesis of T2DM are not understood completely.
Collapse
|
46
|
Role of genetic polymorphisms of ion channels in the pathophysiology of coronary microvascular dysfunction and ischemic heart disease. Basic Res Cardiol 2013; 108:387. [PMID: 24068186 PMCID: PMC3898136 DOI: 10.1007/s00395-013-0387-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 08/13/2013] [Accepted: 09/11/2013] [Indexed: 01/14/2023]
Abstract
Conventionally, ischemic heart disease (IHD) is equated with large vessel coronary disease. However, recent evidence has suggested a role of compromised microvascular regulation in the etiology of IHD. Because regulation of coronary blood flow likely involves activity of specific ion channels, and key factors involved in endothelium-dependent dilation, we proposed that genetic anomalies of ion channels or specific endothelial regulators may underlie coronary microvascular disease. We aimed to evaluate the clinical impact of single-nucleotide polymorphisms in genes encoding for ion channels expressed in the coronary vasculature and the possible correlation with IHD resulting from microvascular dysfunction. 242 consecutive patients who were candidates for coronary angiography were enrolled. A prospective, observational, single-center study was conducted, analyzing genetic polymorphisms relative to (1) NOS3 encoding for endothelial nitric oxide synthase (eNOS); (2) ATP2A2 encoding for the Ca2+/H+-ATPase pump (SERCA); (3) SCN5A encoding for the voltage-dependent Na+ channel (Nav1.5); (4) KCNJ8 and KCNJ11 encoding for the Kir6.1 and Kir6.2 subunits of K-ATP channels, respectively; and (5) KCN5A encoding for the voltage-gated K+ channel (Kv1.5). No significant associations between clinical IHD manifestations and polymorphisms for SERCA, Kir6.1, and Kv1.5 were observed (p > 0.05), whereas specific polymorphisms detected in eNOS, as well as in Kir6.2 and Nav1.5 were found to be correlated with IHD and microvascular dysfunction. Interestingly, genetic polymorphisms for ion channels seem to have an important clinical impact influencing the susceptibility for microvascular dysfunction and IHD, independent of the presence of classic cardiovascular risk factors.
Collapse
|
47
|
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
|
48
|
Anuradha CV. Phytochemicals targeting genes relevant for type 2 diabetes. Can J Physiol Pharmacol 2013; 91:397-411. [PMID: 23745945 DOI: 10.1139/cjpp-2012-0350] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Nutrigenomic approaches based on ethnopharmacology and phytotherapy concepts have revealed that type 2 diabetes mellitus (T2DM) may be susceptible to dietary intervention. Interaction between bioactive food components and the genome may influence cell processes and modulate the onset and progression of the disease. T2DM, characterized by insulin resistance and beta cell dysfunction, is one of the leading causes of death and disability. Despite the great advances that have been made in the understanding and management of this complex, multifactorial disease, T2DM has become a worldwide epidemic in the 21st century. Population and family studies have revealed a strong genetic component of T2DM, and a number of candidate genes have been identified in humans. Variations in the gene sequences such as single nucleotide polymorphisms, explain the individual differences in traits like disease susceptibility and response to treatment. A clear understanding of how nutrients affect the expression of genes should facilitate the development of individualized intervention and, eventually, treatment strategies for T2DM. Review of the literature identified many phytochemicals/extracts from traditional medicinal plants that can target diabetogenic genes. This review focuses on the genetic aspects of T2DM, nutrient modification of genes relevant for diabetes, and future prospects of nutritional therapy of T2DM.
Collapse
Affiliation(s)
- Carani Venkatraman Anuradha
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar - 608 002, Tamil Nadu, India.
| |
Collapse
|
49
|
Abstract
ATP-sensitive potassium (KATP) channels were first discovered in the heart 30 years ago. Reconstitution of KATP channel activity by coexpression of members of the pore-forming inward rectifier gene family (Kir6.1, KCNJ8, and Kir6.2 KCNJ11) with sulfonylurea receptors (SUR1, ABCC8, and SUR2, ABCC9) of the ABCC protein subfamily has led to the elucidation of many details of channel gating and pore properties. In addition, the essential roles of Kir6.x and SURx subunits in generating cardiac and vascular KATP(2) and the detrimental consequences of genetic deletions or mutations in mice have been recognized. However, despite this extensive body of knowledge, there has been a paucity of defined roles of KATP subunits in human cardiovascular diseases, although there are reports of association of a single Kir6.1 variant with the J-wave syndrome in the ECG, and 2 isolated studies have reported association of loss of function mutations in SUR2 with atrial fibrillation and heart failure. Two new studies convincingly demonstrate that mutations in the SUR2 gene are associated with Cantu syndrome, a complex multi-organ disorder characterized by hypertrichosis, craniofacial dysmorphology, osteochondrodysplasia, patent ductus arteriosus, cardiomegaly, pericardial effusion, and lymphoedema. This realization of previously unconsidered consequences provides significant insight into the roles of the KATP channel in the cardiovascular system and suggests novel therapeutic possibilities.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
50
|
Shimajiri Y, Yamana A, Morita S, Furuta H, Furuta M, Sanke T. Kir6.2 E23K polymorphism is related to secondary failure of sulfonylureas in non-obese patients with type 2 diabetes. J Diabetes Investig 2013; 4:445-9. [PMID: 24843693 PMCID: PMC4025112 DOI: 10.1111/jdi.12070] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 01/17/2013] [Accepted: 01/28/2013] [Indexed: 11/27/2022] Open
Abstract
Aims/Introduction The Kir6.2 E23K polymorphism was studied with a special reference to secondary sulfonylurea (SU) failure in non‐obese patients with type 2 diabetes. Materials and Methods We recruited 278 non‐obese (body mass index ≤30.0 kg/m2) Japanese patients with type 2 diabetes who had a history of SU treatment (for 11.2 ± 6.3 years) and compared the frequency of the secondary SU failure among the genotypes of the polymorphism. Genotyping of the Kir6.2 E23K was carried out by polymerase chain reaction‐restriction fragment length polymorphism. Results The genotype frequencies of the polymorphism were similar to those previously reported in Japanese patients with type 2 diabetes. The frequency with which patients deteriorated into secondary SU failure was significantly higher in those with the KK genotype than those with EE or EK genotypes. Among 214 patients who eventually received insulin therapy because of secondary SU failure, the period of SU treatment in those with the KK genotype was significantly shorter than those with the EE or EK genotype, although the period from diagnosis to the start of SU treatment was not significantly different. Conclusions These data suggest that the Kir6.2 E23K polymorphism is related to the acceleration of secondary SU failure in non‐obese Japanese patients with type 2 diabetes.
Collapse
Affiliation(s)
| | - Akiko Yamana
- Department of Clinical Laboratory Medicine Tokyo Japan
| | - Shuhei Morita
- Department of Clinical Laboratory Medicine Tokyo Japan
| | - Hiroto Furuta
- First Department of Medicine Wakayama Medical University Wakayama Japan
| | - Machi Furuta
- Department of Clinical Laboratory Medicine Tokyo Japan
| | - Tokio Sanke
- Department of Clinical Laboratory Medicine Tokyo Japan ; Institute for Diabetes Fuchu Hospital Tokyo Japan
| |
Collapse
|