151
|
Hizel C, Tremblay J, Bartlett G, Hamet P. Introduction. PROGRESS AND CHALLENGES IN PRECISION MEDICINE 2017:1-34. [DOI: 10.1016/b978-0-12-809411-2.00001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
152
|
Spears LD, Renth AL, McKuin MR, Kennedy AR, Andrisse S, Briggs NE, Fisher JS. A role for ataxia telangiectasia mutated in insulin-independent stimulation of glucose transport. TRENDS IN CELL & MOLECULAR BIOLOGY 2017; 12:49-56. [PMID: 30542240 PMCID: PMC6287632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Literature reports suggest that ataxia telangiectasia mutated (ATM) can activate the AMP-activated protein kinase (AMPK), a protein that can stimulate glucose transport in skeletal muscle. We hypothesized that 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an AMPK activator, would increase glucose transport in mouse extensor digitorum longus (EDL) muscles in an ATM-dependent manner. AICAR-stimulated glucose transport was prevented by the ATM inhibitor KU-55933 despite normal stimulation of AMPK phosphorylation. Consistent with this, AICAR caused AMPK phosphorylation but not an increase of glucose transport in ATM-deficient (ATM-/-) muscles. S231 of TBC1D1 matches the sequence motif of ATM substrates, and phosphorylation of this site is known to inhibit TBC1D1 and lead to increased glucose transport. Accordingly, we assessed TBC1D1 phosphorylation and found that AICAR-stimulated phosphorylation of TBC1D1 at S231 did not occur in ATM-/- muscles. However, activation of ATM without activation of AMPK was insufficient to increase TBC1D1 phosphorylation. The data suggest that ATM plays a role in AICAR-stimulated glucose transport downstream of AMPK.
Collapse
Affiliation(s)
| | - Allyson L. Renth
- Department of Biology, Saint Louis University, St. Louis, Missouri, USA
| | - Michael R. McKuin
- Department of Biology, Saint Louis University, St. Louis, Missouri, USA
| | - Anne R. Kennedy
- Department of Biology, Saint Louis University, St. Louis, Missouri, USA
| | | | - Nell E. Briggs
- Department of Biology, Saint Louis University, St. Louis, Missouri, USA
| | | |
Collapse
|
153
|
Zaharenko L, Kalnina I, Geldnere K, Konrade I, Grinberga S, Židzik J, Javorský M, Lejnieks A, Nikitina-Zake L, Fridmanis D, Peculis R, Radovica-Spalvina I, Hartmane D, Pugovics O, Tkáč I, Klimčáková L, Pīrāgs V, Klovins J. Single nucleotide polymorphisms in the intergenic region between metformin transporter OCT2 and OCT3 coding genes are associated with short-term response to metformin monotherapy in type 2 diabetes mellitus patients. Eur J Endocrinol 2016; 175:531-540. [PMID: 27609360 DOI: 10.1530/eje-16-0347] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVES High variability in clinical response to metformin is often observed in type 2 diabetes (T2D) patients, and it highlights the need for identification of genetic components affecting the efficiency of metformin therapy. Aim of this observational study is to evaluate the role of tagSNPs (tagging single nucleotide polymorphisms) from genomic regions coding for six metformin transporter genes with respect to the short-term efficiency. DESIGN 102 tagSNPs in 6 genes coding for metformin transporters were genotyped in the group of 102 T2D patients treated with metformin for 3 months. METHODS Most significant hits were analyzed in the group of 131 T2D patients from Slovakia. Pharmacokinetic study in 25 healthy nondiabetic volunteers was conducted to investigate the effects of identified polymorphisms. RESULTS In the discovery group of 102 patients, minor alleles of rs3119309, rs7757336 and rs2481030 were significantly nominally associated with metformin inefficiency (P = 1.9 × 10-6 to 8.1 × 10-6). Effects of rs2481030 and rs7757336 did not replicate in the group of 131 T2DM patients from Slovakia alone, whereas rs7757336 was significantly associated with a reduced metformin response in combined group. In pharmacokinetic study, group of individuals harboring risk alleles of rs7757336 and rs2481030 displayed significantly reduced AUC∞ of metformin in plasma. CONCLUSIONS For the first time, we have identified an association between the lack of metformin response and SNPs rs3119309 and rs7757336 located in the 5' flanking region of the genes coding for Organic cation transporter 2 and rs2481030 located in the 5' flanking region of Organic cation transporter 3 that was supported by the results of a pharmacokinetic study on 25 healthy volunteers.
Collapse
Affiliation(s)
| | - Ineta Kalnina
- Latvian Biomedical Research and Study CentreRiga, Latvia
| | - Kristine Geldnere
- Pauls Stradins Clinical University HospitalRiga, Latvia
- Faculty of MedicineUniversity of Latvia, Riga, Latvia
| | - Ilze Konrade
- Riga East Clinical University HospitalRiga, Latvia
- Riga Stradins UniversityRiga, Latvia
| | | | - Jozef Židzik
- Faculty of MedicineP. J. Šafárik University, Košice, Slovakia
| | - Martin Javorský
- Faculty of MedicineP. J. Šafárik University, Košice, Slovakia
| | - Aivars Lejnieks
- Riga East Clinical University HospitalRiga, Latvia
- Riga Stradins UniversityRiga, Latvia
| | | | | | - Raitis Peculis
- Latvian Biomedical Research and Study CentreRiga, Latvia
| | | | | | | | - Ivan Tkáč
- Faculty of MedicineP. J. Šafárik University, Košice, Slovakia
| | | | - Valdis Pīrāgs
- Pauls Stradins Clinical University HospitalRiga, Latvia
- Faculty of MedicineUniversity of Latvia, Riga, Latvia
| | - Janis Klovins
- Latvian Biomedical Research and Study CentreRiga, Latvia
| |
Collapse
|
154
|
Luizon MR, Eckalbar WL, Wang Y, Jones SL, Smith RP, Laurance M, Lin L, Gallins PJ, Etheridge AS, Wright F, Zhou Y, Molony C, Innocenti F, Yee SW, Giacomini KM, Ahituv N. Genomic Characterization of Metformin Hepatic Response. PLoS Genet 2016; 12:e1006449. [PMID: 27902686 PMCID: PMC5130177 DOI: 10.1371/journal.pgen.1006449] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 10/25/2016] [Indexed: 12/26/2022] Open
Abstract
Metformin is used as a first-line therapy for type 2 diabetes (T2D) and prescribed for numerous other diseases. However, its mechanism of action in the liver has yet to be characterized in a systematic manner. To comprehensively identify genes and regulatory elements associated with metformin treatment, we carried out RNA-seq and ChIP-seq (H3K27ac, H3K27me3) on primary human hepatocytes from the same donor treated with vehicle control, metformin or metformin and compound C, an AMP-activated protein kinase (AMPK) inhibitor (allowing to identify AMPK-independent pathways). We identified thousands of metformin responsive AMPK-dependent and AMPK-independent differentially expressed genes and regulatory elements. We functionally validated several elements for metformin-induced promoter and enhancer activity. These include an enhancer in an ataxia telangiectasia mutated (ATM) intron that has SNPs in linkage disequilibrium with a metformin treatment response GWAS lead SNP (rs11212617) that showed increased enhancer activity for the associated haplotype. Expression quantitative trait locus (eQTL) liver analysis and CRISPR activation suggest that this enhancer could be regulating ATM, which has a known role in AMPK activation, and potentially also EXPH5 and DDX10, its neighboring genes. Using ChIP-seq and siRNA knockdown, we further show that activating transcription factor 3 (ATF3), our top metformin upregulated AMPK-dependent gene, could have an important role in gluconeogenesis repression. Our findings provide a genome-wide representation of metformin hepatic response, highlight important sequences that could be associated with interindividual variability in glycemic response to metformin and identify novel T2D treatment candidates. Metformin is among the most widely prescribed drugs. It is used as a first line therapy for type 2 diabetes (T2D), and for additional diseases including cancer. The variability in response to metformin is substantial and can be caused by genetic factors. However, the molecular mechanisms of metformin action are not fully known. Here, we used various genomic assays to analyze human liver cells treated with or without metformin and identified in a genome-wide manner thousands of differentially expressed genes and gene regulatory elements affected by metformin. Follow up functional assays identified several novel genes and regulatory elements to be associated with metformin response. These include ATF3, a gene that showed gluconeogenesis repression upon metformin response and a potential regulatory element of the ATM gene that is associated with metformin treatment differences through genome-wide association studies. Combined, this work identifies several novel genes and gene regulatory elements that can be activated due to metformin treatment and thus provides candidate sequences in the human genome where nucleotide variation can lead to differences in metformin response. It also enables the identification and prioritization of novel candidates for T2D treatment.
Collapse
Affiliation(s)
- Marcelo R. Luizon
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Department of General Biology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Walter L. Eckalbar
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Yao Wang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Stacy L. Jones
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Robin P. Smith
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Megan Laurance
- Library and Center for Knowledge Management, University of California San Francisco, San Francisco, California, United States of America
| | - Lawrence Lin
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Paul J. Gallins
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Amy S. Etheridge
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Fred Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Yihui Zhou
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Cliona Molony
- Merck Research Labs, Merck & Co. Inc., Kenilworth, New Jersey, United States of America
| | - Federico Innocenti
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Kathleen M. Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
155
|
Sacco F, Calderone A, Castagnoli L, Cesareni G. The cell-autonomous mechanisms underlying the activity of metformin as an anticancer drug. Br J Cancer 2016; 115:1451-1456. [PMID: 27875520 PMCID: PMC5155371 DOI: 10.1038/bjc.2016.385] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/12/2016] [Accepted: 10/23/2016] [Indexed: 12/23/2022] Open
Abstract
The biguanide drug metformin profoundly affects cell metabolism, causing an impairment of the cell energy balance and triggering a plethora of pleiotropic effects that vary depending on the cellular or environmental context. Interestingly, a decade ago, it was observed that metformin-treated diabetic patients have a significantly lower cancer risk. Although a variety of in vivo and in vitro observations emphasising the role of metformin as anticancer drug have been reported, the underlying mechanisms are still poorly understood. Here, we discuss our current understanding of the molecular mechanisms that are perturbed by metformin treatment and that might be relevant to understand its antitumour activities. We focus on the cell-autonomous mechanisms modulating growth and death of cancer cells.
Collapse
Affiliation(s)
- Francesca Sacco
- Department of Biochemistry, Max Plank Institute, Martinsried (Munich) 82152, Germany
| | - Alberto Calderone
- IBBE-CNR at the Bioinformatics and Computational Biology Unit, Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| |
Collapse
|
156
|
Kung CP, Murphy ME. The role of the p53 tumor suppressor in metabolism and diabetes. J Endocrinol 2016; 231:R61-R75. [PMID: 27613337 PMCID: PMC5148674 DOI: 10.1530/joe-16-0324] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/12/2022]
Abstract
In the context of tumor suppression, p53 is an undisputedly critical protein. Functioning primarily as a transcription factor, p53 helps fend off the initiation and progression of tumors by inducing cell cycle arrest, senescence or programmed cell death (apoptosis) in cells at the earliest stages of precancerous development. Compelling evidence, however, suggests that p53 is involved in other aspects of human physiology, including metabolism. Indeed, recent studies suggest that p53 plays a significant role in the development of metabolic diseases, including diabetes, and further that p53's role in metabolism may also be consequential to tumor suppression. Here, we present a review of the literature on the role of p53 in metabolism, diabetes, pancreatic function, glucose homeostasis and insulin resistance. Additionally, we discuss the emerging role of genetic variation in the p53 pathway (single-nucleotide polymorphisms) on the impact of p53 in metabolic disease and diabetes. A better understanding of the relationship between p53, metabolism and diabetes may one day better inform the existing and prospective therapeutic strategies to combat this rapidly growing epidemic.
Collapse
Affiliation(s)
- Che-Pei Kung
- Department of Internal MedicineWashington University School of Medicine, St Louis, Missouri, USA
| | - Maureen E Murphy
- Department of Internal MedicineWashington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
157
|
Floyd JS, Psaty BM. The Application of Genomics in Diabetes: Barriers to Discovery and Implementation. Diabetes Care 2016; 39:1858-1869. [PMID: 27926887 PMCID: PMC5079615 DOI: 10.2337/dc16-0738] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/16/2016] [Indexed: 02/03/2023]
Abstract
The emerging availability of genomic and electronic health data in large populations is a powerful tool for research that has drawn interest in bringing precision medicine to diabetes. In this article, we discuss the potential application of genomics to the prediction, prevention, and treatment of diabetes, and we use examples from other areas of medicine to illustrate some of the challenges involved in conducting genomics research in human populations and implementing findings in practice. At this time, a major barrier to the application of genomics in diabetes care is the lack of actionable genomic findings. Whether genomic information should be used in clinical practice requires a framework for evaluating the validity and clinical utility of this approach, an improved integration of genomic data into electronic health records, and the clinical decision support and educational resources for clinicians to use these data. Efforts to identify optimal approaches in all of these domains are in progress and may help to bring diabetes into the era of genomic medicine.
Collapse
Affiliation(s)
- James S Floyd
- Cardiovascular Health Research Unit and Departments of Epidemiology and Medicine, University of Washington, Seattle, WA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit and Departments of Epidemiology and Medicine, University of Washington, Seattle, WA
- Department of Health Services, University of Washington, Seattle, WA
- Group Health Research Institute, Seattle, WA
| |
Collapse
|
158
|
Goswami S, Yee SW, Xu F, Sridhar SB, Mosley JD, Takahashi A, Kubo M, Maeda S, Davis RL, Roden DM, Hedderson MM, Giacomini KM, Savic RM. A Longitudinal HbA1c Model Elucidates Genes Linked to Disease Progression on Metformin. Clin Pharmacol Ther 2016; 100:537-547. [PMID: 27415606 PMCID: PMC5534241 DOI: 10.1002/cpt.428] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 12/20/2022]
Abstract
One-third of type-2 diabetic patients respond poorly to metformin. Despite extensive research, the impact of genetic and nongenetic factors on long-term outcome is unknown. In this study we combine nonlinear mixed effect modeling with computational genetic methodologies to identify predictors of long-term response. In all, 1,056 patients contributed their genetic, demographic, and long-term HbA1c data. The top nine variants (of 12,000 variants in 267 candidate genes) accounted for approximately one-third of the variability in the disease progression parameter. Average serum creatinine level, age, and weight were determinants of symptomatic response; however, explaining negligible variability. Two single nucleotide polymorphisms (SNPs) in CSMD1 gene (rs2617102, rs2954625) and one SNP in a pharmacologically relevant SLC22A2 gene (rs316009) influenced disease progression, with minor alleles leading to less and more favorable outcomes, respectively. Overall, our study highlights the influence of genetic factors on long-term HbA1c response and provides a computational model, which when validated, may be used to individualize treatment.
Collapse
Affiliation(s)
- S Goswami
- University of California, San Francisco, San Francisco, California, USA
| | - S W Yee
- University of California, San Francisco, San Francisco, California, USA
| | - F Xu
- Kaiser Permanente Northern California, Oakland, California, USA
| | - S B Sridhar
- Kaiser Permanente Northern California, Oakland, California, USA
| | - J D Mosley
- Vanderbilt University, Nashville, Tennessee, USA
| | - A Takahashi
- RIKEN Institute, Center for Genomic Medicine, Saitama, Japan
| | - M Kubo
- RIKEN Institute, Center for Genomic Medicine, Saitama, Japan
| | - S Maeda
- RIKEN Institute, Center for Genomic Medicine, Saitama, Japan
| | - R L Davis
- Kaiser Permanente Georgia, Atlanta, Georgia, USA
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - D M Roden
- Vanderbilt University, Nashville, Tennessee, USA
| | - M M Hedderson
- Kaiser Permanente Northern California, Oakland, California, USA
| | - K M Giacomini
- University of California, San Francisco, San Francisco, California, USA.
| | - R M Savic
- University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
159
|
U/G SNP rs111904020 in 3′UTR of STAT3 regulated by miR-214 promotes hepatocellular carcinoma development in Chinese population. Tumour Biol 2016; 37:14629-14635. [DOI: 10.1007/s13277-016-5352-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/07/2016] [Indexed: 01/04/2023] Open
|
160
|
Chang C, Zhang K, Veluchamy A, Hébert HL, Looker HC, Colhoun HM, Palmer CNA, Meng W. A Genome-Wide Association Study Provides New Evidence That CACNA1C Gene is Associated With Diabetic Cataract. Invest Ophthalmol Vis Sci 2016; 57:2246-50. [PMID: 27124316 PMCID: PMC4855826 DOI: 10.1167/iovs.16-19332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Purpose Diabetic cataract is one of the major eye complications of diabetes. It was reported that cataract occurs two to five times more frequently in patients with diabetes compared with those with no diabetes. The purpose of this study was to identify genetic contributors of diabetic cataract based on a genome-wide association approach using a well-defined Scottish diabetic cohort. Methods We adapted linked e-health records to define diabetic cataract. A diabetic cataract case in this study was defined as a type 2 diabetic patient who has ever been recorded in the linked e-health records to have cataracts in both eyes or who had previous cataract extraction surgeries in at least one eye. A control in this study was defined as a type 2 diabetic individual who has never been diagnosed as cataract in the linked e-health records and had no history of cataract surgeries. A standard genome-wide association approach was applied. Results Overall, we have 2341 diabetic cataract cases and 2878 controls in the genetics of diabetes audit and research in Tayside Scotland (GoDARTS) dataset. We found that the P value of rs2283290 in the CACNA1C gene was 8.81 × 10−10, which has reached genome-wide significance. We also identified that the blood calcium level was statistically different between diabetic cataract cases and controls. Conclusions We identified supporting evidence that CACNA1C gene is associated with diabetic cataract. The role of calcium in the cataractogenesis needs to be reevaluated in future studies.
Collapse
Affiliation(s)
- Cheng Chang
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Kaida Zhang
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Abirami Veluchamy
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Harry L Hébert
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Helen C Looker
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Helen M Colhoun
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Colin N A Palmer
- Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Weihua Meng
- Division of Population Health Sciences Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
161
|
Zhou K, Yee SW, Seiser EL, van Leeuwen N, Tavendale R, Bennett AJ, Groves CJ, Coleman RL, van der Heijden AA, Beulens JW, de Keyser CE, Zaharenko L, Rotroff DM, Out M, Jablonski KA, Chen L, Javorský M, Židzik J, Levin AM, Williams LK, Dujic T, Semiz S, Kubo M, Chien HC, Maeda S, Witte JS, Wu L, Tkáč I, Kooy A, van Schaik RHN, Stehouwer CDA, Logie L, MetGen Investigators, DPP Investigators, ACCORD Investigators, Sutherland C, Klovins J, Pirags V, Hofman A, Stricker BH, Motsinger-Reif AA, Wagner MJ, Innocenti F, 't Hart LM, Holman RR, McCarthy MI, Hedderson MM, Palmer CNA, Florez JC, Giacomini KM, Pearson ER. Variation in the glucose transporter gene SLC2A2 is associated with glycemic response to metformin. Nat Genet 2016; 48:1055-1059. [PMID: 27500523 PMCID: PMC5007158 DOI: 10.1038/ng.3632] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/30/2016] [Indexed: 02/06/2023]
Abstract
Metformin is the first-line antidiabetic drug with over 100 million users worldwide, yet its mechanism of action remains unclear. Here the Metformin Genetics (MetGen) Consortium reports a three-stage genome-wide association study (GWAS), consisting of 13,123 participants of different ancestries. The C allele of rs8192675 in the intron of SLC2A2, which encodes the facilitated glucose transporter GLUT2, was associated with a 0.17% (P = 6.6 × 10(-14)) greater metformin-induced reduction in hemoglobin A1c (HbA1c) in 10,577 participants of European ancestry. rs8192675 was the top cis expression quantitative trait locus (cis-eQTL) for SLC2A2 in 1,226 human liver samples, suggesting a key role for hepatic GLUT2 in regulation of metformin action. Among obese individuals, C-allele homozygotes at rs8192675 had a 0.33% (3.6 mmol/mol) greater absolute HbA1c reduction than T-allele homozygotes. This was about half the effect seen with the addition of a DPP-4 inhibitor, and equated to a dose difference of 550 mg of metformin, suggesting rs8192675 as a potential biomarker for stratified medicine.
Collapse
Affiliation(s)
- Kaixin Zhou
- School of Medicine, University of Dundee, Dundee, UK
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Eric L Seiser
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Nienke van Leeuwen
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Amanda J Bennett
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Christopher J Groves
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Ruth L Coleman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Amber A van der Heijden
- Department of General Practice, EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Joline W Beulens
- Department of Epidemiology and Biostatistics, EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Linda Zaharenko
- Latvian Genome Data Base (LGDB), Riga, Latvia
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Daniel M Rotroff
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Mattijs Out
- Treant Zorggroep, Location Bethesda, Hoogeveen, the Netherlands
- Bethesda Diabetes Research Centre, Hoogeveen, the Netherlands
| | | | - Ling Chen
- Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Jozef Židzik
- Faculty of Medicine, Šafárik University, Košice, Slovakia
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan, USA
| | - L Keoki Williams
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, Michigan, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, USA
| | - Tanja Dujic
- School of Medicine, University of Dundee, Dundee, UK
- Faculty of Pharmacy, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Sabina Semiz
- Faculty of Pharmacy, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
- Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Huan-Chieh Chien
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara, Japan
| | - John S Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
- Department of Urology, University of California, San Francisco, San Francisco, California, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, USA
| | - Longyang Wu
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Ivan Tkáč
- Faculty of Medicine, Šafárik University, Košice, Slovakia
| | - Adriaan Kooy
- Treant Zorggroep, Location Bethesda, Hoogeveen, the Netherlands
- Bethesda Diabetes Research Centre, Hoogeveen, the Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine and Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Lisa Logie
- School of Medicine, University of Dundee, Dundee, UK
| | | | | | | | | | - Janis Klovins
- Latvian Genome Data Base (LGDB), Riga, Latvia
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Valdis Pirags
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
- Department of Endocrinology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Inspectorate of Healthcare, Heerlen, the Netherlands
| | - Alison A Motsinger-Reif
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| | - Michael J Wagner
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, Center for Pharmacogenomics and Individualized Therapy, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Leen M 't Hart
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Epidemiology and Biostatistics, EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rury R Holman
- Diabetes Trials Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Monique M Hedderson
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | | | - Jose C Florez
- Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Metabolism, Broad Institute, Cambridge, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
162
|
Fodor A, Karnieli E. Challenges of implementing personalized (precision) medicine: a focus on diabetes. Per Med 2016; 13:485-497. [DOI: 10.2217/pme-2016-0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The concept of personalized (precision) medicine (PM) emphasizes the scientific and technological innovations that enable the physician to tailor disease prediction, diagnosis and treatment to the individual patient, based on a personalized data-driven approach. The major challenge for the medical systems is to translate the molecular and genomic advances into clinical available means. Patients and healthcare providers, the pharmaceutical and diagnostic industries manifest a growing interest in PM. Multiple stakeholders need adaptation and re-engineering for successful clinical implementation of PM. Drawing primarily from the field of ‘diabetes’, this article will summarize the main challenges to implementation of PM into current medical practice and some of the approaches currently being implemented to overcome these challenges.
Collapse
Affiliation(s)
- Adriana Fodor
- University of Medicine & Pharmacy 'Iuliu Hatieganu', Cluj-Napoca, Romania
| | - Eddy Karnieli
- Galil Center for Telemedicine, Medical Informatics & Personalized Medicine, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
163
|
Javorský M, Gotthardová I, Klimčáková L, Kvapil M, Židzik J, Schroner Z, Doubravová P, Gala I, Dravecká I, Tkáč I. A missense variant in GLP1R gene is associated with the glycaemic response to treatment with gliptins. Diabetes Obes Metab 2016; 18:941-4. [PMID: 27160388 DOI: 10.1111/dom.12682] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/27/2016] [Accepted: 05/01/2016] [Indexed: 12/25/2022]
Abstract
Gliptins act by increasing endogenous incretin levels. Glucagon-like peptide-1 receptor (GLP1R) and glucose-dependent insulinotropic peptide receptor (GIPR) are their indirect drug targets. Variants of GLP1R and GIPR have previously been associated with the incretin effect. The aim of the present pilot study was to examine associations of the GLP1R and GIPR gene variants with the glycaemic response to gliptins. A total of 140 consecutive patients with type 2 diabetes were followed-up 6 months after initiation of gliptin treatment. GLP1R rs6923761 (Gly168Ser) and GIPR rs10423928 genotyping was performed using real-time PCR, with subsequent high-resolution melting analysis. The main study outcome was reduction in glycated haemoglobin (HbA1c) after treatment. GLP1R Gly168Ser variant was significantly associated with reduction in HbA1c in an additive model (β = -0.33, p = 0.011). The mean reduction in HbA1c in Ser/Ser homozygotes was significantly lower compared with Gly-allele carriers [0.12 ± 0.23% vs. 0.80 ± 0.09% (1.3 ± 2.5 mmol/mol vs. 8.7 ± 1.0 mmol/mol); p = 0.008]. In conclusion, GLP1R missense variant was associated with a reduced response to gliptin treatment. The genotype-related effect size of ∼0.7% (8 mmol/mol) is equal to an average effect of gliptin treatment and makes this variant a candidate for use in precision medicine.
Collapse
Affiliation(s)
- M Javorský
- Faculty of Medicine, Department of Internal Medicine 4, Šafárik University, Košice, Slovakia
- Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| | - I Gotthardová
- Faculty of Medicine, Department of Internal Medicine 4, Šafárik University, Košice, Slovakia
- Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| | - L Klimčáková
- Faculty of Medicine, Department of Internal Medicine 4, Šafárik University, Košice, Slovakia
- Faculty of Medicine, Department of Medicine Biology, Šafárik University, Košice, Slovakia
| | - M Kvapil
- Faculty of Medicine 2, Department of Medicine, Charles University, Praha, Czech Republic
- Faculty Hospital in Motol, Department of Medicine, Praha, Czech Republic
| | - J Židzik
- Faculty of Medicine, Department of Internal Medicine 4, Šafárik University, Košice, Slovakia
| | - Z Schroner
- Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| | - P Doubravová
- Faculty Hospital in Motol, Department of Medicine, Praha, Czech Republic
| | - I Gala
- Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| | - I Dravecká
- Faculty of Medicine, Department of Internal Medicine 4, Šafárik University, Košice, Slovakia
- Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| | - I Tkáč
- Faculty of Medicine, Department of Internal Medicine 4, Šafárik University, Košice, Slovakia
- Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| |
Collapse
|
164
|
Sam WJ, Roza O, Hon YY, Alfaro RM, Calis KA, Reynolds JC, Yanovski JA. Effects of SLC22A1 Polymorphisms on Metformin-Induced Reductions in Adiposity and Metformin Pharmacokinetics in Obese Children With Insulin Resistance. J Clin Pharmacol 2016; 57:219-229. [PMID: 27407018 DOI: 10.1002/jcph.796] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 12/27/2022]
Abstract
Steady-state population pharmacokinetics of a noncommercial immediate-release metformin (hydrochloride) drug product were characterized in 28 severely obese children with insulin resistance. The concentration-time profiles with double peaks were well described by a 1-compartment model with 2 absorption sites. Mean population apparent clearance (CL/F) was 68.1 L/h, and mean apparent volume of distribution (V/F) was 28.8 L. Body weight was a covariate of CL/F and V/F. Estimated glomerular filtration rate was a significant covariate of CL/F (P < .001). SLC22A1 genotype did not significantly affect metformin pharmacokinetics. The response to 6 months of metformin treatment (HbA1c , homeostasis model assessment for insulin resistance, fasting insulin, and glucose changes) did not differ between SLC22A1 wild-type subjects and carriers of presumably low-activity SLC22A1 alleles. However, SLC22A1 variant carriers had smaller reductions in percentage of total trunk fat after metformin therapy, although the percentage reduction in trunk fat was small. The median % change in trunk fat was -2.20% (-9.00% to 0.900%) and -1.20% (-2.40% to 7.30%) for the SLC22A1 wild-type subjects and variant carriers, respectively. Future study is needed to evaluate the effects of SLC22A1 polymorphisms on metformin-mediated weight reduction in obese children.
Collapse
Affiliation(s)
- Wai Johnn Sam
- Clinical Pharmacokinetics Research Laboratory, Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Orsolya Roza
- Section on Growth and Obesity, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.,Institute of Pharmacognosy, University of Szeged, Szeged, Hungary
| | - Yuen Yi Hon
- Clinical Pharmacokinetics Research Laboratory, Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA.,Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Raul M Alfaro
- Clinical Pharmacokinetics Research Laboratory, Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Karim A Calis
- Clinical Pharmacokinetics Research Laboratory, Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA.,Office of the Clinical Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - James C Reynolds
- Nuclear Medicine Division, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Jack A Yanovski
- Section on Growth and Obesity, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
165
|
Yang P, Heredia VO, Beltramo DM, Soria NW. Pharmacogenetics and personalized treatment of type 2 diabetes mellitus. Int J Diabetes Dev Ctries 2016. [DOI: 10.1007/s13410-016-0517-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
166
|
Lyssenko V, Bianchi C, Del Prato S. Personalized Therapy by Phenotype and Genotype. Diabetes Care 2016; 39 Suppl 2:S127-36. [PMID: 27440825 DOI: 10.2337/dcs15-3002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Valeriya Lyssenko
- Department of Translational Pathophysiology, Steno Diabetes Center A/S, Gentofte, Denmark Diabetes and Endocrinology, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Cristina Bianchi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Del Prato
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
167
|
Pharmacogenomics in type 2 diabetes: oral antidiabetic drugs. THE PHARMACOGENOMICS JOURNAL 2016; 16:399-410. [DOI: 10.1038/tpj.2016.54] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/08/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023]
|
168
|
Abstract
As with other complex diseases, unbiased association studies followed by physiological and experimental characterization have for years formed a paradigm for identifying genes or processes of relevance to type 2 diabetes mellitus (T2D). Recent large-scale common and rare variant genome-wide association studies (GWAS) suggest that substantially larger association studies are needed to identify most T2D loci in the population. To hasten clinical translation of genetic discoveries, new paradigms are also required to aid specialized investigation of nascent hypotheses. We argue for an integrated T2D knowledgebase, designed for a worldwide community to access aggregated large-scale genetic data sets, as one paradigm to catalyse convergence of these efforts.
Collapse
|
169
|
Thomas PPM, Alshehri SM, van Kranen HJ, Ambrosino E. The impact of personalized medicine of Type 2 diabetes mellitus in the global health context. Per Med 2016; 13:381-393. [DOI: 10.2217/pme-2016-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Advances in the fields of genomic sciences have given rise to personalized medicine. This new paradigm draws upon a patient's genetic and metabolic makeup in order to tailor diagnostics and treatment. Personalized medicine holds remarkable promises to improve prevention and management of chronic diseases of global relevance, such as Type 2 diabetes mellitus (T2DM). This review article aims at summarizing the evidence from genome-based sciences on T2DM risk and management in different populations and in the Global Health context. Opinions from leading experts in the field were also included. Based on these findings, strengths and weaknesses of personalized approach to T2DM in a global context are delineated. Implications for future research and implementation on that subject are discussed.
Collapse
Affiliation(s)
- Pierre Paul Michel Thomas
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
| | - Salih Mohammed Alshehri
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
| | - Henk J van Kranen
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
- National Institute for Public Health & the Environment, Bilthoven 3721 MA, The Netherlands
| | - Elena Ambrosino
- Institute for Public Health Genomics, Department of Genetics & CellBiology, School for Oncology & Developmental Biology (GROW), Faculty of Health, Medicine & LifeSciences, Maastricht University, Maastricht 6200 MD, The Netherlands
| |
Collapse
|
170
|
Florez JC. Precision Medicine in Diabetes: Is It Time? Diabetes Care 2016; 39:1085-8. [PMID: 27289125 DOI: 10.2337/dc16-0586] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/23/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Jose C Florez
- Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA; Metabolism Program and Program in Medical and Population Genetics, Broad Institute, Cambridge, MA; and Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
171
|
Abstract
In this Perspective, Jose Florez discusses how information from genetics and genomics may be able to contribute to prevention of type 2 diabetes and predicting individual responses to behavioral and other interventions.
Collapse
|
172
|
Abstract
Personalized medicine aims at better targeting therapeutic intervention to the individual to maximize benefit and minimize harm. Type 2 diabetes (T2D) is a heterogeneous disease from a genetic, pathophysiological and clinical point of view. Thus, the response to any antidiabetic medication may considerably vary between individuals. Numerous glucose-lowering agents, with different mechanisms of action, have been developed, a diversified armamentarium that offers the possibility of a patient-centred therapeutic approach. In the current clinical practice, a personalized approach is only based upon phenotype, taking into account patient and disease individual characteristics. If this approach may help increase both efficacy and safety outcomes, there remains considerable room for improvement. In recent years, many efforts were taken to identify genetic and genotype SNP's (Single Nucleotide Polymorphism's) variants that influence the pharmacokinetics, pharmacodynamics, and ultimately the therapeutic response of oral glucose-lowering drugs. This approach mainly concerns metformin, sulphonylureas, meglitinides and thiazolidinediones, with only scarce data concerning gliptins and gliflozins yet. However, the contribution of pharmacogenetics and pharmacogenomics to personalized therapy still needs to mature greatly before routine clinical implementation is possible. This review discusses both opportunities and challenges of precision medicine and how this new paradigm may lead to a better individualized treatment of T2D.
Collapse
Affiliation(s)
- André J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, University of Liège, Liège, Belgium; Clinical Pharmacology Unit, CHU Liège, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium.
| |
Collapse
|
173
|
Endocrine abnormalities in ataxia telangiectasia: findings from a national cohort. Pediatr Res 2016; 79:889-94. [PMID: 26891003 DOI: 10.1038/pr.2016.19] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/21/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ataxia telangiectasia (AT) is a genetic multisystem disorder, presenting with progressive ataxia, immune deficiency, and propensity toward malignancy. Endocrine abnormalities (growth retardation, reproductive dysfunction, and diabetes) have been described, however detailed information regarding this aspect is lacking. We aimed to characterize endocrine anomalies and growth patterns in a large cohort of AT patients. METHODS Retrospective study comprising all 52 patients (aged 2-26.2 y) followed at a national AT Clinic. Anthropometric and laboratory measurements were extracted from the charts. RESULTS Median height-SDS was already subnormal during infancy, remaining negative throughout follow up to adulthood. Height-SDS was more impaired than weight-SDS up to age 4 y, thereafter weight-SDS steadily decreased, resulting in progressively lower BMI-SDS. IGF-I-SDS was low (-1.53 ± 1.54), but did not correlate with height-SDS. Gonadal failure was present in all 13 females older than 10 y but only in one male. Two patients had diabetes and 10 had dyslipidemia. Vitamin D deficiency was observed in 52.2% of the evaluated patients. CONCLUSION Our results suggest a primary growth abnormality in AT, rather than secondary to nutritional impairment or disease severity. Sex hormone replacement should be considered for female patients. Vitamin D levels should be followed and supplementation given if needed.
Collapse
|
174
|
Abstract
Personalized medicine, otherwise called stratified or precision medicine, aims to better target intervention to the individual to maximize benefit and minimize harm. This review discusses how diabetes aetiology, pathophysiology and patient genotype influence response to or side effects of the commonly used diabetes treatments. C-peptide is a useful biomarker that is underused to guide treatment choice, severe insulin deficiency predicts non-response to glucagon-like peptide-1 receptor agonists, and thiazolidinediones are more effective in insulin-resistant patients. The field of pharmacogenetics is now yielding clinically important results, with three examples outlined: sulphonylurea sensitivity in patients with HNF1A maturity-onset diabetes of the young; sulphonylurea sensitivity in patients with Type 2 diabetes with reduced function alleles at CYP2C9, resulting in reduced metabolism of sulphonylureas; and severe metformin intolerance associated with reduced function organic cation transporter 1 (OCT1) variants, exacerbated by drugs that also inhibit OCT1. Genome-wide approaches and the potential of other 'omics', including metagenomics and metabolomics, are then outlined, highlighting the complex interacting networks that we need to understand before we can truly personalize diabetes treatments.
Collapse
Affiliation(s)
- E R Pearson
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, UK
| |
Collapse
|
175
|
Zhou K, Pedersen HK, Dawed AY, Pearson ER. Pharmacogenomics in diabetes mellitus: insights into drug action and drug discovery. Nat Rev Endocrinol 2016; 12:337-46. [PMID: 27062931 DOI: 10.1038/nrendo.2016.51] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Genomic studies have greatly advanced our understanding of the multifactorial aetiology of type 2 diabetes mellitus (T2DM) as well as the multiple subtypes of monogenic diabetes mellitus. In this Review, we discuss the existing pharmacogenetic evidence in both monogenic diabetes mellitus and T2DM. We highlight mechanistic insights from the study of adverse effects and the efficacy of antidiabetic drugs. The identification of extreme sulfonylurea sensitivity in patients with diabetes mellitus owing to heterozygous mutations in HNF1A represents a clear example of how pharmacogenetics can direct patient care. However, pharmacogenomic studies of response to antidiabetic drugs in T2DM has yet to be translated into clinical practice, although some moderate genetic effects have now been described that merit follow-up in trials in which patients are selected according to genotype. We also discuss how future pharmacogenomic findings could provide insights into treatment response in diabetes mellitus that, in addition to other areas of human genetics, facilitates drug discovery and drug development for T2DM.
Collapse
Affiliation(s)
- Kaixin Zhou
- School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Helle Krogh Pedersen
- Department of Systems Biology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Adem Y Dawed
- School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Ewan R Pearson
- School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| |
Collapse
|
176
|
Tkáč I, Gotthardová I. Pharmacogenetic aspects of the treatment of Type 2 diabetes with the incretin effect enhancers. Pharmacogenomics 2016; 17:795-804. [PMID: 27166975 DOI: 10.2217/pgs-2016-0011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Incretin effect enhancers are drugs used in the treatment of Type 2 diabetes and include GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors (gliptins). Variants in several genes were shown to be involved in the physiology of incretin secretion. Only two gene variants have evidence also from pharmacogenetic studies. TCF7L2 rs7903146 C>T and CTRB1/2 rs7202877 T>G minor allele carriers were both associated with a smaller reduction in HbA1c after gliptin treatment when compared with major allele carriers. After replication in further studies, these observations could be of clinical significance in helping to identify patients with potentially lower or higher response to gliptin treatment.
Collapse
Affiliation(s)
- Ivan Tkáč
- Department of Internal Medicine 4, Šafárik University, Faculty of Medicine, Rastislavova 43, 041 90 Košice, Slovakia.,Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| | - Ivana Gotthardová
- Department of Internal Medicine 4, Šafárik University, Faculty of Medicine, Rastislavova 43, 041 90 Košice, Slovakia.,Department of Internal Medicine 4, Pasteur University Hospital, Košice, Slovakia
| |
Collapse
|
177
|
Dawed AY, Zhou K, Pearson ER. Pharmacogenetics in type 2 diabetes: influence on response to oral hypoglycemic agents. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2016; 9:17-29. [PMID: 27103840 PMCID: PMC4827904 DOI: 10.2147/pgpm.s84854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is one of the leading causes of morbidity and mortality, consuming a significant proportion of public health spending. Oral hypoglycemic agents (OHAs) are the frontline treatment approaches after lifestyle changes. However, huge interindividual variation in response to OHAs results in unnecessary treatment failure. In addition to nongenetic factors, genetic factors are thought to contribute to much of such variability, highlighting the importance of the potential of pharmacogenetics to improve therapeutic outcome. Despite the presence of conflicting results, significant progress has been made in an effort to identify the genetic markers associated with pharmacokinetics, pharmacodynamics, and ultimately therapeutic response and/or adverse outcomes to OHAs. As such, this article presents a comprehensive review of current knowledge on pharmacogenetics of OHAs and provides insights into knowledge gaps and future directions.
Collapse
Affiliation(s)
- Adem Yesuf Dawed
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Kaixin Zhou
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| | - Ewan Robert Pearson
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
178
|
Nelson MR, Johnson T, Warren L, Hughes AR, Chissoe SL, Xu CF, Waterworth DM. The genetics of drug efficacy: opportunities and challenges. Nat Rev Genet 2016; 17:197-206. [PMID: 26972588 DOI: 10.1038/nrg.2016.12] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lack of sufficient efficacy is the most common cause of attrition in late-phase drug development. It has long been envisioned that genetics could drive stratified drug development by identifying those patient subgroups that are most likely to respond. However, this vision has not been realized as only a small proportion of drugs have been found to have germline genetic predictors of efficacy with clinically meaningful effects, and so far all but one were found after drug approval. With the exception of oncology, systematic application of efficacy pharmacogenetics has not been integrated into drug discovery and development across the industry. Here, we argue for routine, early and cumulative screening for genetic predictors of efficacy, as an integrated component of clinical trial analysis. Such a strategy would identify clinically relevant predictors that may exist at the earliest possible opportunity, allow these predictors to be integrated into subsequent clinical development and provide mechanistic insights into drug disposition and patient-specific factors that influence response, therefore paving the way towards more personalized medicine.
Collapse
Affiliation(s)
- Matthew R Nelson
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| | - Toby Johnson
- Target Sciences, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Liling Warren
- GlaxoSmithKline, Durham, North Carolina 27713, USA
- Acclarogen, Cambridge CB4 0WS, UK
| | - Arlene R Hughes
- PAREXEL International, Research Triangle Park, North Carolina 27713, USA
| | | | - Chun-Fang Xu
- Target Sciences, GlaxoSmithKline, Stevenage SG1 2NY, UK
| | - Dawn M Waterworth
- Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania 19406, USA
| |
Collapse
|
179
|
Sacco F, Silvestri A, Posca D, Pirrò S, Gherardini PF, Castagnoli L, Mann M, Cesareni G. Deep Proteomics of Breast Cancer Cells Reveals that Metformin Rewires Signaling Networks Away from a Pro-growth State. Cell Syst 2016; 2:159-71. [PMID: 27135362 DOI: 10.1016/j.cels.2016.02.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/02/2015] [Accepted: 02/01/2016] [Indexed: 12/25/2022]
Abstract
Metformin is the most frequently prescribed drug for type 2 diabetes. In addition to its hypoglycemic effects, metformin also lowers cancer incidence. This anti-cancer activity is incompletely understood. Here, we profiled the metformin-dependent changes in the proteome and phosphoproteome of breast cancer cells using high-resolution mass spectrometry. In total, we quantified changes of 7,875 proteins and 15,813 phosphosites after metformin changes. To interpret these datasets, we developed a generally applicable strategy that overlays metformin-dependent changes in the proteome and phosphoproteome onto a literature-derived network. This approach suggested that metformin treatment makes cancer cells more sensitive to apoptotic stimuli and less sensitive to pro-growth stimuli. These hypotheses were tested in vivo; as a proof-of-principle, we demonstrated that metformin inhibits the p70S6K-rpS6 axis in a PP2A-phosphatase dependent manner. In conclusion, analysis of deep proteomics reveals both detailed and global mechanisms that contribute to the anti-cancer activity of metformin.
Collapse
Affiliation(s)
- Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Daniela Posca
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Pirrò
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Matthias Mann
- Department Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany.
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Istituto Ricovero e Cura a Carattere Scientifico, Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
180
|
Guleria A, Chandna S. ATM kinase: Much more than a DNA damage responsive protein. DNA Repair (Amst) 2016; 39:1-20. [PMID: 26777338 DOI: 10.1016/j.dnarep.2015.12.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 12/21/2015] [Accepted: 12/21/2015] [Indexed: 11/22/2022]
Abstract
ATM, mutation of which causes Ataxia telangiectasia, has emerged as a cardinal multifunctional protein kinase during past two decades as evidenced by various studies from around the globe. Further to its well established and predominant role in DNA damage response, ATM has also been understood to help in maintaining overall functional integrity of cells; since its mutation, inactivation or deficiency results in a variety of pathological manifestations besides DNA damage. These include oxidative stress, metabolic syndrome, mitochondrial dysfunction as well as neurodegeneration. Recently, high throughput screening using proteomics, metabolomics and transcriptomic studies revealed several proteins which might be acting as substrates of ATM. Studies that can help in identifying effective regulatory controls within the ATM-mediated pathways/mechanisms can help in developing better therapeutics. In fact, more in-depth understanding of ATM-dependent cellular signals could also help in the treatment of variety of other disease conditions since these pathways seem to control many critical cellular functions. In this review, we have attempted to put together a detailed yet lucid picture of the present-day understanding of ATM's role in various pathophysiological conditions involving DNA damage and beyond.
Collapse
Affiliation(s)
- Ayushi Guleria
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Delhi 110054, India
| | - Sudhir Chandna
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Delhi 110054, India.
| |
Collapse
|
181
|
Connelly PJ, Smith N, Chadwick R, Exley AR, Shneerson JM, Pearson ER. Recessive mutations in the cancer gene Ataxia Telangiectasia Mutated (ATM), at a locus previously associated with metformin response, cause dysglycaemia and insulin resistance. Diabet Med 2016; 33:371-5. [PMID: 26606753 PMCID: PMC4832393 DOI: 10.1111/dme.13037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/18/2015] [Indexed: 12/31/2022]
Abstract
AIM To investigate glucose and insulin metabolism in participants with ataxia telangiectasia in the absence of a diagnosis of diabetes. METHODS A standard oral glucose tolerance test was performed in participants with ataxia telangiectasia (n = 10) and in a control cohort (n = 10). Serial glucose and insulin measurements were taken to permit cohort comparisons of glucose-insulin homeostasis and indices of insulin secretion and sensitivity. RESULTS During the oral glucose tolerance test, the 2-h glucose (6.75 vs 4.93 mmol/l; P = 0.029), insulin concentrations (285.6 vs 148.5 pmol/l; P = 0.043), incremental area under the curve for glucose (314 vs 161 mmol/l/min; P = 0.036) and incremental area under the curve for insulin (37,720 vs 18,080 pmol/l/min; P = 0.03) were higher in participants with ataxia telangiectasia than in the controls. There were no significant differences between groups in fasting glucose, insulin concentrations or insulinogenic index measurement (0.94 vs 0.95; P = 0.95). The Matsuda index, reflecting whole-body insulin sensitivity, was lower in participants with ataxia telangiectasia (5.96 vs 11.03; P = 0.019) than in control subjects. CONCLUSIONS Mutations in Ataxia Telangiectasia Mutated (ATM) that cause ataxia telangiectasia are associated with elevated glycaemia and low insulin sensitivity in participants without diabetes. This indicates a role of ATM in glucose and insulin metabolic pathways.
Collapse
Affiliation(s)
- P J Connelly
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, UK
| | - N Smith
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, UK
| | - R Chadwick
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - A R Exley
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - J M Shneerson
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - E R Pearson
- Division of Cardiovascular and Diabetes Medicine, Medical Research Institute, University of Dundee, Dundee, UK
| |
Collapse
|
182
|
Staiger H, Schaeffeler E, Schwab M, Häring HU. Pharmacogenetics: Implications for Modern Type 2 Diabetes Therapy. Rev Diabet Stud 2016; 12:363-76. [PMID: 27111121 DOI: 10.1900/rds.2015.12.363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Many clinical treatment studies have reported remarkable interindividual variability in the response to pharmaceutical drugs, and uncovered the existence of inadequate treatment response, non-response, and even adverse drug reactions. Pharmacogenetics addresses the impact of genetic variants on treatment outcome including side-effects. In recent years, it has also entered the field of clinical diabetes research. In modern type 2 diabetes therapy, metformin is established as first-line drug. The latest pharmaceutical developments, including incretin mimetics, dipeptidyl peptidase 4 inhibitors (gliptins), and sodium/glucose cotransporter 2 inhibitors (gliflozins), are currently experiencing a marked increase in clinical use, while the prescriptions of α-glucosidase inhibitors, sulfonylureas, meglitinides (glinides), and thiazolidinediones (glitazones) are declining, predominantly because of reported side-effects. This review summarizes the current knowledge about gene-drug interactions observed in therapy studies with the above drugs. We report drug interactions with candidate genes involved in the pharmacokinetics (e.g., drug transporters) and pharmacodynamics (drug targets and downstream signaling steps) of the drugs, with known type 2 diabetes risk genes and previously unknown genes derived from hypothesis-free approaches such as genome-wide association studies. Moreover, some new and promising candidate genes for future pharmacogenetic assessment are highlighted. Finally, we critically appraise the current state of type 2 diabetes pharmacogenetics in the light of its impact on therapeutic decisions, and we refer to major problems, and make suggestions for future efforts in this field to help improve the clinical relevance of the results, and to establish genetically determined treatment failure.
Collapse
Affiliation(s)
- Harald Staiger
- Interfaculty Centre for Pharmacogenomics and Pharma Research at the University of Tübingen, Tübingen, Germany
| | - Elke Schaeffeler
- Interfaculty Centre for Pharmacogenomics and Pharma Research at the University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Interfaculty Centre for Pharmacogenomics and Pharma Research at the University of Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Interfaculty Centre for Pharmacogenomics and Pharma Research at the University of Tübingen, Tübingen, Germany
| |
Collapse
|
183
|
Jiang G, Luk AO, Yang X, Wang Y, Tam CHT, Lau SH, Ozaki R, Kong APS, Tong PC, Chow CC, Chan JCN, So WY, Ma RCW. Progression to treatment failure among Chinese patients with type 2 diabetes initiated on metformin versus sulphonylurea monotherapy--The Hong Kong Diabetes Registry. Diabetes Res Clin Pract 2016; 112:57-64. [PMID: 26703273 DOI: 10.1016/j.diabres.2015.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/07/2015] [Accepted: 11/09/2015] [Indexed: 11/24/2022]
Abstract
AIMS To assess the development of treatment failure in Chinese patients with type 2 diabetes mellitus (T2DM) initiated on metformin or sulphonylurea (SU) monotherapy, with consideration of various potential sources of biases. METHODS A 1:1-matched new metformin and SU user cohort on immortal time and mean propensity score after multiple imputation was selected from a cohort of 5889 Chinese patients with T2DM. Treatment failure was defined as progression to (i) combination oral anti-hyperglycemia drug therapy, (ii) insulin use, or (iii) a treatment haemoglobin A1c (HbA1c) >7.5% (58 mmol/mol). Stratified Cox regression analysis on the matched pairs was employed to examine the associations between initial monotherapy and onset of treatment failure. RESULTS Of 554 new metformin and 840 new SU users, 380 were matched. During a median follow-up duration of 3 years, 173 (45.6%) metformin users and 220 (57.9%) SU users experienced treatment failure (annual failure rates of 15% and 19%, respectively). The median time from monotherapy starting to treatment failure was 3.0 [inter-quartile range (IQR): 1.8-5.4] years for metformin users, versus 1.8 (IQR: 0.9-4.1) years for SU users (p<0.001). Stratified Cox regression analysis showed significantly lower risk of treatment failure for metformin users (HR [95% CI], 0.62[0.47-0.81]; p<0.001). Consistent results were found in analyses based on traditional adjustment schemes with or without imputation. CONCLUSIONS By systematically incorporating new-user design, multiple imputation and matching methods, we found that Chinese patients with T2DM initiated on metformin monotherapy were associated with a significant delay in the onset of treatment failure compared to SU monotherapy.
Collapse
Affiliation(s)
- Guozhi Jiang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Andrea O Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ying Wang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Siu Him Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Risa Ozaki
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Peter C Tong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chun Chung Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yee So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong.
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
184
|
Bondar IA, Shabelnikova OY, Sokolova EA, Pyankova OV, Filipenko ML. Phenotypic and genetic characteristics of patients with type 2 diabetes with different responses to metformin therapy in Novosibirsk region. DIABETES MELLITUS 2016. [DOI: 10.14341/dm2004146-47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Aim: The purpose of this study was to examine the phenotypic and genetic characteristics of patients with type 2 diabetes mellitus (T2DM) with different responses to treatment with metformin (MF) in the Novosibirsk region. Materials and methods: We examined 460 patients with T2DM in the Novosibirsk region. Patients were divided into groups according to their HbA1c level: patients who achieved the target HbA1c level during MF therapy (n = 209) and those who did not reach the target HbA1c level (n=251). Genotyping of ATM (rs11212617) was performed using polymerase chain reaction by TaqMan. Results: Patients who achieved the target HbA1c level during MF treatment (good response) were older (61. 1±9. 1 years vs. 57. 4±8. 4 years, p=0. 001), had later onset of diabetes (54. 6 ± 10. 1 years vs. 49. 2±8. 5 years, p = 0. 0001) and shorter duration of diabetes (6. 5±5. 9 years vs. 8. 2±6. 1 years, p=0. 03) compared with those who did not achieve the target HbA1c level. There was no statistically significant association between ATM rs11212617 and achieving the target HbA1c level among all patients [odds ratio (OR)=0. 94, 95% confidence interval = (0. 73–1. 23), p=0. 67] or those with MF monotherapy [OR=0. 90, (0. 65–1. 25), p=0. 54] or combination therapy [OR=1. 02, (0. 72–1. 43), p=0. 92]. There was an effect of age on response to MF therapy in all three groups (all patients: p=0. 001, MF monotherapy group: p=0. 04, combination therapy group: p=0. 0009). In the MF monotherapy group, low dose MF was associated with a good response (p=0. 03), and in the combination therapy group, males were more likely to have a good response (p=0. 003). Patients with genotype C/C or A/C for ATM (rs11212617) compared with those with genotype A/A were more likely to have high levels of triglycerides [2. 33 (1. 52–4. 2) mmol/l, 2. 09 (1. 35–3. 0) mmol/l and 1. 99 (1. 49–3. 21) mmol/l, respectively, p=0. 001], coronary heart disease (CHD) (13. 4%, 13. 4% and 9. 6%, respectively, p=0. 009) and myocardial infarction (7. 8%, 3. 2% and 4. 0%, respectively, p=0. 001). Conclusion: Patients with T2DM who had a good response to MF therapy were older, more likely to be male and had a later onset of T2DM. Genotype C/C for ATM rs11212617 was associated with high triglycerides, CHD and myocardial infarction. ATM rs11212617 was not associated with response to MF therapy in the Novosibirsk region.
Collapse
|
185
|
Shokri F, Ghaedi H, Ghafouri Fard S, Movafagh A, Abediankenari S, Mahrooz A, Kashi Z, Omrani MD. Impact of ATM and SLC22A1 Polymorphisms on Therapeutic Response to Metformin in Iranian Diabetic Patients. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2016; 5:1-7. [PMID: 27386433 PMCID: PMC4916778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Metabolic syndrome and its pathological sequel, type 2 diabetes are considered as important global health problems. Metformin is the most common drug prescribed for patients with this disorder. Consequently, understanding the genetic pathways involved in pharmacokinetics and pharmacodynamics of this drug can have a considerable effect on the personalized treatment of type 2 diabetes. In this study, we evaluated the association between rs11212617 polymorphism of ATM gene and rs628031 of SLC22A1 gene with response to treatment in newly diagnosed type 2 diabetes patients. We genotyped rs11212617 and rs628031 polymorphism by PCR based restriction fragment length polymorphism (RFLP) and assessed the role of this polymorphisms on response to treatment in 140 patients who have been recently diagnosed with type 2 diabetes and were under monotherapy with metformin for 6 months. Response to metformin was defined by HbA1c and fasting blood sugar (FBS) values. Based on such evaluations, patients were divided into two groups: responders (n= 63) and non-responders (n= 77). No significant association was found between these polymorphisms and response to treatment (OR= 0.86, [95% CI 0.52-1.41], P= 0.32) for rs11212617 and (OR= 0.45, [95% CI 0.64-1.76], P= 0.45) for rs 628031. The reported gene variants in ATM and SLC22A1 are not significantly associated with metformin treatment response in type 2 diabetic patients in an Iranian population.
Collapse
Affiliation(s)
- Fazlollah Shokri
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamid Ghaedi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolfazl Movafagh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Abediankenari
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Abdolkarim Mahrooz
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Zahra Kashi
- Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Corresponding author: Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. E-mail:
| |
Collapse
|
186
|
Meng W, Deshmukh HA, van Zuydam NR, Liu Y, Donnelly LA, Zhou K, Morris AD, Colhoun HM, Palmer CNA, Smith BH. A genome-wide association study suggests an association of Chr8p21.3 (GFRA2) with diabetic neuropathic pain. Eur J Pain 2015; 19:392-9. [PMID: 24974787 PMCID: PMC4737240 DOI: 10.1002/ejp.560] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2014] [Indexed: 12/19/2022]
Abstract
Background Neuropathic pain, caused by a lesion or a disease affecting the somatosensory system, is one of the most common complications in diabetic patients. The purpose of this study is to identify genetic factors contributing to this type of pain in a general diabetic population. Method We accessed the Genetics of Diabetes Audit and Research Tayside (GoDARTS) datasets that contain prescription information and monofilament test results for 9439 diabetic patients, among which 6927 diabetic individuals were genotyped by Affymetrix SNP6.0 or Illumina OmniExpress chips. Cases of neuropathic pain were defined as diabetic patients with a prescription history of at least one of five drugs specifically indicated for the treatment of neuropathic pain and in whom monofilament test result was positive for sensory neuropathy in at least one foot. Controls were individuals who did not have a record of receiving any opioid analgesics. Imputation of non‐genotyped SNPs was performed by IMPUTE2, with reference files from 1000 Genomes Phase I datasets. Results After data cleaning and relevant exclusions, imputed genotypes of 572 diabetic neuropathic pain cases and 2491 diabetic controls were used in the Fisher's exact test. We identified a cluster in the Chr8p21.3, next to GFRA2 with a lowest p‐value of 1.77 × 10−7 at rs17428041. The narrow‐sense heritability of this phenotype was 11.00%. Conclusion This genome‐wide association study on diabetic neuropathic pain suggests new evidence for the involvement of variants near GFRA2 with the disorder, which needs to be verified in an independent cohort and at the molecular level.
Collapse
Affiliation(s)
- W Meng
- Division of Population Health Sciences, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Abstract
OBJECTIVE The aim of this study was to determine the intrapair similarity in trough steady-state plasma concentrations of metformin in monozygotic and dizygotic twin pairs. METHODS We included 16 twin pairs (eight monozygotic and eight dizygotic twin pairs) for this study after contacting 524 twin pairs. They were dosed with metformin to steady state (1 g twice daily) for 6 days and on day 7, the trough concentration of metformin was determined 12 h after the last dose. RESULTS There was no strong intrapair similarity in trough steady-state plasma concentrations of metformin in either dizygotic or monozygotic twin pairs. CONCLUSION The trough steady-state plasma concentration of metformin does not appear to be tightly genetically regulated. The interpretation of this finding is limited by the small sample size.
Collapse
|
188
|
Abstract
p53 has been studied intensively as a major tumour suppressor that detects oncogenic events in cancer cells and eliminates them through senescence (a permanent non-proliferative state) or apoptosis. Consistent with this role, p53 activity is compromised in a high proportion of all cancer types, either through mutation of the TP53 gene (encoding p53) or changes in the status of p53 modulators. p53 has additional roles, which may overlap with its tumour-suppressive capacity, in processes including the DNA damage response, metabolism, aging, stem cell differentiation and fertility. Moreover, many mutant p53 proteins, termed 'gain-of-function' (GOF), acquire new activities that help drive cancer aggression. p53 is regulated mainly through protein turnover and operates within a negative-feedback loop with its transcriptional target, MDM2 (murine double minute 2), an E3 ubiquitin ligase which mediates the ubiquitylation and proteasomal degradation of p53. Induction of p53 is achieved largely through uncoupling the p53-MDM2 interaction, leading to elevated p53 levels. Various stress stimuli acting on p53 (such as hyperproliferation and DNA damage) use different, but overlapping, mechanisms to achieve this. Additionally, p53 activity is regulated through critical context-specific or fine-tuning events, mediated primarily through post-translational mechanisms, particularly multi-site phosphorylation and acetylation. In the present review, I broadly examine these events, highlighting their regulatory contributions, their ability to integrate signals from cellular events towards providing most appropriate response to stress conditions and their importance for tumour suppression. These are fascinating aspects of molecular oncology that hold the key to understanding the molecular pathology of cancer and the routes by which it may be tackled therapeutically.
Collapse
|
189
|
Zhang Y, Storr SJ, Johnson K, Green AR, Rakha EA, Ellis IO, Morgan DAL, Martin SG. Involvement of metformin and AMPK in the radioresponse and prognosis of luminal versus basal-like breast cancer treated with radiotherapy. Oncotarget 2015; 5:12936-49. [PMID: 25427448 PMCID: PMC4350336 DOI: 10.18632/oncotarget.2683] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/04/2014] [Indexed: 11/25/2022] Open
Abstract
Metformin is under evaluation as a potential anticancer agent. Expression of total and phospho(Thr172)-adenosine monophosphate-activated kinase-α (AMPKα and pAMPKα(Thr172) respectively), a main metformin target, was examined in radiotherapy treated breast cancers and metformin's ability to modulate Trx system expression and breast cancer radiosensitivity evaluated in vitro. AMPKα and pAMPKα(Thr172) expression was assessed using a discovery (n=166) and validation cohort (n=609). Metformin's role in regulating radioresponse, and Trx family expression, was examined via clonogenic assays and Western blots. Intracellular reactive oxygen species (ROS) levels, cell cycle progression and apoptosis were assessed by flow cytometry. High AMPKα expression associated with improved local recurrence-free (P=0.019), relapse-free (P=0.016) and breast cancer-specific survival (P=0.000065) and was, from multivariate analysis, an independent prognostic factor from the discovery cohort. From the validation cases AMPKα expression associated with relapse-free and breast cancer-specific survival in luminal breast cancers. Metformin substantially increased radiosensitivity, intracellular ROS levels and reduced Trx expression, in luminal breast cancer cells, but had little effect on basal phenotype cells. In conclusion, high AMPKα expression associates with improved prognosis, especially in luminal breast cancer. Metformin preferentially radiosensitises luminal breast cancer cells, potentially due to alterations to intracellular ROS levels via modulation of Trx family protein expression.
Collapse
Affiliation(s)
- Yimin Zhang
- Academic Unit of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - Sarah J Storr
- Academic Unit of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - Kerstie Johnson
- Clinical Oncology, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - Andrew R Green
- Histopathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - Emad A Rakha
- Histopathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - Ian O Ellis
- Histopathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - David A L Morgan
- Clinical Oncology, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| | - Stewart G Martin
- Academic Unit of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham University Hospitals NHS Trust, City Hospital Campus, Nottingham, UK
| |
Collapse
|
190
|
Reni M, Dugnani E, Cereda S, Belli C, Balzano G, Nicoletti R, Liberati D, Pasquale V, Scavini M, Maggiora P, Sordi V, Lampasona V, Ceraulo D, Di Terlizzi G, Doglioni C, Falconi M, Piemonti L. (Ir)relevance of Metformin Treatment in Patients with Metastatic Pancreatic Cancer: An Open-Label, Randomized Phase II Trial. Clin Cancer Res 2015; 22:1076-85. [PMID: 26459175 DOI: 10.1158/1078-0432.ccr-15-1722] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/04/2015] [Indexed: 12/16/2022]
Abstract
PURPOSE We aimed to assess the safety and efficacy of metformin for treating patients with metastatic pancreatic cancer and to identify endocrine and metabolic phenotypic features or tumor molecular markers associated with sensitivity to metformin antineoplastic action. EXPERIMENTAL DESIGN We designed an open-label, randomized, phase II trial to assess the efficacy of adding metformin to a standard systemic therapy with cisplatin, epirubicin, capecitabine, and gemcitabine (PEXG) in patients with metastatic pancreatic cancer. Patients ages 18 years or older with metastatic pancreatic cancer were randomly assigned (1:1) to receive PEXG every 4 weeks in combination or not with 2 g oral metformin daily. The primary endpoint was 6-months progression-free survival (PFS-6) in the intention-to-treat population. RESULTS Between August 2010 and January 2014, we randomly assigned 60 patients to receive PEXG with (n = 31) or without metformin (n = 29). At the preplanned interim analysis, the study was ended for futility. PFS-6 was 52% [95% confidence interval (CI), 33-69] in the control group and 42% (95% CI, 24-59) in the metformin group (P = 0.61). Furthermore, there was no difference in disease-free survival and overall survival between groups. Despite endocrine metabolic modifications induced by metformin, there was no correlation with the outcome. Single-nucleotide polymorphism rs11212617 predicted glycemic response, but not tumor response to metformin. Gene expression on tumor tissue did not predict tumor response to metformin. CONCLUSIONS Addition of metformin at the dose commonly used in diabetes did not improve outcome in patients with metastatic pancreatic cancer treated with standard systemic therapy. See related commentary by Yang and Rustgi, p. 1031.
Collapse
Affiliation(s)
- Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Erica Dugnani
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Cereda
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carmen Belli
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianpaolo Balzano
- Pancreatic Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Nicoletti
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Liberati
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Pasquale
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Maggiora
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Domenica Ceraulo
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gaetano Di Terlizzi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Doglioni
- Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy. Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy. Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
191
|
Pharmacogenetics and individual responses to treatment of hyperglycemia in type 2 diabetes. Pharmacogenet Genomics 2015; 25:475-84. [DOI: 10.1097/fpc.0000000000000160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
192
|
Shafaee A, Dastyar DZ, Islamian JP, Hatamian M. Inhibition of tumor energy pathways for targeted esophagus cancer therapy. Metabolism 2015; 64:1193-8. [PMID: 26271140 DOI: 10.1016/j.metabol.2015.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/18/2015] [Accepted: 07/13/2015] [Indexed: 11/18/2022]
Abstract
Interest in targeting cancer metabolism has been renewed in recent years with the discovery that many cancer related pathways have a profound effect on metabolism and that many tumors become dependent on specific metabolic processes. Accelerated glucose uptake during anaerobic glycolysis and loss of regulation between glycolytic metabolism and respiration, are the major metabolic changes found in malignant cells. The non-metabolizable glucose analog, 2-deoxy-D-glucose inhibits glucose synthesis and adenosine triphosphate production. The adenosine monophosphate-activated protein kinase (AMPK) is a key sensor of cellular energy and AMPK is a potential target for cancer prevention and/or treatment. Metformin is an activator of AMPK which inhibits protein synthesis and gluconeogenesis during cellular stress. This article reviews the status of clinical and laboratory researches exploring targeted therapies via metabolic pathways for treatment of esophageal cancer.
Collapse
Affiliation(s)
- Abbas Shafaee
- Department of Radiology, School of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Zarei Dastyar
- Department of Medical Radiation Science, School of Paramedicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalil Pirayesh Islamian
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Milad Hatamian
- Department of Medical Physics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
193
|
Xiao D, Zhang SM, Li X, Yin JY, Gong WJ, Zheng Y, Xu XJ, Lin X, Ji LN, Liu RR, Tang Q, Zhang W, Zhou HH, Han XY, Liu ZQ. IL-1B rs1143623 and EEF1A1P11-RPL7P9 rs10783050 polymorphisms affect the glucose-lowing efficacy of metformin in Chinese overweight or obese Type 2 diabetes mellitus patients. Pharmacogenomics 2015; 16:1621-9. [PMID: 26401715 DOI: 10.2217/pgs.15.95] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim: To investigate the potential genetic effect on metformin efficacy in overweight or obese Chinese Type 2 diabetes mellitus (T2DM) patients. Patients & methods: 768 SNPs in or close to 207 genes were genotyped in 84 patients treated with metformin + glibenclamide/Xiaoke Pill. Significant SNPs were then verified in 107 recent-onset overweight or obese T2DM patients treated with metformin alone. Genotyping was done by Illumina GoldenGate Assay. Results: In the discovery stage, 22 SNPs were nominally significant. IL1B rs1143623 (p = 0.011) and EEF1A1P11-RPL7P9 rs10783050 (p = 0.021) were still significantly associated with the relative change of HbA1c in the replication stage. Conclusion: IL1B rs1143623 and EEF1A1P11-RPL7P9 rs10783050 polymorphisms may contribute to metformin's glucose-lowing efficacy in overweight or obese Chinese T2DM patients.
Collapse
Affiliation(s)
- Di Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, People's Republic of China
| | - Si-Min Zhang
- Department of Endocrinology, The People's Hospital of Peking University, Beijing 100044, People's Republic of China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, People's Republic of China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, People's Republic of China
| | - Wei-Jing Gong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
| | - Yi Zheng
- The Maternal & Child Health Hospital of Hunan Province, Changsha 410008, People's Republic of China
| | - Xiao-Jing Xu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
| | - Xu Lin
- Institute of Nutrition Science of Shanghai School for Biological Sciences, Chinese Academy of Sciences, Shanghai 20031, People's Republic of China
| | - Li-Nong Ji
- Department of Endocrinology, The People's Hospital of Peking University, Beijing 100044, People's Republic of China
| | - Rang-Ru Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
| | - Qiang Tang
- Department of Pharmacy, The Huaihua Third People's Hospital, Huaihua 418000, People's Republic of China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, People's Republic of China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, People's Republic of China
| | - Xue-Yao Han
- Department of Endocrinology, The People's Hospital of Peking University, Beijing 100044, People's Republic of China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang 421001, People's Republic of China
| |
Collapse
|
194
|
Iyengar SK, Sedor JR, Freedman BI, Kao WHL, Kretzler M, Keller BJ, Abboud HE, Adler SG, Best LG, Bowden DW, Burlock A, Chen YDI, Cole SA, Comeau ME, Curtis JM, Divers J, Drechsler C, Duggirala R, Elston RC, Guo X, Huang H, Hoffmann MM, Howard BV, Ipp E, Kimmel PL, Klag MJ, Knowler WC, Kohn OF, Leak TS, Leehey DJ, Li M, Malhotra A, März W, Nair V, Nelson RG, Nicholas SB, O’Brien SJ, Pahl MV, Parekh RS, Pezzolesi MG, Rasooly RS, Rotimi CN, Rotter JI, Schelling JR, Seldin MF, Shah VO, Smiles AM, Smith MW, Taylor KD, Thameem F, Thornley-Brown DP, Truitt BJ, Wanner C, Weil EJ, Winkler CA, Zager PG, Igo RP, Hanson RL, Langefeld CD, Family Investigation of Nephropathy and Diabetes (FIND). Genome-Wide Association and Trans-ethnic Meta-Analysis for Advanced Diabetic Kidney Disease: Family Investigation of Nephropathy and Diabetes (FIND). PLoS Genet 2015; 11:e1005352. [PMID: 26305897 PMCID: PMC4549309 DOI: 10.1371/journal.pgen.1005352] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 06/10/2015] [Indexed: 11/28/2022] Open
Abstract
Diabetic kidney disease (DKD) is the most common etiology of chronic kidney disease (CKD) in the industrialized world and accounts for much of the excess mortality in patients with diabetes mellitus. Approximately 45% of U.S. patients with incident end-stage kidney disease (ESKD) have DKD. Independent of glycemic control, DKD aggregates in families and has higher incidence rates in African, Mexican, and American Indian ancestral groups relative to European populations. The Family Investigation of Nephropathy and Diabetes (FIND) performed a genome-wide association study (GWAS) contrasting 6,197 unrelated individuals with advanced DKD with healthy and diabetic individuals lacking nephropathy of European American, African American, Mexican American, or American Indian ancestry. A large-scale replication and trans-ethnic meta-analysis included 7,539 additional European American, African American and American Indian DKD cases and non-nephropathy controls. Within ethnic group meta-analysis of discovery GWAS and replication set results identified genome-wide significant evidence for association between DKD and rs12523822 on chromosome 6q25.2 in American Indians (P = 5.74x10-9). The strongest signal of association in the trans-ethnic meta-analysis was with a SNP in strong linkage disequilibrium with rs12523822 (rs955333; P = 1.31x10-8), with directionally consistent results across ethnic groups. These 6q25.2 SNPs are located between the SCAF8 and CNKSR3 genes, a region with DKD relevant changes in gene expression and an eQTL with IPCEF1, a gene co-translated with CNKSR3. Several other SNPs demonstrated suggestive evidence of association with DKD, within and across populations. These data identify a novel DKD susceptibility locus with consistent directions of effect across diverse ancestral groups and provide insight into the genetic architecture of DKD. Type 2 diabetes is the most common cause of severe kidney disease worldwide and diabetic kidney disease (DKD) associates with premature death. Individuals of non-European ancestry have the highest burden of type 2 DKD; hence understanding the causes of DKD remains critical to reducing health disparities. Family studies demonstrate that genes regulate the onset and progression of DKD; however, identifying these genes has proven to be challenging. The Family Investigation of Diabetes and Nephropathy consortium (FIND) recruited a large multi-ethnic collection of individuals with type 2 diabetes with and without kidney disease in order to detect genes associated with DKD. FIND discovered and replicated a DKD-associated genetic locus on human chromosome 6q25.2 (rs955333) between the SCAF8 and CNKSR genes. Findings were supported by significantly different expression of genes in this region from kidney tissue of subjects with, versus without DKD. The present findings identify a novel kidney disease susceptibility locus in individuals with type 2 diabetes which is consistent across subjects of differing ancestries. In addition, FIND results provide a rich catalogue of genetic variation in DKD patients for future research on the genetic architecture regulating this common and devastating disease.
Collapse
Affiliation(s)
- Sudha K. Iyengar
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail: (SKI); (JRS); (BIF)
| | - John R. Sedor
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Departments of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail: (SKI); (JRS); (BIF)
| | - Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail: (SKI); (JRS); (BIF)
| | - W. H. Linda Kao
- Department of Epidemiology and Medicine, John Hopkins University, Baltimore, Maryland, United States of America
| | - Matthias Kretzler
- Department of Internal Medicine/Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Benjamin J. Keller
- Department of Internal Medicine/Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hanna E. Abboud
- Department of Medicine/Nephrology, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Sharon G. Adler
- Department of Medicine, Division of Nephrology and Hypertension, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Lyle G. Best
- Missouri Breaks Industries Research, Timber Lake, South Dakota, United States of America
| | - Donald W. Bowden
- Department of Biochemistry, Center for Human Genomics, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Allison Burlock
- Department of Internal Medicine/Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Shelley A. Cole
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Mary E. Comeau
- Center for Public Health Genomics and Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, United States of America
| | - Jeffrey M. Curtis
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, United States of America
| | - Jasmin Divers
- Center for Public Health Genomics and Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, United States of America
| | - Christiane Drechsler
- University Hospital Würzburg, Renal Division and Comprehensive Heart Failure Center, Würzburg, Germany
| | - Ravi Duggirala
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Robert C. Elston
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Huateng Huang
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Barbara V. Howard
- MedStar Health Research Institute, Hyattsville, Maryland, United States of America
| | - Eli Ipp
- Department of Medicine, Section of Diabetes and Metabolism, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Paul L. Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, United States of America
| | - Michael J. Klag
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - William C. Knowler
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, United States of America
| | - Orly F. Kohn
- Department of Medicine, University of Chicago Medicine, Chicago, Illinois, United States of America
| | - Tennille S. Leak
- Department of Internal Medicine/Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - David J. Leehey
- Department of Medicine, Loyola School of Medicine, Maywood, Illinois, United States of America
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Alka Malhotra
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, United States of America
| | - Winfried März
- Heidelberg University and Synlab Academy, University of Graz, Graz, Austria
| | - Viji Nair
- Department of Internal Medicine/Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Robert G. Nelson
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, United States of America
| | - Susanne B. Nicholas
- Department of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Stephen J. O’Brien
- Theodosius Dobzhansky Center for Genome Bioinformatics, St. Petersburg, Russia, and Oceanographic Center, Nova Southeastern University, Ft. Lauderdale, Florida, United States of America
| | - Madeleine V. Pahl
- Department of Medicine, University of California, Irvine, Irvine, California, United States of America
| | - Rulan S. Parekh
- Departments of Paediatrics and Medicine, Hospital for Sick Children, University Health Network and the University of Toronto, Toronto, Ontario, Canada
| | - Marcus G. Pezzolesi
- Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rebekah S. Rasooly
- National Institute of Diabetes and Digestive Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Charles N. Rotimi
- Center for Research on Genomics and Global Health, Bethesda, Maryland, United States of America
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Jeffrey R. Schelling
- Departments of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Michael F. Seldin
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Davis, California, United States of America
| | - Vallabh O. Shah
- Department of Biochemistry & Molecular Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Adam M. Smiles
- Joslin Diabetes Center, Section on Genetics and Epidemiology, Boston, Massachusetts, United States of America
| | - Michael W. Smith
- National Human Genome Research Institute, Rockville, Maryland, United States of America
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Farook Thameem
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | | | - Barbara J. Truitt
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Christoph Wanner
- Department of Medicine, Division of Nephrology, University Hospital Würzburg, Würzburg, Germany
| | - E. Jennifer Weil
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, United States of America
| | - Cheryl A. Winkler
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Philip G. Zager
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Robert P. Igo
- Department of Epidemiology & Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Robert L. Hanson
- National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, United States of America
| | - Carl D. Langefeld
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | | |
Collapse
|
195
|
Meng W, Deshmukh HA, Donnelly LA, Torrance N, Colhoun HM, Palmer CNA, Smith BH. A Genome-wide Association Study Provides Evidence of Sex-specific Involvement of Chr1p35.1 (ZSCAN20-TLR12P) and Chr8p23.1 (HMGB1P46) With Diabetic Neuropathic Pain. EBioMedicine 2015; 2:1386-93. [PMID: 26629533 PMCID: PMC4634194 DOI: 10.1016/j.ebiom.2015.08.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/30/2015] [Accepted: 08/01/2015] [Indexed: 12/23/2022] Open
Abstract
Neuropathic pain is defined as pain arising as a direct consequence of a lesion or a disease affecting the somatosensory system and it affects around 1 in 4 diabetic patients in the UK. The purpose of this genome-wide association study (GWAS) was to identify genetic contributors to this disorder. Cases of neuropathic pain were defined as diabetic patients with a multiple prescription history of at least one of five drugs specifically indicated for the treatment of neuropathic pain. Controls were diabetic individuals who were not prescribed any of these drugs, nor amitriptyline, carbamazepine, or nortriptyline. Overall, 961 diabetic neuropathic pain cases and 3260 diabetic controls in the Genetics of Diabetes Audit and Research Tayside (GoDARTS) cohort were identified. We found a cluster in the Chr1p35.1 (ZSCAN20-TLR12P) with a lowest P value of 2.74 × 10(- 7) at rs71647933 in females and a cluster in the Chr8p23.1, next to HMGB1P46 with a lowest P value of 8.02 × 10(- 7) at rs6986153 in males. Sex-specific narrow sense heritability was higher in males (30.0%) than in females (14.7%). This GWAS on diabetic neuropathic pain provides evidence for the sex-specific involvement of Chr1p35.1 (ZSCAN20-TLR12P) and Chr8p23.1 (HMGB1P46) with the disorder, indicating the need for further research.
Collapse
Affiliation(s)
- Weihua Meng
- Division of Population Health Sciences, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| | - Harshal A Deshmukh
- Division of Population Health Sciences, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| | - Louise A Donnelly
- Centre for Pharmacogenetics and Pharmacogenomics, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | | | | | - Nicola Torrance
- Division of Population Health Sciences, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| | - Helen M Colhoun
- Division of Population Health Sciences, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| | - Colin N A Palmer
- Centre for Pharmacogenetics and Pharmacogenomics, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Blair H Smith
- Division of Population Health Sciences, Medical Research Institute, Ninewells Hospital and School of Medicine, University of Dundee, Dundee DD2 4BF, UK
| |
Collapse
|
196
|
Bhatt S, Gupta MK, Khamaisi M, Martinez R, Gritsenko MA, Wagner BK, Guye P, Busskamp V, Shirakawa J, Wu G, Liew CW, Clauss TR, Valdez I, El Ouaamari A, Dirice E, Takatani T, Keenan HA, Smith RD, Church G, Weiss R, Wagers AJ, Qian WJ, King GL, Kulkarni RN. Preserved DNA Damage Checkpoint Pathway Protects against Complications in Long-Standing Type 1 Diabetes. Cell Metab 2015; 22:239-52. [PMID: 26244933 PMCID: PMC4589213 DOI: 10.1016/j.cmet.2015.07.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/18/2015] [Accepted: 07/20/2015] [Indexed: 01/03/2023]
Abstract
The mechanisms underlying the development of complications in type 1 diabetes (T1D) are poorly understood. Disease modeling of induced pluripotent stem cells (iPSCs) from patients with longstanding T1D (disease duration ≥ 50 years) with severe (Medalist +C) or absent to mild complications (Medalist -C) revealed impaired growth, reprogramming, and differentiation in Medalist +C. Genomics and proteomics analyses suggested differential regulation of DNA damage checkpoint proteins favoring protection from cellular apoptosis in Medalist -C. In silico analyses showed altered expression patterns of DNA damage checkpoint factors among the Medalist groups to be targets of miR200, whose expression was significantly elevated in Medalist +C serum. Notably, neurons differentiated from Medalist +C iPSCs exhibited enhanced susceptibility to genotoxic stress that worsened upon miR200 overexpression. Furthermore, knockdown of miR200 in Medalist +C fibroblasts and iPSCs rescued checkpoint protein expression and reduced DNA damage. We propose miR200-regulated DNA damage checkpoint pathway as a potential therapeutic target for treating complications of diabetes.
Collapse
Affiliation(s)
- Shweta Bhatt
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Manoj K Gupta
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Mogher Khamaisi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rachael Martinez
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Bridget K Wagner
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Patrick Guye
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Volker Busskamp
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Shirakawa
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Gongxiong Wu
- Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Chong Wee Liew
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Therese R Clauss
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Ivan Valdez
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Abdelfattah El Ouaamari
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Ercument Dirice
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Tomozumi Takatani
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Hillary A Keenan
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - George Church
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Amy J Wagers
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Howard Hughes Medical Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - George L King
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
197
|
Zhou Y, Ye W, Wang Y, Jiang Z, Meng X, Xiao Q, Zhao Q, Yan J. Genetic variants of OCT1 influence glycemic response to metformin in Han Chinese patients with type-2 diabetes mellitus in Shanghai. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:9533-9542. [PMID: 26464716 PMCID: PMC4583948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 07/21/2015] [Indexed: 06/05/2023]
Abstract
AIMS/HYPOTHESIS Genetic variation in OCT1 can influence the glycemic response to metformin. We evaluated the effects of the OCT1 single-nucleotide polymorphisms (SNPs), rs1867351, rs4709400, rs628031, and rs2297374, on metformin efficacy in type-2 diabetes mellitus (DM) patients. METHODS We performed a single-center prospective analysis of the distributions of these SNPs in a cohort of Han Chinese subjects in Shanghai, China (HCS), and evaluated the effects of each SNP on glycemic control in HCS DM patients following 3 months of incident metformin treatment. RESULTS The allele frequencies of rs4709400 and rs628031 in our HCS control group differed from those previously reported for Han Chinese subjects in Beijing (HCB), as well as those previously reported for Caucasians and Africans, whereas the allele frequencies of rs1867351 and rs2297374 were more similar to those in HCB subjects. The DM patients with the rs1867351 T/T or rs4709400 G/G genotype exhibited greater reductions in postprandial plasma glucose (PPG), compared to those with different genotypes of these SNPs. The DM patients with the rs2297374 C/T, rs4709400 G/G, or rs628031 G/G genotype exhibited greater reductions in fasting plasma glucose (FPG), and those with the rs1867351 T/T, rs628031 A/A, or rs2297374 C/T genotype exhibited greater reductions in HbA1c , compared to those with different genotypes of these SNPs. Conclusions /interpretation: The rs1867351, rs4709400, rs628031, and rs2297374 SNPs of OCT1 have selective effects on FPG, PPG, and HbA1c in HCS DM patients in response to metformin treatment. Future studies of these SNPs in larger samples of HCS DM patients are warranted.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Weiwei Ye
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Yi Wang
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Zhikui Jiang
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Xiangying Meng
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Qian Xiao
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Qian Zhao
- Department of Endocrinology, Dahua Hospital Shanghai, China
| | - Jian Yan
- Department of Endocrinology, Dahua Hospital Shanghai, China
| |
Collapse
|
198
|
Abstract
The introduction of several new drug groups into the treatment of type 2 diabetes in the past few decades leads to an increased requirement for an individualized treatment approach. A personalized treatment is important from the point of view of both efficacy and safety. Recent guidelines are based mainly on entirely phenotypic characteristics such as diabetes duration, presence of macrovascular complications, or risk of hypoglycemia with the use of individual drugs. So far, genetic knowledge is used to guide treatment in the monogenic forms of diabetes. With the accumulating pharmacogenetic evidence in type 2 diabetes, there are reasonable expectations that genetics might help in the adjustment of drug doses to reduce severe side effects, as well as to make better therapeutic choices among the drugs available for the treatment of diabetes.
Collapse
Affiliation(s)
- Ivan Tkáč
- Department of Internal Medicine 4, P. J. Šafárik University, Faculty of Medicine, L. Pasteur University Hospital, Rastislavova 43, 041 90, Košice, Slovakia,
| |
Collapse
|
199
|
Dorajoo R, Liu J, Boehm BO. Genetics of Type 2 Diabetes and Clinical Utility. Genes (Basel) 2015; 6:372-84. [PMID: 26110315 PMCID: PMC4488669 DOI: 10.3390/genes6020372] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/02/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023] Open
Abstract
A large proportion of heritability of type 2 diabetes (T2D) has been attributed to inherent genetics. Recent genetic studies, especially genome-wide association studies (GWAS), have identified a multitude of variants associated with T2D. It is thus reasonable to question if these findings may be utilized in a clinical setting. Here we briefly review the identification of risk loci for T2D and discuss recent efforts and propose future work to utilize these loci in clinical setting-for the identification of individuals who are at particularly high risks of developing T2D and for the stratification of specific health-care approaches for those who would benefit most from such interventions.
Collapse
Affiliation(s)
- Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, 138672, Singapore.
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, 138672, Singapore.
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore.
| | - Bernhard O Boehm
- Genome Institute of Singapore, Agency for Science, Technology and Research, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore.
- Imperial College London, London, SW7 2AZ, UK.
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore.
| |
Collapse
|
200
|
Pollastro C, Ziviello C, Costa V, Ciccodicola A. Pharmacogenomics of Drug Response in Type 2 Diabetes: Toward the Definition of Tailored Therapies? PPAR Res 2015; 2015:415149. [PMID: 26161088 PMCID: PMC4486250 DOI: 10.1155/2015/415149] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/24/2015] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes is one of the major causes of mortality with rapidly increasing prevalence. Pharmacological treatment is the first recommended approach after failure in lifestyle changes. However, a significant number of patients shows-or develops along time and disease progression-drug resistance. In addition, not all type 2 diabetic patients have the same responsiveness to drug treatment. Despite the presence of nongenetic factors (hepatic, renal, and intestinal), most of such variability is due to genetic causes. Pharmacogenomics studies have described association between single nucleotide variations and drug resistance, even though there are still conflicting results. To date, the most reliable approach to investigate allelic variants is Next-Generation Sequencing that allows the simultaneous analysis, on a genome-wide scale, of nucleotide variants and gene expression. Here, we review the relationship between drug responsiveness and polymorphisms in genes involved in drug metabolism (CYP2C9) and insulin signaling (ABCC8, KCNJ11, and PPARG). We also highlight the advancements in sequencing technologies that to date enable researchers to perform comprehensive pharmacogenomics studies. The identification of allelic variants associated with drug resistance will constitute a solid basis to establish tailored therapeutic approaches in the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Carla Pollastro
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
- DiST, Department of Science and Technology, “Parthenope” University of Naples, Centro Direzionale, Isola C4, 80143 Naples, Italy
| | - Carmela Ziviello
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso”, National Research Council, Via Pietro Castellino 111, 80131 Naples, Italy
- DiST, Department of Science and Technology, “Parthenope” University of Naples, Centro Direzionale, Isola C4, 80143 Naples, Italy
| |
Collapse
|