51
|
Goodarzi MO, Palmer ND, Cui J, Guo X, Chen YDI, Taylor KD, Raffel LJ, Wagenknecht LE, Buchanan TA, Hsueh WA, Rotter JI. Classification of Type 2 Diabetes Genetic Variants and a Novel Genetic Risk Score Association With Insulin Clearance. J Clin Endocrinol Metab 2020; 105:dgz198. [PMID: 31714576 PMCID: PMC7059988 DOI: 10.1210/clinem/dgz198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Genome-wide association studies have identified more than 450 single nucleotide polymorphisms (SNPs) for type 2 diabetes (T2D). OBJECTIVE To facilitate use of these SNPs in future genetic risk score (GRS)-based analyses, we aimed to classify the SNPs based on physiology. We also sought to validate GRS associations with insulin-related traits in deeply phenotyped Mexican Americans. DESIGN, SETTING, AND PARTICIPANTS A total of 457 T2D SNPs from the literature were assigned physiologic function based on association studies and cluster analyses. All SNPs (All-GRS), beta-cell (BC-GRS), insulin resistance (IR-GRS), lipodystrophy (Lipo-GRS), and body mass index plus lipids (B + L-GRS) were evaluated for association with diabetes and indices of insulin secretion (from oral glucose tolerance test), insulin sensitivity and insulin clearance (from euglycemic clamp), and adiposity and lipid markers in 1587 Mexican Americans. RESULTS Of the 457 SNPs, 52 were classified as BC, 30 as IR, 12 as Lipo, 12 as B + L, whereas physiologic function of 351 was undefined. All-GRS was strongly associated with T2D. Among nondiabetic Mexican Americans, BC-GRS was associated with reduced insulinogenic index, IR-GRS was associated with reduced insulin sensitivity, and Lipo-GRS was associated with reduced adiposity. B + L-GRS was associated with increased insulin clearance. The latter did not replicate in an independent cohort wherein insulin clearance was assessed by a different method. CONCLUSIONS Supporting their utility, BC-GRS, IR-GRS, and Lipo-GRS, based on SNPs discovered largely in Europeans, exhibited expected associations in Mexican Americans. The novel association of B + L-GRS with insulin clearance suggests that impaired ability to reduce insulin clearance in compensation for IR may play a role in the pathogenesis of T2D. Whether this applies to other ethnic groups remains to be determined.
Collapse
Affiliation(s)
- Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, US
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, US
| | - Jinrui Cui
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, US
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, US
| | - Yii-Der I Chen
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, US
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, US
| | - Leslie J Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, US
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, US
| | - Thomas A Buchanan
- Department of Physiology and Biophysics and Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, US
| | - Willa A Hsueh
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Wexner Medical Center, The Ohio State University, Columbus, US
| | - Jerome I Rotter
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, US
| |
Collapse
|
52
|
Han X, Wei Y, Hu H, Wang J, Li Z, Wang F, Long T, Yuan J, Yao P, Wei S, Wang Y, Zhang X, Guo H, Yang H, Wu T, He M. Genetic Risk, a Healthy Lifestyle, and Type 2 Diabetes: the Dongfeng-Tongji Cohort Study. J Clin Endocrinol Metab 2020; 105:5696594. [PMID: 31900493 DOI: 10.1210/clinem/dgz325] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The objective of this study is to examine whether healthy lifestyle could reduce diabetes risk among individuals with different genetic profiles. DESIGN A prospective cohort study with a median follow-up of 4.6 years from the Dongfeng-Tongji cohort was performed. PARTICIPANTS A total of 19 005 individuals without diabetes at baseline participated in the study. MAIN VARIABLE MEASURE A healthy lifestyle was determined based on 6 factors: nonsmoker, nondrinker, healthy diet, body mass index of 18.5 to 23.9 kg/m2, waist circumference less than 85 cm for men and less than 80 cm for women, and higher level of physical activity. Associations of combined lifestyle factors and incident diabetes were estimated using Cox proportional hazard regression. A polygenic risk score of 88 single-nucleotide polymorphisms previously associated with diabetes was constructed to test for association with diabetes risk among 7344 individuals, using logistic regression. RESULTS A total of 1555 incident diabetes were ascertained. Per SD increment of simple and weighted genetic risk score was associated with a 1.39- and 1.34-fold higher diabetes risk, respectively. Compared with poor lifestyle, intermediate and ideal lifestyle were reduced to a 23% and 46% risk of incident diabetes, respectively. Association of lifestyle with diabetes risk was independent of genetic risk. Even among individuals with high genetic risk, intermediate and ideal lifestyle were separately associated with a 29% and 49% lower risk of diabetes. CONCLUSION Genetic and combined lifestyle factors were independently associated with diabetes risk. A healthy lifestyle could lower diabetes risk across different genetic risk categories, emphasizing the benefit of entire populations adhering to a healthy lifestyle.
Collapse
Affiliation(s)
- Xu Han
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yue Wei
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hua Hu
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jing Wang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhaoyang Li
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Fei Wang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Tengfei Long
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jing Yuan
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ping Yao
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Sheng Wei
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Youjie Wang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Huan Guo
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Handong Yang
- Dongfeng Central Hospital, Dongfeng Motor Corporation and Hubei University of Medicine, Shiyan, Hubei, P.R. China
| | - Tangchun Wu
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Meian He
- Department of Occupational and Environmental Health and Key Laboratory of Environmental and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
53
|
Zhang FF, Liu YH, Wang DW, Liu TS, Yang Y, Guo JM, Pan Y, Zhang YF, Du H, Li L, Jin L. Obesity-induced reduced expression of the lncRNA ROIT impairs insulin transcription by downregulation of Nkx6.1 methylation. Diabetologia 2020; 63:811-824. [PMID: 32008054 DOI: 10.1007/s00125-020-05090-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Although obesity is a predisposing factor for pancreatic beta cell dysfunction, the mechanisms underlying its negative effect on insulin-secreting cells is still poorly understood. The aim of this study was to identify islet long non-coding RNAs (lncRNAs) involved in obesity-mediated beta cell dysfunction. METHODS RNA sequencing was performed to analyse the islets of high-fat diet (HFD)-fed mice and those of normal chow-fed mice (NCD). The function in beta cells of the selected lncRNA 1810019D21Rik (referred to in this paper as ROIT [regulator of insulin transcription]) was assessed after its overexpression or knockdown in MIN6 cells and primary islet cells, as well as in siRNA-treated mice. Then, RNA pull-down, RNA immunoprecipitation, coimmunoprecipitation and bisulphite sequencing were performed to investigate the mechanism of ROIT regulation of islet function. RESULTS ROIT was dramatically downregulated in the islets of the obese mice, as well as in the sera of obese donors with type 2 diabetes, and was suppressed by HNF1B. Overexpression of ROIT in MIN6 cells and islets led to improved glucose homeostasis and insulin transcription. Investigation of the mechanism involved showed that ROIT bound to DNA methyltransferase 3a and caused its degradation through the ubiquitin proteasome pathway, which blocked the methylation of the Nkx6.1 promoter. CONCLUSIONS/INTERPRETATION These findings functionally suggest a novel link between obesity and beta cell dysfunction via ROIT. Elucidating a precise mechanism for the effect of obesity on lncRNA expression will broaden our understanding of the pathophysiological development of diabetes and facilitate the design of better tools for diabetes prevention and treatment. DATA AVAILABILITY The raw RNA sequencing data are available from the NCBI Gene Expression Omnibus (GEO series accession number GSE139991).
Collapse
Affiliation(s)
- Fang Fang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Yu Hong Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Dan Wei Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Ting Sheng Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Jia Min Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Yan Feng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China
| | - Hong Du
- Department of Endocrinology, Nanjing Jinling Hospital, 305 Zhongshan East Road, Nanjing, People's Republic of China
| | - Ling Li
- Department of Endocrinology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang Avenue, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
54
|
Kim H, Yoon BH, Oh CM, Lee J, Lee K, Song H, Kim E, Yi K, Kim MY, Kim H, Kim YK, Seo EH, Heo H, Kim HJ, Lee J, Suh JM, Koo SH, Seong JK, Kim S, Ju YS, Shong M, Kim M, Kim H. PRMT1 Is Required for the Maintenance of Mature β-Cell Identity. Diabetes 2020; 69:355-368. [PMID: 31848151 DOI: 10.2337/db19-0685] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 12/12/2019] [Indexed: 11/13/2022]
Abstract
Loss of functional β-cell mass is an essential feature of type 2 diabetes, and maintaining mature β-cell identity is important for preserving a functional β-cell mass. However, it is unclear how β-cells achieve and maintain their mature identity. Here we demonstrate a novel function of protein arginine methyltransferase 1 (PRMT1) in maintaining mature β-cell identity. Prmt1 knockout in fetal and adult β-cells induced diabetes, which was aggravated by high-fat diet-induced metabolic stress. Deletion of Prmt1 in adult β-cells resulted in the immediate loss of histone H4 arginine 3 asymmetric dimethylation (H4R3me2a) and the subsequent loss of β-cell identity. The expression levels of genes involved in mature β-cell function and identity were robustly downregulated as soon as Prmt1 deletion was induced in adult β-cells. Chromatin immunoprecipitation sequencing and assay for transposase-accessible chromatin sequencing analyses revealed that PRMT1-dependent H4R3me2a increases chromatin accessibility at the binding sites for CCCTC-binding factor (CTCF) and β-cell transcription factors. In addition, PRMT1-dependent open chromatin regions may show an association with the risk of diabetes in humans. Together, our results indicate that PRMT1 plays an essential role in maintaining β-cell identity by regulating chromatin accessibility.
Collapse
Affiliation(s)
- Hyunki Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Byoung-Ha Yoon
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Joonyub Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Kanghoon Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Heein Song
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eunha Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Kijong Yi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Mi-Young Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center, Seoul, Republic of Korea
| | - Hyeongseok Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Eun-Hye Seo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Haejeong Heo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Hee-Jin Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Junguee Lee
- Department of Pathology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, Republic of Korea
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Mirang Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
55
|
Cao M, Zhang L, Chen T, Shi A, Xie K, Li Z, Xu J, Chen Z, Ji C, Wen J. Genetic Susceptibility to Gestational Diabetes Mellitus in a Chinese Population. Front Endocrinol (Lausanne) 2020; 11:247. [PMID: 32390949 PMCID: PMC7188786 DOI: 10.3389/fendo.2020.00247] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 04/03/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction: New genetic variants associated with susceptibility to obesity and metabolic diseases have been discovered in recent genome-wide association (GWA) studies. The aim of this study was to investigate the association of theses risk variants with gestational diabetes mellitus (GDM). Methods: We performed a case-control study including 964 unrelated pregnant women with GDM and 1,021 pregnant women with normal glucose tolerance (as controls). A total of 33 genetic variants confirmed by GWA studies for obesity and metabolic diseases were selected and measured. Results: We observed that FTO rs1121980 and KCNQ1 rs163182 conferred a decreased GDM risk in the dominant and additive model [additive model: OR (95% CI) = 0.79 (0.67-0.94), P = 0.007 for rs1121980; OR(95%CI) = 0.84 (0.73-0.96), P = 0.009 for rs163182], whereas MC4R rs12970134 and PROX1 rs340841 conferred an increased GDM risk in the dominant, recessive, and additive model [additive model: OR(95%CI) = 1.25 (1.07-1.46), P = 0.006 for rs12970134; OR(95%CI) = 1.22 (1.07-1.39), P = 0.002 for rs340841). With the increasing number of risk alleles of the four significant SNPs, GDM risk was significantly increased in a dose-dependent manner (Ptrend < 0.001). And the significant positive associations between the weighted genetic risk score and risk of GDM persisted. Further function annotation indicated that these four SNPs may fall on the functional elements of human pancreatic islets. The genotype-phenotype associations indicated that these SNPs may contribute to GDM by affecting the expression levels of their nearby or distant genes. Conclusion: Our study suggests that FTO rs1121980, KCNQ1 rs163182, MC4R rs12970134, and PROX1 rs340841 may be markers for susceptibility to GDM in a Chinese population.
Collapse
Affiliation(s)
- Minkai Cao
- Department of Obstetrics, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Le Zhang
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Chen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Aiwu Shi
- Department of MICU, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Kaipeng Xie
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengying Li
- Department of Neonatology, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, China
| | - Jianjuan Xu
- Department of Obstetrics, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Zhong Chen
- Department of Obstetrics, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
- *Correspondence: Zhong Chen
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
- Chenbo Ji
| | - Juan Wen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
- Juan Wen
| |
Collapse
|
56
|
Afshardoost S, Sarhangi N, Afshari M, Aghaei Meybodi HR, Hasanzad M. The influence of a genetic variant in the KCNQ1 gene on type 2 diabetes mellitus development. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
57
|
Song C, Wang M, Fang H, Gong W, Mao D, Ding C, Fu Q, Feng G, Chen Z, Ma Y, Yao Y, Liu A. Effects of variants of 50 genes on diabetes risk among the Chinese population born in the early 1960s. J Diabetes 2019; 11:857-868. [PMID: 30907055 PMCID: PMC6850447 DOI: 10.1111/1753-0407.12922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/06/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Genome-wide association studies have identified loci that significantly increase diabetes risk. This study explored the genetic susceptibility in relation to diabetes risk in adulthood among a Chinese population born in the early 1960s. METHODS In all, 2129 subjects (833 males, 1296 females) were selected from the cross-sectional 2010 to 2012 China National Nutrition and Health Survey. Fifty diabetes-related single nucleotide polymorphisms (SNPs) were detected. Two diabetes genetic risk scores (GRSs) based on the 50 diabetes-predisposing variants were developed to examine the association of these SNPs with diabetes risk. RESULTS Associations were found between diabetes risk and SNPs in the MTNR1B (rs10830963), KLHDC5 (rs10842994), GRK5 (rs10886471), cyclindependentkinase 5 regulatory subunit associated protein 1 (rs10946398), adaptorrelated protein complex 3 subunit sigma 2 (rs2028299), diacylglycerol kinase beta/transmembrane protein 195 (rs2191349), SREBF chaperone (rs4858889), ankyrin1 (rs516946), RAS guanyl releasing protein 1 (rs7403531), and zinc finger AN1-type containing 3 (rs9470794) genes. As a continuous variable, with a 1-point increase in the GRS or weighted (w) GRS, fasting plasma glucose (FPG) increased 0.045 and 0.044 mM, respectively (P < 0.001 for both), after adjusting for confounders. Both GRS and wGRS showed an association with diabetes, with a multivariable-adjusted odds ratio (95% confidence interval) of 1.09 (1.00-1.19) and 1.12 (1.03-1.22), respectively, among all subjects. No significant associations were found between the GRS or wGRS and impaired fasting glucose or impaired glucose tolerance. CONCLUSIONS The data suggest the association of 10 SNPs and the GRS or wGRS with diabetes risk. Genetic susceptibility to diabetes may synergistically affect the risk of diabetes in adulthood.
Collapse
Affiliation(s)
- Chao Song
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Meng Wang
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Hongyun Fang
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Weiyan Gong
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Deqian Mao
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Caicui Ding
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Qiqi Fu
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Ganyu Feng
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Zheng Chen
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Yanning Ma
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Yecheng Yao
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| | - Ailing Liu
- Chinese Center for Disease Control and PreventionNational Institute for Nutrition and HealthBeijingChina
| |
Collapse
|
58
|
Cholesteryl Ester Transfer Protein Genetic Variants Associated with Risk for Type 2 Diabetes and Diabetic Kidney Disease in Taiwanese Population. Genes (Basel) 2019; 10:genes10100782. [PMID: 31597401 PMCID: PMC6826370 DOI: 10.3390/genes10100782] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/24/2019] [Accepted: 10/02/2019] [Indexed: 12/11/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) plays an important role in lipid metabolism. Low levels of high-density lipoprotein cholesterol (HDL-C) increase the risk of type 2 diabetes (T2D). This study investigated CETP gene variants to assess the risk of T2D and specific complications of diabetic kidney disease (DKD) and diabetic retinopathy. Towards this, a total of 3023 Taiwanese individuals (1383 without T2D, 1640 with T2D) were enrolled in this study. T2D mice (+Leprdb/+Leprdb, db/db) were used to determine CETP expression in tissues. The A-alleles of rs3764261, rs4783961, and rs1800775 variants were found to be independently associated with 2.86, 1.71, and 0.91 mg/dL increase in HDL-C per allele, respectively. In addition, the A-allele of rs4783961 was significantly associated with a reduced T2D risk (odds ratio (OR), 0.82; 95% confidence interval (CI), 0.71–0.96)), and the A-allele of rs1800775 was significantly related to a lowered DKD risk (OR, 0.78; 95% CI, 0.64–0.96). CETP expression was significantly decreased in the T2D mice kidney compared to that in the control mice (T2D mice, 0.16 ± 0.01 vs. control mice, 0.21 ± 0.02; p = 0.02). These collective findings indicate that CETP variants in the promoter region may affect HDL-C levels. Taiwanese individuals possessing an allele associated with higher HDL-C levels had a lower risk of T2D and DKD.
Collapse
|
59
|
Huang YC, Chen CC, Wang TY, Nguyen HTT, Chen YH, Wu CM, Chang YW, Liao WL, Tsai FJ. C-Reactive Protein Gene Variants and Their Serum Levels in Early Adult-onset Type 2 Diabetes Mellitus. In Vivo 2019; 33:1685-1690. [PMID: 31471424 DOI: 10.21873/invivo.11656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/09/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND/AIM C-Reactive protein (CRP) is a common marker of inflammation. Elevated CRP levels have been associated with increased risk of development of type 2 diabetes mellitus (T2DM). This study aimed to evaluate the association of CRP gene polymorphisms with early-onset T2DM and the effect of genetic variants on CRP level. MATERIALS AND METHODS In total, 948 individuals with early-onset (n=271) or late-onset (n=677) T2DM were enrolled in the study. Five single-nucleotide polymorphisms (SNPs) in the CRP gene, namely rs3093077, rs2808630, rs1800947, rs11265263, and rs11265265, were selected for genotyping, and CRP levels were measured. RESULTS Genotypic, allelic, and haplotype frequencies of these five SNPs were not significantly different between patients with early- and those with late-onset. T2DM Higher serum CRP levels were independently associated with the C-allele of rs3093077 and T-allele of rs11265265 (p<0.001). Furthermore, the C-allele of rs3093077 was associated with higher CRP level in both early- (p=0.016) and late-onset (p<0.001) T2DM. CONCLUSION CRP gene variants may contribute to the risk of early-onset T2DM by affecting the serum CRP level.
Collapse
Affiliation(s)
- Yu-Chuen Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan, R.O.C.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Ching-Chu Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan, R.O.C.,Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Tzu-Yuan Wang
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Hung Tran The Nguyen
- International Master's Program of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan, R.O.C
| | - Chia-Ming Wu
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Ya-Wen Chang
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan, R.O.C. .,Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Fuu-Jen Tsai
- Department of Medical Research, Medical Genetics and Pediatrics, China Medical University Hospital, Taichung, Taiwan, R.O.C. .,Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
60
|
Toraih EA, Abdelghany AA, Abd El Fadeal NM, Al Ageeli E, Fawzy MS. Deciphering the role of circulating lncRNAs: RNCR2, NEAT2, CDKN2B-AS1, and PVT1 and the possible prediction of anti-VEGF treatment outcomes in diabetic retinopathy patients. Graefes Arch Clin Exp Ophthalmol 2019; 257:1897-1913. [DOI: 10.1007/s00417-019-04409-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/17/2019] [Accepted: 06/29/2019] [Indexed: 12/13/2022] Open
|
61
|
Dai W, Lee D. Interfering with long chain noncoding RNA ANRIL expression reduces heart failure in rats with diabetes by inhibiting myocardial oxidative stress. J Cell Biochem 2019; 120:18446-18456. [PMID: 31211466 DOI: 10.1002/jcb.29162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/31/2022]
Abstract
This study is performed to elucidate whether long-chain noncoding RNA ANRIL has an effect on diabetes, and further explore the mechanism of ANRIL in diabetes. The rat model of diabetes was established via intraperitoneal injection of streptozotocin. The modeled rats were grouped into normal, diabetes, siRNA-NC, and ANRIL siRNA groups. Besides, the expression of ANRIL, cardiac function, inflammatory factor levels, cardiomyocyte apoptosis, and levels of oxidative stress index were all determined. Upregulated ANRIL was found in myocardial tissue of diabetic rats. Downregulated ANRIL improved cardiac function index and the expression of inflammatory factors, improved the pathological state of myocardial tissue and myocardial remodeling, decreased myocardial collagen deposition area and cardiomyocyte apoptosis and reduced the oxidative level of myocardial tissue in diabetic rats. This present study suggests that upregulated ANRIL is found in myocardial tissue of diabetic rats. Additionally, silencing of ANRIL reduces myocardial injury in diabetes by inhibiting myocardial oxidative stress.
Collapse
Affiliation(s)
- Wenxin Dai
- Department of Polymer_Nano Science and Technology, Polymer Fusion Research Center, Chonbuk National University, Jeonju, Korea.,Fourth Ward of Medical Care Center, Hainan Provincial People's Hospital, Haikou, Hainan, China
| | - Dongwon Lee
- Department of Polymer_Nano Science and Technology, Polymer Fusion Research Center, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
62
|
Systematic analysis of genes and diseases using PheWAS-Associated networks. Comput Biol Med 2019; 109:311-321. [PMID: 31128465 DOI: 10.1016/j.compbiomed.2019.04.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 02/08/2023]
|
63
|
Abu-Remaileh M, Abu-Remaileh M, Akkawi R, Knani I, Udi S, Pacold ME, Tam J, Aqeilan RI. WWOX somatic ablation in skeletal muscles alters glucose metabolism. Mol Metab 2019; 22:132-140. [PMID: 30755385 PMCID: PMC6437662 DOI: 10.1016/j.molmet.2019.01.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/18/2019] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE WWOX, a well-established tumor suppressor, is frequently lost in cancer and plays important roles in DNA damage response and cellular metabolism. METHODS We re-analyzed several genome-wide association studies (GWAS) using the Type 2 Diabetes Knowledge Portal website to uncover WWOX's association with metabolic syndrome (MetS). Using several engineered mouse models, we studied the effect of somatic WWOX loss on glucose homeostasis. RESULTS Several WWOX variants were found to be strongly associated with MetS disorders. In mouse models, somatic ablation of Wwox in skeletal muscle (WwoxΔSKM) results in weight gain, glucose intolerance, and insulin resistance. Furthermore, WwoxΔSKM mice display reduced amounts of slow-twitch fibers, decreased mitochondrial quantity and activity, and lower glucose oxidation levels. Mechanistically, we found that WWOX physically interacts with the cellular energy sensor AMP-activated protein kinase (AMPK) and that its loss is associated with impaired activation of AMPK, and with significant accumulation of the hypoxia inducible factor 1 alpha (HIF1α) in SKM. CONCLUSIONS Our studies uncover an unforeseen role of the tumor suppressor WWOX in whole-body glucose homeostasis and highlight the intimate relationship between cancer progression and metabolic disorders, particularly obesity and type-2 diabetes. SUBJECT AREAS Genetics, Metabolic Syndrome, Diabetes.
Collapse
Affiliation(s)
- Muhannad Abu-Remaileh
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Monther Abu-Remaileh
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Rania Akkawi
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ibrahim Knani
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Shiran Udi
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Micheal E Pacold
- Department of Radiation Oncology, New York University Langone Medical Center, 522 First Avenue, Smilow 907, New York, NY, USA
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Rami I Aqeilan
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel; Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
64
|
Mannino GC, Andreozzi F, Sesti G. Pharmacogenetics of type 2 diabetes mellitus, the route toward tailored medicine. Diabetes Metab Res Rev 2019; 35:e3109. [PMID: 30515958 PMCID: PMC6590177 DOI: 10.1002/dmrr.3109] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic disease that has reached the levels of a global epidemic. In order to achieve optimal glucose control, it is often necessary to rely on combination therapy of multiple drugs or insulin because uncontrolled glucose levels result in T2DM progression and enhanced risk of complications and mortality. Several antihyperglycemic agents have been developed over time, and T2DM pharmacotherapy should be prescribed based on suitability for the individual patient's characteristics. Pharmacogenetics is the branch of genetics that investigates how our genome influences individual responses to drugs, therapeutic outcomes, and incidence of adverse effects. In this review, we evaluated the pharmacogenetic evidences currently available in the literature, and we identified the top informative genetic variants associated with response to the most common anti-diabetic drugs: metformin, DPP-4 inhibitors/GLP1R agonists, thiazolidinediones, and sulfonylureas/meglitinides. Overall, we found 40 polymorphisms for each drug class in a total of 71 loci, and we examined the possibility of encouraging genetic screening of these variants/loci in order to critically implement decision-making about the therapeutic approach through precision medicine strategies. It is possible then to anticipate that when the clinical practice will take advantage of the genetic information of the diabetic patients, this will provide a useful resource for the prevention of T2DM progression, enabling the identification of the precise drug that is most likely to be effective and safe for each patient and the reduction of the economic impact on a global scale.
Collapse
Affiliation(s)
- Gaia Chiara Mannino
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Francesco Andreozzi
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| | - Giorgio Sesti
- Department of Medical and Surgical SciencesUniversity Magna Graecia of CatanzaroCatanzaroItaly
| |
Collapse
|
65
|
Tulay P, Temel S, Ergoren M. Investigation of KCNQ1 polymorphisms as biomarkers for cardiovascular diseases in the Turkish Cypriots for establishing preventative medical measures. Int J Biol Macromol 2019; 124:537-540. [DOI: 10.1016/j.ijbiomac.2018.11.227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/14/2018] [Accepted: 11/25/2018] [Indexed: 01/24/2023]
|
66
|
Liu Y, Wang T, Zhang M, Chen P, Yu Y. Down-regulation of myocardial infarction associated transcript 1 improves myocardial ischemia-reperfusion injury in aged diabetic rats by inhibition of activation of NF-κB signaling pathway. Chem Biol Interact 2019; 300:111-122. [PMID: 30611788 DOI: 10.1016/j.cbi.2019.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study is performed to investigate the effect of long chain noncoding RNA myocardial infarction associated transcript 1 (MIRT1) on myocardial ischemia reperfusion (I/R) injury in aged diabetic rats. METHODS The aged diabetic rat model and myocardial I/R injury model were established. Through injecting MIRT1 siRNA into caudal vein of rats, the cardiac function, myocardial pathological injury, myocardial fibrosis, cardiomyocytes apoptosis, oxidative stress and inflammatory injury of myocardial tissue of rats were measured. RESULTS For diabetic I/R rats, the expression of MIRT1 in myocardial tissue was increased, the activation of nuclear factor kappa B (NF-κB) signaling pathway was increased, the degree of damage to cardiac function was aggravated, the area of myocardial pathological injury and myocardial fibrosis was enlarged, the degree of cardiomyocytes apoptosis was increased, the degree of oxidative stress and inflammatory injury was increased. After inhibiting the expression of MIRT1, the activation of NF-κB signaling pathway was inhibited, the damage of cardiac function and cardiomyopathy was alleviated, the area of myocardial fibrosis was decreased, the degree of myocardial apoptosis was decreased, the degree of oxidative stress and inflammatory injury was obviously improved. CONCLUSION Our study highlights that down-regulation of MIRT1 improves myocardial I/R injury in aged diabetic rats by inhibition of activation of NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yaoxia Liu
- Department of Endocrinology, West China School of Medicine/ West China Hospital, Sichuan University, Chengdu, 610041, PR China; Department of Endocrinology in Elderly, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610041, PR China
| | - Tao Wang
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, PR China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, PR China; Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases of Sichuan Province, Chengdu, 610041, PR China
| | - Min Zhang
- Department of Endocrinology in Elderly, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610041, PR China
| | - Ping Chen
- Department of Endocrinology in Elderly, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610041, PR China
| | - Yerong Yu
- Department of Endocrinology, West China School of Medicine/ West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
67
|
Yang S, Chen X, Yang M, Zhao X, Chen Y, Zhao H, Liu C, Shen C. The variant at TGFBRAP1 is significantly associated with type 2 diabetes mellitus and affects diabetes-related miRNA expression. J Cell Mol Med 2019; 23:83-92. [PMID: 30461200 PMCID: PMC6307842 DOI: 10.1111/jcmm.13885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/06/2018] [Indexed: 01/09/2023] Open
Abstract
While the transforming growth factor-β1 (TGF-β1) regulates the growth and proliferation of pancreatic β-cells, its receptors trigger the activation of Smad network and subsequently induce the insulin resistance. A case-control was conducted to evaluate the associations of the polymorphisms of TGF-β1 receptor-associated protein 1 (TGFBRAP1) and TGF-β1 receptor 2 (TGFBR2) with type 2 diabetes mellitus (T2DM), and its genetic effects on diabetes-related miRNA expression. miRNA microarray chip was used to screen T2DM-related miRNA and 15 differential expressed miRNAs were further validated in 75 T2DM and 75 normal glucose tolerance (NGT). The variation of rs2241797 (T/C) at TGFBRAP1 showed significant association with T2DM in case-control study, and the OR (95% CI) of dominant model for cumulative effects was 1.204 (1.060-1.370), Bonferroni corrected P < 0.05. Significant differences in the fast glucose and HOMA-β indices were observed amongst the genotypes of rs2241797. The expression of has-miR-30b-5p and has-miR-93-5p was linearly increased across TT, TC, and CC genotypes of rs2241797 in NGT, Ptrend values were 0.024 and 0.016, respectively. Our findings suggest that genetic polymorphisms of TGFBRAP1 may contribute to the genetic susceptibility of T2DM by mediating diabetes-related miRNA expression.
Collapse
Affiliation(s)
- Song Yang
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu UniversityPeople's Hospital of Yixing CityYixingChina
| | - Xiaotian Chen
- Department of EpidemiologySchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Mengyao Yang
- Department of Clinical EpidemiologyGeriatric Hospital of Nanjing Medical UniversityJiangsu Province Geriatric InstituteNanjingChina
| | - Xianghai Zhao
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu UniversityPeople's Hospital of Yixing CityYixingChina
| | - Yanchun Chen
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu UniversityPeople's Hospital of Yixing CityYixingChina
| | - Hailong Zhao
- Division of Communicable Disease ControlHuai's Centre for Disease Control and PreventionHuaianChina
| | - Chunlan Liu
- Department of EpidemiologySchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Chong Shen
- Department of EpidemiologySchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Clinical EpidemiologyGeriatric Hospital of Nanjing Medical UniversityJiangsu Province Geriatric InstituteNanjingChina
| |
Collapse
|
68
|
Inhibition of Lincpint expression affects insulin secretion and apoptosis in mouse pancreatic β cells. Int J Biochem Cell Biol 2018; 104:171-179. [DOI: 10.1016/j.biocel.2018.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/02/2018] [Accepted: 09/06/2018] [Indexed: 12/27/2022]
|
69
|
Suwandhi L, Hausmann S, Braun A, Gruber T, Heinzmann SS, Gálvez EJC, Buck A, Legutko B, Israel A, Feuchtinger A, Haythorne E, Staiger H, Heni M, Häring HU, Schmitt-Kopplin P, Walch A, Cáceres CG, Tschöp MH, Rutter GA, Strowig T, Elsner M, Ussar S. Chronic d-serine supplementation impairs insulin secretion. Mol Metab 2018; 16:191-202. [PMID: 30093356 PMCID: PMC6157639 DOI: 10.1016/j.molmet.2018.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/07/2018] [Accepted: 07/16/2018] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE The metabolic role of d-serine, a non-proteinogenic NMDA receptor co-agonist, is poorly understood. Conversely, inhibition of pancreatic NMDA receptors as well as loss of the d-serine producing enzyme serine racemase have been shown to modulate insulin secretion. Thus, we aim to study the impact of chronic and acute d-serine supplementation on insulin secretion and other parameters of glucose homeostasis. METHODS We apply MALDI FT-ICR mass spectrometry imaging, NMR based metabolomics, 16s rRNA gene sequencing of gut microbiota in combination with a detailed physiological characterization to unravel the metabolic action of d-serine in mice acutely and chronically treated with 1% d-serine in drinking water in combination with either chow or high fat diet feeding. Moreover, we identify SNPs in SRR, the enzyme converting L-to d-serine and two subunits of the NMDA receptor to associate with insulin secretion in humans, based on the analysis of 2760 non-diabetic Caucasian individuals. RESULTS We show that chronic elevation of d-serine results in reduced high fat diet intake. In addition, d-serine leads to diet-independent hyperglycemia due to blunted insulin secretion from pancreatic beta cells. Inhibition of alpha 2-adrenergic receptors rapidly restores glycemia and glucose tolerance in d-serine supplemented mice. Moreover, we show that single nucleotide polymorphisms (SNPs) in SRR as well as in individual NMDAR subunits are associated with insulin secretion in humans. CONCLUSION Thus, we identify a novel role of d-serine in regulating systemic glucose metabolism through modulating insulin secretion.
Collapse
Affiliation(s)
- Lisa Suwandhi
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Simone Hausmann
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Alexander Braun
- Institute of Groundwater Ecology, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Tim Gruber
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Germany
| | - Silke S Heinzmann
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Research Unit Analytical BioGeoChemistry, Department of Environmental Sciences, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Eric J C Gálvez
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Achim Buck
- Research Unit Analytical Pathology, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Beata Legutko
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany
| | - Andreas Israel
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Elizabeth Haythorne
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, UK
| | - Harald Staiger
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Martin Heni
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany; Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard Karls University Tübingen, 72076 Tübingen, Germany; Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Philippe Schmitt-Kopplin
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Research Unit Analytical BioGeoChemistry, Department of Environmental Sciences, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Cristina García Cáceres
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany
| | - Matthias H Tschöp
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Germany
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, UK
| | - Till Strowig
- Research Group Microbial Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Elsner
- Institute of Groundwater Ecology, Helmholtz Center Munich, 85764 Neuherberg, Germany; Analytical Chemistry and Water Chemistry, Technical University of Munich, 81377 Munich, Germany
| | - Siegfried Ussar
- RG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748 Garching, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| |
Collapse
|
70
|
Genetic validation study of protein tyrosine phosphatase receptor type D (PTPRD) gene variants and risk for antipsychotic-induced weight gain. J Neural Transm (Vienna) 2018; 126:27-33. [DOI: 10.1007/s00702-018-1921-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/27/2018] [Indexed: 01/08/2023]
|
71
|
Liu Y, Wang C, Chen Y, Yuan Z, Yu T, Zhang W, Tang F, Gu J, Xu Q, Chi X, Ding L, Xue F, Zhang C. A variant in KCNQ1 gene predicts metabolic syndrome among northern urban Han Chinese women. BMC MEDICAL GENETICS 2018; 19:153. [PMID: 30157802 PMCID: PMC6114251 DOI: 10.1186/s12881-018-0652-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 07/23/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous studies have reported that the potassium voltage-gated channel subfamily Q member 1 (KCNQ1) gene is associated with diabetes in both European and Asian population. This study aims to find a predictable single nucleotide polymorphism (SNP) to predict the risk of metabolic syndrome (MetS) through investigating the association of SNP in KCNQ1 gene with MetS in Han Chinese women of northern urban area. METHODS Six SNPs were selected and genotyped in 1381 unrelated women aged 21 and above, who have had physical check-up in Shandong Provincial Qianfoshan Hospital. Cox proportional model was conducted to access the association between SNPs and MetS. RESULTS Sixty one women developed MetS between 2010 and 2015 during the 3055 person-year of follow-up. The cumulative incidence density was 19.964/1000 person-year. The SNP rs163182 was associated with MetS both in the additive genetic model (RR = 1.658, 95% CI: 1.144-2.402) and in the recessive genetic model (RR = 2.461, 95% CI: 1.347-4.496). It remained significant after adjustment. This relationship was also observed in MetS components (BMI and SBP). CONCLUSION A novel association between rs163182 and MetS was found in this study, which can predict the occurrence of MetS among northern urban Han Chinese women. More investigations are needed to be done to assess the possible pathway in which KCNQ1 gene affects MetS.
Collapse
Affiliation(s)
- Yafei Liu
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China.,Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Rd, Jinan, 250014, China
| | - Chunxia Wang
- Jinan Kingmed Center for Clinical Laboratory Co, Ltd., 554 Zhengfeng Rd, Jinan, 250010, Shandong, China
| | - Yafei Chen
- Linyi Centre for Adverse Drug Reaction Monitoring, Linyi, 276000, Shandong, China
| | - Zhongshang Yuan
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China
| | - Tao Yu
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China
| | - Wenchao Zhang
- Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Rd, Jinan, 250014, China
| | - Fang Tang
- Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Rd, Jinan, 250014, China
| | - Jianhua Gu
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China
| | - Qinqin Xu
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China
| | - Xiaotong Chi
- Department of Imaging and Nuclear Medicine, Taishan Medical University, 619 Changcheng Rd, Tai'an, 271016, Shandong, China
| | - Lijie Ding
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China
| | - Fuzhong Xue
- Division of Biostatistics, School of Public Health, Shandong University, 44 Wenhua Xilu, Jinan, 250010, Shandong, China.
| | - Chengqi Zhang
- Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Rd, Jinan, 250014, China.
| |
Collapse
|
72
|
Admixture mapping and fine-mapping of type 2 diabetes susceptibility loci in African American women. J Hum Genet 2018; 63:1109-1117. [PMID: 30135545 PMCID: PMC6202164 DOI: 10.1038/s10038-018-0503-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/18/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
African American women are disproportionately affected by type 2 diabetes. Genetic factors may explain part of the excess risk. More than 100 genetic variants have been associated with risk of type 2 diabetes, but most studies have been conducted in white populations. Two genome-wide association studies (GWAS) in African Americans have identified three novel genetic variants only. We conducted admixture mapping using 2,918 ancestral informative markers in 2,632 cases of type 2 diabetes and 2,596 controls nested in the ongoing Black Women’s Health Study cohort, with the goal of identifying genomic loci with local African ancestry associated with type 2 diabetes. In addition, we performed replication analysis of 71 previously identified index SNPs, and fine-mapped those genetic loci to identify better or new genetic variants associated with type 2 diabetes in African Americans. We found that individual African ancestry was associated with higher risk of type 2 diabetes. In addition, we identified two genomic regions, 3q26 and 12q23, with excess of African ancestry associated with higher risk of type 2 diabetes. Lastly, we replicated 8 out of 71 index SNPs from previous GWAS, including, for the first time in African Americans, the X-linked rs5945326 SNP near the DUSP9 gene. In addition, our fine-mapping efforts suggest independent signals at five loci. Our detailed analysis identified two genomic regions associated with risk of type 2 diabetes, and showed that many genetic risk variants are shared across ancestries.
Collapse
|
73
|
Morris AP. Progress in defining the genetic contribution to type 2 diabetes susceptibility. Curr Opin Genet Dev 2018; 50:41-51. [DOI: 10.1016/j.gde.2018.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/30/2022]
|
74
|
Hsueh WC, Bennett PH, Esparza-Romero J, Urquidez-Romero R, Valencia ME, Ravussin E, Williams RC, Knowler WC, Baier LJ, Schulz LO, Hanson RL. Analysis of type 2 diabetes and obesity genetic variants in Mexican Pima Indians: Marked allelic differentiation among Amerindians at HLA. Ann Hum Genet 2018; 82:287-299. [PMID: 29774533 DOI: 10.1111/ahg.12252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 02/11/2018] [Accepted: 03/08/2018] [Indexed: 01/21/2023]
Abstract
Prevalence of diabetes and obesity in Mexican Pima Indians is low, while prevalence in US Pima Indians is high. Although lifestyle likely accounts for much of the difference, the role of genetic factors is not well explored. To examine this, we genotyped 359 single nucleotide polymorphisms, including established type 2 diabetes and obesity variants from genome-wide association studies (GWAS) and 96 random markers, in 342 Mexican Pimas. A multimarker risk score of obesity variants was associated with body mass index (BMI; β = 0.81 kg/m2 per SD, P = 0.0066). The mean value of the score was lower in Mexican Pimas than in US Pimas (P = 4.3 × 10-11 ), and differences in allele frequencies at established loci could account for approximately 7% of the population difference in BMI; however, the difference in risk scores was consistent with evolutionary neutrality given genetic distance. To identify loci potentially under recent natural selection, allele frequencies at 283 variants were compared between US and Mexican Pimas, accounting for genetic distance. The largest differences were seen at HLA markers (e.g., rs9271720, difference = 0.75, P = 8.7 × 10-9 ); genetic distances at HLA were greater than at random markers (P = 1.6 × 10-46 ). Analyses of GWAS data in 937 US Pimas also showed sharing of alleles identical by descent at HLA that exceeds its genomic expectation (P = 7.0 × 10-10 ). These results suggest that, in addition to the widely recognized balancing selection at HLA, recent directional selection may also occur, resulting in marked allelic differentiation between closely related populations.
Collapse
Affiliation(s)
- Wen-Chi Hsueh
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Peter H Bennett
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Julian Esparza-Romero
- Departamento de Nutrición Pública y Salud, Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Sonora, México
| | - Rene Urquidez-Romero
- Instituto de Ciencias Biomédicas, Departamento de Ciencias de la Salud, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez, Chihuahua, México
| | - Mauro E Valencia
- Departamento de Nutrición Pública y Salud, Coordinación de Nutrición, Centro de Investigación en Alimentación y Desarrollo, Hermosillo, Sonora, México
| | - Eric Ravussin
- Pennington Biomedical Research Center, Louisiana State University Systems, Baton Rouge, LA, USA
| | - Robert C Williams
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - William C Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Leslie J Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Leslie O Schulz
- College of Health and Human Services, Northern Arizona University, Flagstaff, AZ, USA
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| |
Collapse
|
75
|
Pervjakova N, Kukushkina V, Haller T, Kasela S, Joensuu A, Kristiansson K, Annilo T, Perola M, Salomaa V, Jousilahti P, Metspalu A, Mägi R. Genome-wide analysis of nuclear magnetic resonance metabolites revealed parent-of-origin effect on triglycerides in medium very low-density lipoprotein in PTPRD gene. Biomark Med 2018. [DOI: 10.2217/bmm-2018-0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The aim of the study was to explore the parent-of-origin effects (POEs) on a range of human nuclear magnetic resonance metabolites. Materials & methods: We search for POEs in 14,815 unrelated individuals from Estonian and Finnish cohorts using POE method for the genotype data imputed with 1000 G reference panel and 82 nuclear magnetic resonance metabolites. Results: Meta-analysis revealed the evidence of POE for the variant rs1412727 in PTPRD gene for the metabolite: triglycerides in medium very low-density lipoprotein. No POEs were detected for genetic variants that were previously known to have main effect on circulating metabolites. Conclusion: We demonstrated possibility to detect POEs for human metabolites, but the POEs are weak, and therefore it is hard to detect those using currently available sample sizes.
Collapse
Affiliation(s)
- N Pervjakova
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular & Cell Biology, University of Tartu, Tartu 51010, Estonia
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, Hammersmith Hospital, UK
| | - V Kukushkina
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular & Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - T Haller
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
| | - S Kasela
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
| | - A Joensuu
- National Institute for Health & Welfare (THL), Department of Public Health Solutions, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - K Kristiansson
- National Institute for Health & Welfare (THL), Department of Public Health Solutions, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - T Annilo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular & Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - M Perola
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
- National Institute for Health & Welfare (THL), Department of Public Health Solutions, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | - V Salomaa
- National Institute for Health & Welfare (THL), Department of Public Health Solutions, Helsinki, Finland
| | - P Jousilahti
- National Institute for Health & Welfare (THL), Department of Public Health Solutions, Helsinki, Finland
| | - A Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular & Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - R Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
| |
Collapse
|
76
|
Vuckovic D, Mezzavilla M, Cocca M, Morgan A, Brumat M, Catamo E, Concas MP, Biino G, Franzè A, Ambrosetti U, Pirastu M, Gasparini P, Girotto G. Whole-genome sequencing reveals new insights into age-related hearing loss: cumulative effects, pleiotropy and the role of selection. Eur J Hum Genet 2018; 26:1167-1179. [PMID: 29725052 PMCID: PMC6057993 DOI: 10.1038/s41431-018-0126-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 02/05/2018] [Accepted: 02/13/2018] [Indexed: 01/17/2023] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory disorder in the elderly. Although not directly life threatening, it contributes to loss of autonomy and is associated with anxiety, depression and cognitive decline. To search for genetic risk factors underlying ARHL, a large whole-genome sequencing (WGS) approach has been carried out in a cohort of 212 cases and controls, both older than 50 years to select genes characterized by a burden of variants specific to cases or controls. Accordingly, the total variation load per gene was compared and two groups were detected: 375 genes more variable in cases and 371 more variable in controls. In both cases, Gene Ontology analysis showed that the largest enrichment for biological processes (fold > 5, p-value = 0.042) was the “sensory perception of sound”, suggesting cumulative genetic effects were involved. Replication confirmed 141 genes, while additional analysis based on natural selection led to a prioritization of 21 genes. The majority of them (20 out of 21) showed positive expression in mouse cochlea cDNA and were associated with two functional pathways. Among them, two genes were previously associated with hearing (CSMD1 and PTRPD) and re-sequenced in a large Italian cohort of ARHL patients (N = 389). Results led to the identification of six coding variants not detected in cases so far, suggesting a possible protective role, which requires investigation. In conclusion, we show that this multistep strategy (WGS, selection, expression, pathway analysis and targeted re-sequencing) can provide major insights into the molecular characterization of complex diseases such as ARHL.
Collapse
Affiliation(s)
- Dragana Vuckovic
- Medical Sciences, Chirurgical and Health Department, University of Trieste, Trieste, Italy. .,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy.
| | - Massimo Mezzavilla
- Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Massimiliano Cocca
- Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Anna Morgan
- Medical Sciences, Chirurgical and Health Department, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Marco Brumat
- Medical Sciences, Chirurgical and Health Department, University of Trieste, Trieste, Italy
| | - Eulalia Catamo
- Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Maria Pina Concas
- Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, Italy
| | - Annamaria Franzè
- Ceinge Advanced Biotechnology, Naples, Italy.,Neuroscience, Reproductive and Odontology Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Umberto Ambrosetti
- UO Audiology, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Milan, Italy.,Audiology Unit, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Paolo Gasparini
- Medical Sciences, Chirurgical and Health Department, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Giorgia Girotto
- Medical Sciences, Chirurgical and Health Department, University of Trieste, Trieste, Italy.,Medical Genetics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| |
Collapse
|
77
|
Chen T, Xu J, Liu G, Liu H, Chen M, Qin Y, Wu W, Xia Y, Ji C, Guo X, Wen J, Wang X. Genetic variants in PTPRD and risk of gestational diabetes mellitus. Oncotarget 2018; 7:76101-76107. [PMID: 27738328 PMCID: PMC5340179 DOI: 10.18632/oncotarget.12599] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/29/2016] [Indexed: 12/16/2022] Open
Abstract
Genome-wide association studies (GWASs) showed that two single nucleotide polymorphisms (SNPs) (rs17584499 and rs649891) in the protein tyrosine phosphatase receptor type D (PTPRD) were associated with type 2 diabetes (T2D). We sought to determine the influence of the PTPRD variants on the gestational diabetes mellitus (GDM) risk. In this research, two SNPs in PTPRD reported in T2D GWASs and six PTPRD expression-related SNPs were genotyped in 964 GDM cases and 1,021 controls using the Sequenom platform. Logistic regression analyses in additive models showed consistently significant associations of PTPRD rs10511544 A>C, rs10756026 T>A and rs10809070 C>G with a decreased risk of GDM [adjusted OR (95% CI) = 0.83 (0.72-0.97) for rs10511544; adjusted OR (95% CI) = 0.81 (0.70-0.94) for rs10756026; adjusted OR (95% CI) = 0.78 (0.65-0.92) for rs10809070]. Furthermore, the risk of GDM was significantly decreased with an increasing number of variant alleles of the three SNPs in a dose-dependent manner (Ptrend = 0.008). Moreover, the haplotype containing variant alleles of the three SNPs were significantly associated with a decreased risk of GDM [adjusted OR (95% CI) = 0.77 (0.64-0.92), P = 0.005], when compared with the most frequent haplotype. However, there were no significant associations for the SNPs reported in the T2D GWASs. Altogether, these findings indicate that the variants of rs10511544, rs10756026 and rs10809070 in PTPRD may contribute to a decreased susceptibility to GDM. Further validation in different ethnic backgrounds and biological function analyses are needed.
Collapse
Affiliation(s)
- Ting Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.,Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Juan Xu
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China.,Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Guangquan Liu
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Heng Liu
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yufeng Qin
- Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China.,Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Xirong Guo
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China.,Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Juan Wen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China.,Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
78
|
Khodakheir TN, Badpa F, Ranjbar N, Saravani R, Galavi HR. Combination and haplotype effects of protein tyrosine phosphatase receptor type D gene polymorphisms on type 2 diabetes in a sample of Iranian population: A pilot study. Meta Gene 2018. [DOI: 10.1016/j.mgene.2017.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
79
|
A common variant within the HNF1B gene is associated with overall survival of multiple myeloma patients: results from the IMMEnSE consortium and meta-analysis. Oncotarget 2018; 7:59029-59048. [PMID: 27437873 PMCID: PMC5312293 DOI: 10.18632/oncotarget.10665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/19/2016] [Indexed: 01/01/2023] Open
Abstract
Diabetogenic single nucleotide polymorphisms (SNPs) have recently been associated with multiple myeloma (MM) risk but their impact on overall survival (OS) of MM patients has not been analysed yet. In order to investigate the impact of 58 GWAS-identified variants for type 2 diabetes (T2D) on OS of patients with MM, we analysed genotyping data of 936 MM patients collected by the International Multiple Myeloma rESEarch (IMMENSE) consortium and an independent set of 700 MM patients recruited by the University Clinic of Heidelberg. A meta-analysis of the cox regression results of the two sets showed that rs7501939 located in the HNF1B gene negatively impacted OS (HRRec= 1.44, 95% CI = 1.18-1.76, P = 0.0001). The meta-analysis also showed a noteworthy gender-specific association of the SLC30A8rs13266634 SNP with OS. The presence of each additional copy of the minor allele at rs13266634 was associated with poor OS in men whereas no association was seen in women (HRMen-Add = 1.32, 95% CI 1.13-1.54, P = 0.0003). In conclusion, these data suggest that the HNF1Brs7501939 SNP confers poor OS in patients with MM and that a SNP in SLC30A8 affect OS in men.
Collapse
|
80
|
Gudmundsdottir V, Pedersen HK, Allebrandt KV, Brorsson C, van Leeuwen N, Banasik K, Mahajan A, Groves CJ, van de Bunt M, Dawed AY, Fritsche A, Staiger H, Simonis-Bik AMC, Deelen J, Kramer MHH, Dietrich A, Hübschle T, Willemsen G, Häring HU, de Geus EJC, Boomsma DI, Eekhoff EMW, Ferrer J, McCarthy MI, Pearson ER, Gupta R, Brunak S, 't Hart LM. Integrative network analysis highlights biological processes underlying GLP-1 stimulated insulin secretion: A DIRECT study. PLoS One 2018; 13:e0189886. [PMID: 29293525 PMCID: PMC5749727 DOI: 10.1371/journal.pone.0189886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 12/04/2017] [Indexed: 11/18/2022] Open
Abstract
Glucagon-like peptide 1 (GLP-1) stimulated insulin secretion has a considerable heritable component as estimated from twin studies, yet few genetic variants influencing this phenotype have been identified. We performed the first genome-wide association study (GWAS) of GLP-1 stimulated insulin secretion in non-diabetic individuals from the Netherlands Twin register (n = 126). This GWAS was enhanced using a tissue-specific protein-protein interaction network approach. We identified a beta-cell protein-protein interaction module that was significantly enriched for low gene scores based on the GWAS P-values and found support at the network level in an independent cohort from Tübingen, Germany (n = 100). Additionally, a polygenic risk score based on SNPs prioritized from the network was associated (P < 0.05) with glucose-stimulated insulin secretion phenotypes in up to 5,318 individuals in MAGIC cohorts. The network contains both known and novel genes in the context of insulin secretion and is enriched for members of the focal adhesion, extracellular-matrix receptor interaction, actin cytoskeleton regulation, Rap1 and PI3K-Akt signaling pathways. Adipose tissue is, like the beta-cell, one of the target tissues of GLP-1 and we thus hypothesized that similar networks might be functional in both tissues. In order to verify peripheral effects of GLP-1 stimulation, we compared the transcriptome profiling of ob/ob mice treated with liraglutide, a clinically used GLP-1 receptor agonist, versus baseline controls. Some of the upstream regulators of differentially expressed genes in the white adipose tissue of ob/ob mice were also detected in the human beta-cell network of genes associated with GLP-1 stimulated insulin secretion. The findings provide biological insight into the mechanisms through which the effects of GLP-1 may be modulated and highlight a potential role of the beta-cell expressed genes RYR2, GDI2, KIAA0232, COL4A1 and COL4A2 in GLP-1 stimulated insulin secretion.
Collapse
Affiliation(s)
- Valborg Gudmundsdottir
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Helle Krogh Pedersen
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Karla Viviani Allebrandt
- Department of Translational Bioinformatics, R&D Operations, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany
| | - Caroline Brorsson
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nienke van Leeuwen
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Disease Systems Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Wellcome Trust Center for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anubha Mahajan
- Oxford NIHR Biomedical Research Center, Oxford, United Kingdom
| | - Christopher J Groves
- Oxford Center for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Martijn van de Bunt
- Oxford NIHR Biomedical Research Center, Oxford, United Kingdom.,Oxford Center for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Adem Y Dawed
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Andreas Fritsche
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Member of the German Centre for Diabetes Research (DZD), Tübingen, Germany
| | - Harald Staiger
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University, Tübingen, Germany
| | - Annemarie M C Simonis-Bik
- Department of Internal Medicine, Diabetes Center and Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | - Joris Deelen
- Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.,Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Mark H H Kramer
- Department of Internal Medicine, Diabetes Center and Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | - Axel Dietrich
- Department of Translational Bioinformatics, R&D Operations, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany
| | - Thomas Hübschle
- Department GI Endocrinology, R&D Diabetes Division, Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Frankfurt, Germany
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit and the EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Member of the German Centre for Diabetes Research (DZD), Tübingen, Germany
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit and the EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands.,Netherlands Consortium for Healthy Aging, Leiden, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit and the EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Elisabeth M W Eekhoff
- Department of Internal Medicine, Diabetes Center and Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jorge Ferrer
- Section of Epigenomics and Disease, Department of Medicine, Imperial College London, London, United Kingdom.,Genomic Programming of Beta Cells Laboratory, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mark I McCarthy
- Wellcome Trust Center for Human Genetics, University of Oxford, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Center, Oxford, United Kingdom.,Oxford Center for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom
| | - Ewan R Pearson
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ramneek Gupta
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Søren Brunak
- Department of Bio and Health Informatics, Technical University of Denmark, Kongens Lyngby, Denmark.,Novo Nordisk Foundation Center for Protein Research, Disease Systems Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leen M 't Hart
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands.,Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
81
|
Hu C, Jia W. Diabetes in China: Epidemiology and Genetic Risk Factors and Their Clinical Utility in Personalized Medication. Diabetes 2018; 67:3-11. [PMID: 29263166 DOI: 10.2337/dbi17-0013] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 12/15/2022]
Abstract
The incidence of type 2 diabetes (T2D) has rapidly increased over recent decades, and T2D has become a leading public health challenge in China. Compared with European descents, Chinese patients with T2D are diagnosed at a relatively young age and low BMI. A better understanding of the factors contributing to the diabetes epidemic is crucial for determining future prevention and intervention programs. In addition to environmental factors, genetic factors contribute substantially to the development of T2D. To date, more than 100 susceptibility loci for T2D have been identified. Individually, most T2D genetic variants have a small effect size (10-20% increased risk for T2D per risk allele); however, a genetic risk score that combines multiple T2D loci could be used to predict the risk of T2D and to identify individuals who are at a high risk. Furthermore, individualized antidiabetes treatment should be a top priority to prevent complications and mortality. In this article, we review the epidemiological trends and recent progress in the understanding of T2D genetic etiology and further discuss personalized medicine involved in the treatment of T2D.
Collapse
Affiliation(s)
- Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
- Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, People's Republic of China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| |
Collapse
|
82
|
Girard H, Potvin O, Nugent S, Dallaire-Théroux C, Cunnane S, Duchesne S. Faster progression from MCI to probable AD for carriers of a single-nucleotide polymorphism associated with type 2 diabetes. Neurobiol Aging 2017; 64:157.e11-157.e17. [PMID: 29338921 DOI: 10.1016/j.neurobiolaging.2017.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/10/2017] [Accepted: 11/29/2017] [Indexed: 11/27/2022]
Abstract
Sporadic Alzheimer's disease (AD), as opposed to its autosomal dominant form, is likely caused by a complex interaction of genetic, environmental, and health lifestyle factors. Twin studies indicate that sporadic AD heritability could be between 58% and 79%, around half of which is explained by the ε4 allele of the apolipoprotein E (APOE4). We hypothesized that genes associated with known risk factors for AD, namely hypertension, hypercholesterolemia, obesity, diabetes, and cardiovascular disease, would contribute significantly to the remaining heritability. We analyzed 22 AD-associated single-nucleotide polymorphisms (SNPs), associated with these risk factors, that were included in the sequencing data of the Alzheimer's Disease Neuroimaging Initiative 1 data set, which included 355 participants with mild cognitive impairment (MCI). We built survival models with the selected SNPs to predict progression of MCI to probable AD over the 10-year follow-up of the study. The rs391300 SNP, located on the serine racemase (SRR) gene and linked to increased susceptibility to type 2 diabetes, was associated with progression from MCI to probable AD.
Collapse
Affiliation(s)
- Hugo Girard
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, Canada; Département de radiologie, Faculté de médecine, Université Laval, Québec, Canada
| | - Olivier Potvin
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Scott Nugent
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, Canada
| | - Caroline Dallaire-Théroux
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, Canada; Faculté de médecine, Université Laval, Québec, Canada
| | - Stephen Cunnane
- Département de médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Canada
| | - Simon Duchesne
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, Canada; Département de radiologie, Faculté de médecine, Université Laval, Québec, Canada.
| | | |
Collapse
|
83
|
Siddiqui A, Hussain S, Azam A, Muslim I, Maqsood SI, Ahmed W, Murad M, Niazi MK, Ishaq M, Waheed NK, Qamar R, Azam M. ANRIL polymorphism rs1333049, a novel genetic predictor for diabetic retinopathy complication. Meta Gene 2017. [DOI: 10.1016/j.mgene.2017.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
84
|
Sobrin L, Chong YH, Fan Q, Gan A, Stanwyck LK, Kaidonis G, Craig JE, Kim J, Liao WL, Huang YC, Lee WJ, Hung YJ, Guo X, Hai Y, Ipp E, Pollack S, Hancock H, Price A, Penman A, Mitchell P, Liew G, Smith AV, Gudnason V, Tan G, Klein BEK, Kuo J, Li X, Christiansen MW, Psaty BM, Sandow K, Jensen RA, Klein R, Cotch MF, Wang JJ, Jia Y, Chen CJ, Chen YDI, Rotter JI, Tsai FJ, Hanis CL, Burdon KP, Wong TY, Cheng CY. Genetically Determined Plasma Lipid Levels and Risk of Diabetic Retinopathy: A Mendelian Randomization Study. Diabetes 2017; 66:3130-3141. [PMID: 28951389 PMCID: PMC5697951 DOI: 10.2337/db17-0398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/22/2017] [Indexed: 11/17/2022]
Abstract
Results from observational studies examining dyslipidemia as a risk factor for diabetic retinopathy (DR) have been inconsistent. We evaluated the causal relationship between plasma lipids and DR using a Mendelian randomization approach. We pooled genome-wide association studies summary statistics from 18 studies for two DR phenotypes: any DR (N = 2,969 case and 4,096 control subjects) and severe DR (N = 1,277 case and 3,980 control subjects). Previously identified lipid-associated single nucleotide polymorphisms served as instrumental variables. Meta-analysis to combine the Mendelian randomization estimates from different cohorts was conducted. There was no statistically significant change in odds ratios of having any DR or severe DR for any of the lipid fractions in the primary analysis that used single nucleotide polymorphisms that did not have a pleiotropic effect on another lipid fraction. Similarly, there was no significant association in the Caucasian and Chinese subgroup analyses. This study did not show evidence of a causal role of the four lipid fractions on DR. However, the study had limited power to detect odds ratios less than 1.23 per SD in genetically induced increase in plasma lipid levels, thus we cannot exclude that causal relationships with more modest effect sizes exist.
Collapse
Affiliation(s)
- Lucia Sobrin
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA
| | - Yong He Chong
- Duke-NUS Medical School, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Qiao Fan
- Duke-NUS Medical School, National University of Singapore, Singapore
| | - Alfred Gan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Lynn K Stanwyck
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear, Boston, MA
| | - Georgia Kaidonis
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Jihye Kim
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, TX
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Chuen Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Jen Hung
- Department of Internal Medicine, Tri-Service General Hospital, Taipei City, Taiwan
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Yang Hai
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Eli Ipp
- Department of Medicine, LA BioMed, Harbor-UCLA Medical Center, Torrance, CA
| | - Samuela Pollack
- Department of Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Heather Hancock
- Department of Ophthalmology, The University of Mississippi Medical Center, Jackson, MS
| | - Alkes Price
- Department of Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Alan Penman
- Department of Medicine, The University of Mississippi Medical Center, Jackson, MS
| | - Paul Mitchell
- Centre for Vision Research, The Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Gerald Liew
- Centre for Vision Research, The Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Albert V Smith
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Icelandic Heart Association, Kópavogur, Iceland
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Icelandic Heart Association, Kópavogur, Iceland
| | - Gavin Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Barbara E K Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI
| | - Jane Kuo
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
- Clinical and Medical Affairs, CardioDx, Inc., Redwood City, CA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Mark W Christiansen
- Cardiovascular Health Research Unit, Division of General Internal Medicine, University of Washington, Seattle, WA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Division of General Internal Medicine, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | - Kevin Sandow
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Richard A Jensen
- Cardiovascular Health Research Unit, Division of General Internal Medicine, University of Washington, Seattle, WA
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI
| | - Mary Frances Cotch
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Jie Jin Wang
- Duke-NUS Medical School, National University of Singapore, Singapore
- Centre for Vision Research, The Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Yucheng Jia
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Ching J Chen
- Department of Ophthalmology, The University of Mississippi Medical Center, Jackson, MS
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, LA BioMed, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Departments of Medical Genetics, Pediatrics, and Medical Research, China Medical University Hospital, Tiachung, Tiawan
| | - Craig L Hanis
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, TX
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Tien Yin Wong
- Duke-NUS Medical School, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ching-Yu Cheng
- Duke-NUS Medical School, National University of Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
85
|
Ye L, Yang C, Dou JF, Wen LL, Wang WJ, Zheng XD, Zuo XB, Zhou FS, Fan X, Zhang XJ. Genetic factors are stressed variably by onset age-based sample selection in psoriasis: A hint from major histocompatibility complex region-based analysis. J Gene Med 2017; 19. [PMID: 29076194 DOI: 10.1002/jgm.2997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 09/02/2017] [Accepted: 10/13/2017] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Large cohort-based genetic association studies have been established over a decade. However, for certain diseases, different results with respect to the genome-wide association study level have been obtained among studies, even for those conducted within the same ethnic groups. We hypothesized that onset age-based sample variables might have a great impact on the results. METHODS In the present study, we divided psoriasis patients into several subgroups according to the onset age bracket. We conducted genetic association analysis in the major histocompatibility complex (MHC) region of each patient subgroup with shared control subjects. RESULTS We found decreases in the numbers of susceptible variants in each subgroup analysis as the onset age increased in the longitudinal analysis. Meanwhile, the pairwise analysis showed that younger patients exhibited greater numbers of genetic risks in the MHC region compared to elder patients, regardless of whether the cut-off values were defined as 20 or 30 years old. Similar results were also found among 11-20-, 21-30- and 31-40-year-old groups. Furthermore, when combining the results of both the stepwise regression analysis and the HLA-C*06:02 conditioning analysis, different variants were found to be independently associated with each psoriasis subgroup. CONCLUSIONS Onset age-based sample variables influence the results of genetic association studies, at least in MHC region-based genetic analysis. We suggest that caution is required when selecting samples for genetic association studies to prevent confounders that might be a result of onset age.
Collapse
Affiliation(s)
- Lei Ye
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Chao Yang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Jin-Fa Dou
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Lei-Lei Wen
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Wen-Jun Wang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Xiao-Dong Zheng
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Xian-Bo Zuo
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Fu-Sheng Zhou
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Xing Fan
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| | - Xue-Jun Zhang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, PR China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, Anhui, PR China
| |
Collapse
|
86
|
Szabo M, Máté B, Csép K, Benedek T. Genetic Approaches to the Study of Gene Variants and Their Impact on the Pathophysiology of Type 2 Diabetes. Biochem Genet 2017; 56:22-55. [DOI: 10.1007/s10528-017-9827-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022]
|
87
|
Liu Y, Zhao J, Jiang T, Yu M, Jiang G, Hu Y. A pathway analysis of genome-wide association study highlights novel type 2 diabetes risk pathways. Sci Rep 2017; 7:12546. [PMID: 28970525 PMCID: PMC5624908 DOI: 10.1038/s41598-017-12873-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/11/2017] [Indexed: 02/04/2023] Open
Abstract
Genome-wide association studies (GWAS) have been widely used to identify common type 2 diabetes (T2D) variants. However, the known variants just explain less than 20% of the overall estimated genetic contribution to T2D. Pathway-based methods have been applied into T2D GWAS datasets to investigate the biological mechanisms and reported some novel T2D risk pathways. However, few pathways were shared in these studies. Here, we performed a pathway analysis using the summary results from a large-scale meta-analysis of T2D GWAS to investigate more genetic signals in T2D. Here, we selected PLNK and VEGAS to perform the gene-based test and WebGestalt to perform the pathway-based test. We identified 8 shared KEGG pathways after correction for multiple tests in both methods. We confirm previous findings, and highlight some new T2D risk pathways. We believe that our results may be helpful to study the genetic mechanisms of T2D.
Collapse
Affiliation(s)
- Yang Liu
- College of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jing Zhao
- The Department of Obstetrics and Gynaecology, Heilongjiang Provincial Forestry General Hospital, Harbin, Heilongjiang, China
| | - Tao Jiang
- The 224th Hospital of Chinese People's Liberation Army, Harbin, Heilongjiang, China
| | - Mei Yu
- Research institute of Chinese Medicine in Heilongjiang province, Harbin, Heilongjiang, China
| | - Guohua Jiang
- College of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yang Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
88
|
Liao WL, Lee WJ, Chen CC, Lu CH, Chen CH, Chou YC, Lee IT, Sheu WHH, Wu JY, Yang CF, Wang CH, Tsai FJ. Pharmacogenetics of dipeptidyl peptidase 4 inhibitors in a Taiwanese population with type 2 diabetes. Oncotarget 2017; 8:18050-18058. [PMID: 28160554 PMCID: PMC5392306 DOI: 10.18632/oncotarget.14951] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors are oral anti-hyperglycemic drugs enabling effective glycemic control in type 2 diabetes (T2D). Despite DPP-4 inhibitors' advantages, the patients' therapeutic response varies. In this retrospective cohort study, 171 Taiwanese patients with T2D were classified as sensitive or resistant to treatment based on the mean change in HbA1c levels. Using an assumption-free genome-wide association study, 45 single nucleotide polymorphisms (SNPs) involved in the therapeutic response to DPP-4 inhibitors (P < 1 × 10-4) were identified at or near PRKD1, CNTN3, ASK, and LOC10537792. A SNP located within the fourth intron of PRKD1 (rs57803087) was strongly associated with DPP-4 inhibitor response (P = 3.2 × 10-6). This is the first pharmacogenomics study on DPP-4 inhibitor treatment for diabetes in a Taiwanese population. Our data suggest that genes associated with β-cell function and apoptosis are involved in the therapeutic effect of DPP-4 inhibitors, even in the presence of additional oral anti-diabetic drugs. Our findings provide information on how genetic variants influence drug response and may benefit the development of personalized medicine.
Collapse
Affiliation(s)
- Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.,Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan.,School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chieh Hsiang Lu
- Department of Internal Medicine, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi City, Taiwan.,Department of Nursing, College of Nursing and Health Sciences, DAYEH University, Taiwan.,Department of Business management, College of Management, National Chung Cheng University, Taiwan
| | - Chien-Hsiun Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Chun Chou
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans Hospital, Taichung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Defense Medical Center, Taipei, Taiwan.,Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
| | - Jer-Yuarn Wu
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Fan Yang
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chung-Hsing Wang
- Department of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung, Taiwan.,School of Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
89
|
Wood AR, Jonsson A, Jackson AU, Wang N, van Leewen N, Palmer ND, Kobes S, Deelen J, Boquete-Vilarino L, Paananen J, Stančáková A, Boomsma DI, de Geus EJC, Eekhoff EMW, Fritsche A, Kramer M, Nijpels G, Simonis-Bik A, van Haeften TW, Mahajan A, Boehnke M, Bergman RN, Tuomilehto J, Collins FS, Mohlke KL, Banasik K, Groves CJ, McCarthy MI, Pearson ER, Natali A, Mari A, Buchanan TA, Taylor KD, Xiang AH, Gjesing AP, Grarup N, Eiberg H, Pedersen O, Chen YD, Laakso M, Norris JM, Smith U, Wagenknecht LE, Baier L, Bowden DW, Hansen T, Walker M, Watanabe RM, 't Hart LM, Hanson RL, Frayling TM. A Genome-Wide Association Study of IVGTT-Based Measures of First-Phase Insulin Secretion Refines the Underlying Physiology of Type 2 Diabetes Variants. Diabetes 2017; 66:2296-2309. [PMID: 28490609 PMCID: PMC5521867 DOI: 10.2337/db16-1452] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 05/02/2017] [Indexed: 01/19/2023]
Abstract
Understanding the physiological mechanisms by which common variants predispose to type 2 diabetes requires large studies with detailed measures of insulin secretion and sensitivity. Here we performed the largest genome-wide association study of first-phase insulin secretion, as measured by intravenous glucose tolerance tests, using up to 5,567 individuals without diabetes from 10 studies. We aimed to refine the mechanisms of 178 known associations between common variants and glycemic traits and identify new loci. Thirty type 2 diabetes or fasting glucose-raising alleles were associated with a measure of first-phase insulin secretion at P < 0.05 and provided new evidence, or the strongest evidence yet, that insulin secretion, intrinsic to the islet cells, is a key mechanism underlying the associations at the HNF1A, IGF2BP2, KCNQ1, HNF1B, VPS13C/C2CD4A, FAF1, PTPRD, AP3S2, KCNK16, MAEA, LPP, WFS1, and TMPRSS6 loci. The fasting glucose-raising allele near PDX1, a known key insulin transcription factor, was strongly associated with lower first-phase insulin secretion but has no evidence for an effect on type 2 diabetes risk. The diabetes risk allele at TCF7L2 was associated with a stronger effect on peak insulin response than on C-peptide-based insulin secretion rate, suggesting a possible additional role in hepatic insulin clearance or insulin processing. In summary, our study provides further insight into the mechanisms by which common genetic variation influences type 2 diabetes risk and glycemic traits.
Collapse
Affiliation(s)
- Andrew R Wood
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Exeter, U.K
| | - Anna Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| | - Nan Wang
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Diabetes & Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Nienke van Leewen
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ
| | - Joris Deelen
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Lorena Boquete-Vilarino
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Exeter, U.K
| | - Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Alena Stančáková
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, the Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, VU University, Amsterdam, the Netherlands
| | - Elisabeth M W Eekhoff
- Diabetes Center, Internal Medicine Unit, VU University Medical Center, Amsterdam, the Netherlands
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), Tübingen, Germany
| | - Mark Kramer
- Diabetes Center, Internal Medicine Unit, VU University Medical Center, Amsterdam, the Netherlands
| | - Giel Nijpels
- EMGO+ Institute for Health and Care Research, VU University Medical Center, Department of General Practice, Amsterdam, the Netherlands
| | - Annemarie Simonis-Bik
- Diabetes Center, Internal Medicine Unit, VU University Medical Center, Amsterdam, the Netherlands
| | - Timon W van Haeften
- Department of Internal Medicine, Utrecht University Medical Center, Utrecht, the Netherlands
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI
| | - Richard N Bergman
- Diabetes & Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jaakko Tuomilehto
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
- Dasman Diabetes Institute, Dasman, Kuwait
- Department of Clinical Neurosciences and Preventive Medicine, Danube University Krems, Krems, Austria
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Karina Banasik
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Christopher J Groves
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, U.K
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, U.K
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, U.K
- Oxford Biomedical Research Centre, National Institute for Health Research, Churchill Hospital, Oxford, U.K
| | | | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, Italy
| | - Thomas A Buchanan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Diabetes & Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA
| | - Anny H Xiang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Anette P Gjesing
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Eiberg
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yii-Derr Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Leslie Baier
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark Walker
- Institute of Cellular Medicine, Newcastle University, Newcastle, U.K.
| | - Richard M Watanabe
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Diabetes & Obesity Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Leen M 't Hart
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Molecular Epidemiology, 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
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ
| | - Timothy M Frayling
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Exeter, U.K.
| |
Collapse
|
90
|
Pan XH, Huang X, Ruan GP, Pang RQ, Chen Q, Wang JX, He J, Zhao J, Cai XM, Zhao N, Chen Y, Zhu XQ. Umbilical cord mesenchymal stem cells are able to undergo differentiation into functional islet-like cells in type 2 diabetic tree shrews. Mol Cell Probes 2017; 34:1-12. [DOI: 10.1016/j.mcp.2017.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 12/19/2022]
|
91
|
Zarkoob H, Lewinsky S, Almgren P, Melander O, Fakhrai-Rad H. Utilization of genetic data can improve the prediction of type 2 diabetes incidence in a Swedish cohort. PLoS One 2017; 12:e0180180. [PMID: 28700623 PMCID: PMC5507496 DOI: 10.1371/journal.pone.0180180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/12/2017] [Indexed: 01/21/2023] Open
Abstract
The aim of this study was to measure the impact of genetic data in improving the prediction of type 2 diabetes (T2D) in the Malmö Diet and Cancer Study cohort. The current study was performed in 3,426 Swedish individuals and utilizes of a set of genetic and environmental risk data. We first validated our environmental risk model by comparing it to both the Finnish Diabetes Risk Score and the T2D risk model derived from the Framingham Offspring Study. The area under the curve (AUC) for our environmental model was 0.72 [95% CI, 0.69–0.74], which was significantly better than both the Finnish (0.64 [95% CI, 0.61–0.66], p-value < 1 x 10−4) and Framingham (0.69 [95% CI, 0.66–0.71], p-value = 0.0017) risk scores. We then verified that the genetic data has a statistically significant positive correlation with incidence of T2D in the studied population. We also verified that adding genetic data slightly but statistically increased the AUC of a model based only on environmental risk factors (RFs, AUC shift +1.0% from 0.72 to 0.73, p-value = 0.042). To study the dependence of the results on the environmental RFs, we divided the population into two equally sized risk groups based only on their environmental risk and repeated the same analysis within each subpopulation. While there is a statistically significant positive correlation between the genetic data and incidence of T2D in both environmental risk categories, the positive shift in the AUC remains statistically significant only in the category with the lower environmental risk. These results demonstrate that genetic data can be used to increase the accuracy of T2D prediction. Also, the data suggests that genetic data is more valuable in improving T2D prediction in populations with lower environmental risk. This suggests that the impact of genetic data depends on the environmental risk of the studied population and thus genetic association studies should be performed in light of the underlying environmental risk of the population.
Collapse
Affiliation(s)
- Hadi Zarkoob
- BaseHealth Inc., Sunnyvale, California, United States of America
| | - Sarah Lewinsky
- BaseHealth Inc., Sunnyvale, California, United States of America
| | - Peter Almgren
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | |
Collapse
|
92
|
Chen CH, Yang JH, Chiang CWK, Hsiung CN, Wu PE, Chang LC, Chu HW, Chang J, Song IW, Yang SL, Chen YT, Liu FT, Shen CY. Population structure of Han Chinese in the modern Taiwanese population based on 10,000 participants in the Taiwan Biobank project. Hum Mol Genet 2017; 25:5321-5331. [PMID: 27798100 DOI: 10.1093/hmg/ddw346] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
The Taiwan Biobank (TWB) aims to build a nationwide research database that integrates genomic/epigenomic profiles, lifestyle patterns, dietary habits, environmental exposure history and long-term health outcomes of 300,000 residents of Taiwan. We describe here an investigation of the population structure of Han Chinese on this Pacific island using genotype data of 591,048 SNPs in an initial freeze of 10,801 unrelated TWB participants. In addition to the North-South cline reported in other Han Chinese populations, we find the Taiwanese Han Chinese clustered into three cline groups: 5% were of northern Han Chinese ancestry, 79.9% were of southern Han Chinese ancestry, and 14.5% belonged to a third (T) group. We also find that this T group is genetically distinct from neighbouring Southeast Asians and Austronesian tribes but similar to other southern Han Chinese. Interestingly, high degree of LD between HLA haplotype A*33:03-B*58:01, an MHC allele being of pathological relevance, and SNPs across the MHC region was observed in subjects with T origin, but not in other Han Chinese. This suggested the T group individuals may have experienced evolutionary events independent from the other southern Han Chinese. Based on the newly-discovered population structure, we detect different loci susceptible to type II diabetes in individuals with southern and northern Han Chinese ancestries. Finally, as one of the largest dataset currently available for the Chinese population, genome-wide statistics for the 10,810 subjects are made publicly accessible through Taiwan View (https://taiwanview.twbiobank.org.tw/index; date last accessed October 14, 2016) to encourage future genetic research and collaborations with the island Taiwan.
Collapse
Affiliation(s)
- Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan.,National Center for Genome Medicine, Academia Sinica, Taipei City, Taiwan.,School of Chinese Medicine, China Medical University, Taichung City, Taiwan
| | - Jenn-Hwai Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan.,National Center for Genome Medicine, Academia Sinica, Taipei City, Taiwan
| | - Charleston W K Chiang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, UCLA, CA, USA
| | - Chia-Ni Hsiung
- Taiwan Biobank, Academia Sinica, Taipei City, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Pei-Ei Wu
- Taiwan Biobank, Academia Sinica, Taipei City, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Li-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan.,National Center for Genome Medicine, Academia Sinica, Taipei City, Taiwan
| | - Hou-Wei Chu
- Taiwan Biobank, Academia Sinica, Taipei City, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Josh Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - I-Wen Song
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Show-Ling Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Fu-Tong Liu
- Taiwan Biobank, Academia Sinica, Taipei City, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan
| | - Chen-Yang Shen
- Taiwan Biobank, Academia Sinica, Taipei City, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei City, Taiwan.,College of Public Health, China Medical University, Taichung City, Taiwan
| |
Collapse
|
93
|
Chiang HL, Wang NHH, Song IW, Chang CP, Wen MS, Chien YH, Hwu WL, Tsai FJ, Chen YT, Wu JY. Genetic epidemiological study doesn't support GLA IVS4+919G>A variant is a significant mutation in Fabry disease. Mol Genet Metab 2017; 121:22-27. [PMID: 28377241 DOI: 10.1016/j.ymgme.2017.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND The GLA IVS4+919G>A which is linked to late-onset Fabry disease shows high frequency in Taiwan. METHODS To determine whether IVS4+919G>A is a frequent cause of heart disease, we genotyped it in normal controls and other disease cohorts (type 2 diabetes, heart failure, ventricular tachycardia, atrial fibrillation and coronary artery disease). Normal controls and diabetes patients carrying the variant were evaluated for their cardiac condition. Minigene constructs were used to study GLA splicing patterns in different cell lines. RESULTS GLA IVS4+919A was found in 4/1634 males (0.245%) and 2/1634 females (0.123%) in normal controls and in 4/2133 males (0.188%) and 7/1816 females (0.385%) in the type 2 diabetes cohort. Of all the 17 IVS4+919A carriers in these two groups, only two males reported heart-related disease (myocardial infarction and hypertensive heart disease). Furthermore, in the heart disease cohort (n=649), only one male carried the variant. Minigene constructs showed that the AGS (stomach) cell line showed a distinct GLA splicing pattern. CONCLUSION Most subjects carrying GLA IVS4+919A did not show abnormal cardiac phenotypes. The near-absence of GLA IVS4+919A in heart disease cohort suggested that this variant is not a frequent cause of overt heart diseases in Taiwan and that the genotype-phenotype correlation and natural course of the disease need further investigation. We also showed that the GLA IVS4+919G>A nucleotide change did influence alternative splicing in a tissue-specific manner. SYNOPSIS The GLA IVS4+919G>A variant is not a frequent cause of overt heart disease in Taiwan.
Collapse
Affiliation(s)
- Hung-Lun Chiang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - I-Wen Song
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chun-Ping Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ming-Shien Wen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Genetics, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yuan-Tsong Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Department of Pediatrics, Duke University Medical Center, Durham, USA.
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan.
| |
Collapse
|
94
|
Yang S, Zhao H, Xu K, Qian Y, Wu M, Yang T, Chen Y, Zhao X, Chen J, Wen J, Hu Z, Gu HF, Shen H, Shen C. Evaluation of common variants in MG53 and the risk of type 2 diabetes and insulin resistance in Han Chinese. SPRINGERPLUS 2017; 5:612. [PMID: 28443211 PMCID: PMC5395513 DOI: 10.1186/s40064-016-2218-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/22/2016] [Indexed: 11/10/2022]
Abstract
Abnormally increased skeletal-muscle-specific E3 ubiquitin ligase (MG53) is associated with the inhibition of insulin signalling and insulin resistance (IR) in animal models. Four community-based studies of Han Chinese populations were included in this study to test the association of variants of MG53 and type 2 diabetes (T2D). The results showed that rs7186832 and rs12929077 in MG53 were significantly associated with T2D and impaired fasting glucose (IFG) of females in the discovery-stage case-control study and cohort study respectively of rural population but not in the replication sample of urban population. In rural population, the fasting insulin (mU/L) of the subjects with AA, AG and GG genotypes in rs12929077 were 8.70 ± 8.05, 10.71 ± 11.16 and 13.41 ± 14.26, respectively, and increased linearly in T2D cases without medication treatment (P = 0.04). This variant was significantly associated with HOMA-IR (P = 0.020) and HOMA-IS (P = 0.023). In individuals with IFG, the insulin and HOMA-IR of AG carriers were significantly higher than those of AA carriers. In urban population, after glucose loading, there were significant differences in the 30-min glucose, the area under the curve (AUC) of 30-min glucose and the AUC of 120-min glucose according to the genotypes of rs7186832 and rs12929077 in males but not females. Our findings suggest that MG53 variants might confer risk susceptibility to the development of T2D of females and IR particularly in rural population.
Collapse
Affiliation(s)
- Song Yang
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, 214200 China
| | - Hailong Zhao
- Department of Epidemiology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning, Nanjing, 211166 China
| | - Kuangfeng Xu
- Department of Endocrinology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029 China
| | - Yun Qian
- Department of Chronic Non-communicable Disease Control, Wuxi Center for Disease Control and Prevention, Wuxi, 214023 China
| | - Ming Wu
- Institute of Chronic Disease Control, Center for Disease Control and Prevention of Jiangsu Province, Nanjing, 210009 China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029 China
| | - Yanchun Chen
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, 214200 China
| | - Xianghai Zhao
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, 214200 China
| | - Jinfeng Chen
- Department of Clinical Epidemiology, Jiangsu Province Geriatrics Institute, Nanjing, 210024 China
| | - Jinbo Wen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning, Nanjing, 211166 China
| | - Zhibing Hu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning, Nanjing, 211166 China
| | - Harvest F Gu
- Department of Molecular Medicine and Surgery, Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Hongbing Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning, Nanjing, 211166 China
| | - Chong Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Jiangning, Nanjing, 211166 China.,Department of Clinical Epidemiology, Jiangsu Province Geriatrics Institute, Nanjing, 210024 China
| |
Collapse
|
95
|
Huang KC, Li TM, Liu X, Chen JH, Chien WK, Shiao YT, Tsang H, Lin TH, Liao CC, Huang SM, Li JP, Lin CW, Lin JC, Lin CC, Lai CH, Cheng CF, Liang WM, Hung CH, Chen CC, Lin YJ, Tsai FJ. KCNQ1 variants associate with hypertension in type 2 diabetes and affect smooth muscle contractility in vitro. J Cell Physiol 2017; 232:3309-3316. [PMID: 28059450 DOI: 10.1002/jcp.25775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/19/2022]
Abstract
KCNQ1 encodes a potassium voltage-gated channel and represents a susceptibility locus for type 2 diabetes mellitus (T2DM). Here, we explored the association between KCNQ1 polymorphisms and hypertension risk in individuals with T2DM, as well as the role of KCNQ1 in vascular smooth muscle cell contraction in vitro. To investigate the relationship between KCNQ1 and the risk of developing hypertension in patients with T2DM, we divided the T2DM cohort into hypertension (n = 452) and non-hypertension (n = 541) groups. The Mann-Whitney U test, chi-square test, and multivariate regression analyses were used to assess the clinical characteristics and genotypic frequencies. In vitro studies utilized the rat aortic smooth muscle A10 cell line. Patients in the hypertension group were significantly older at the time of enrollment and had higher levels of body mass index, waist-to-hip ratio, and triglyceride than those in the non-hypertension group. The KCNQ1 rs3864884 and rs12576239 genetic variants were associated with hypertension in T2DM. KCNQ1 expression was lower in the individuals with the CC versus the CT and TT genotypes. Smooth muscle cell contractility was inhibited by treatment with a KCNQ1 inhibitor. These results suggest that KCNQ1 might be associated with hypertension in individuals with T2DM.
Collapse
Affiliation(s)
- Kuo-Chin Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Integration of Traditional Chinese and Western Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Te-Mao Li
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Xiang Liu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jin-Hua Chen
- Biostatistics Center and School of Public Health, China Medical University, Taichung, Taiwan.,Biostatistics Center and School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Wen-Kuei Chien
- National Applied Research Laboratories, National Center for High-Performance Computing, Hsinchu, Taiwan
| | - Yi-Tzone Shiao
- Heart Center, China Medical University Hospital, Taichung, Taiwan
| | - Hsinyi Tsang
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Ting-Hsu Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Chu Liao
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Shao-Mei Huang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Ju-Pi Li
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Rheumatism Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Jung-Chun Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chien Lin
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Fung Cheng
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, Taichung, Taiwan
| | - Wen-Miin Liang
- Graduate Institute of Biostatistics, School of Public Health, China Medical University, Taichung, Taiwan
| | - Chien-Hui Hung
- Graduate Institute of Clinical Medical Science, Chang-Gung University, Taipei, Taiwan
| | - Ching-Chu Chen
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ying-Ju Lin
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan.,Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,Asia University, Taichung, Taiwan
| |
Collapse
|
96
|
Plasma miR-21 expression: an indicator for the severity of Type 2 diabetes with diabetic retinopathy. Biosci Rep 2017; 37:BSR20160589. [PMID: 28108673 PMCID: PMC5469322 DOI: 10.1042/bsr20160589] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/18/2017] [Accepted: 01/20/2017] [Indexed: 12/12/2022] Open
Abstract
To investigate the roles of plasma miR-21 in the pathogenic process of Type 2 diabetes (T2D) with diabetic retinopathy (DR). T2D patients included patients without DR (NDR) group, patients with non-proliferative/background DR (BDR) group and patients with proliferative DR (PDR) group. Healthy individuals served as control group. Fasting plasma glucose (FPG), glycosylated haemoglobin (HbA1c), triacylglycerol (TG), total cholesterol (TC), urine creatinine (Cr), fasting blood glucose (FBG), blood urea nitrogen (BUN), low-density lipoprotein cholesterol (LDL-C), fasting insulin (FINS) and plasma miR-21 expression were measured. Quantitative real-time PCR (qRT-PCR) was applied to detect miR-21 expression. Pearson analysis was used to conduct correlation analysis and receiver operating characteristic (ROC) curve was used to analyse the diagnostic value of miR-21 in T2D with DR. Compared with the control group, FBG and HbA1c increased in the NDR group; compared with the control and NDR groups, disease course, HbA1c, FPG levels and homoeostasis model assessment of insulin resistance (HOMA-IR) were increased in the BDR and PDR groups; and compared with the BDR group, disease course, HbA1c and FPG levels were higher in the PDR group. miR-21 expression was higher in the BDR group than the control group, and higher in the PDR group than the BDR group. miR-21 expression was positively related with disease course, HbA1C, FPG and HOMA-IR, and had diagnostic value for T2D with DR and PDR. The plasma miR-21 expression was increased in the development of T2D with DR and can be used as an indicator for the severity of T2D with DR.
Collapse
|
97
|
Chen G, Zhang Z, Adebamowo SN, Liu G, Adeyemo A, Zhou Y, Doumatey AP, Wang C, Zhou J, Yan W, Shriner D, Tekola-Ayele F, Bentley AR, Jiang C, Rotimi CN. Common and rare exonic MUC5B variants associated with type 2 diabetes in Han Chinese. PLoS One 2017; 12:e0173784. [PMID: 28346466 PMCID: PMC5367689 DOI: 10.1371/journal.pone.0173784] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/27/2017] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies have identified over one hundred common genetic risk variants associated with type 2 diabetes (T2D). However, most of the heritability of T2D has not been accounted for. In this study, we investigated the contribution of rare and common variants to T2D susceptibility by analyzing exome array data in 1,908 Han Chinese genotyped with Affymetrix Axiom® Exome Genotyping Arrays. Based on the joint common and rare variants analysis of 57,704 autosomal SNPs within 12,244 genes using Sequence Kernel Association Tests (SKAT), we identified significant associations between T2D and 25 variants (9 rare and 16 common) in MUC5B, p-value 1.01×10−14. This finding was replicated (p = 0.0463) in an independent sample that included 10,401 unrelated individuals. Sixty-six of 1,553 possible haplotypes based on 25 SNPs within MUC5B showed significant association with T2D (Bonferroni corrected p values < 3.2×10−5). The expression level of MUC5B is significantly higher in pancreatic tissues of persons with T2D compared to those without T2D (p-value = 5×10−5). Our findings suggest that dysregulated MUC5B expression may be involved in the pathogenesis of T2D. As a strong candidate gene for T2D, MUC5B may play an important role in the mechanisms underlying T2D etiology and its complications.
Collapse
Affiliation(s)
- Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (CNR); (GC)
| | | | - Sally N. Adebamowo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yanxun Zhou
- Suizhou Central Hospital, Suizhou, Hubei, China
| | - Ayo P. Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fasil Tekola-Ayele
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy R. Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Charles N. Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (CNR); (GC)
| |
Collapse
|
98
|
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]
|
99
|
Chikowore T, van Zyl T, Feskens EJM, Conradie KR. Predictive utility of a genetic risk score of common variants associated with type 2 diabetes in a black South African population. Diabetes Res Clin Pract 2016; 122:1-8. [PMID: 27744072 DOI: 10.1016/j.diabres.2016.09.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 01/24/2023]
Abstract
AIMS To determine the predictive utility of polygenic risk scores of common variants associated with type 2 diabetes derived from the European and Asian ethnicities among a black South African population. METHOD Our study was a case-control study nested within the Prospective Urban and Rural Epidemiological (PURE) study of 178 male and female cases, matched for age and gender with 178 controls. Four types of genetic risk scores (GRS) were developed from 66 selected SNPs. These comprised of beta cell related variants (GRSb), variants which had significant associations with T2D in our study (GRSn), variants from the trans-ethnic meta-analysis (GRStrans) and all the 66 selected SNPs (GRSt). RESULTS Of the GRS's, only GRSn was associated with increased risk of T2D as indicated by an OR (95CI) of 1.21 (1.02-1.43) p-value=0.015. Stratified analysis of age and BMI, indicated the GRSn to be significantly associated with T2D among the non-obese and participants less than 50years. The area under the ROC of the T2D risk factors only was 0.652 (p value<0.001) and with the addition of GRSn it was 0.665 (p value<0.001). CONCLUSIONS The GRS of European and Asian derived variants have limited clinical utility in the black South African population. The inclusion of population specific variants in the GRS is pivotal.
Collapse
Affiliation(s)
- Tinashe Chikowore
- Centre for Excellence in Nutrition, North-West University, Potchefstroom, North West Province 2520, South Africa.
| | - Tertia van Zyl
- Centre for Excellence in Nutrition, North-West University, Potchefstroom, North West Province 2520, South Africa
| | - Edith J M Feskens
- Wageningen University, Division of Human Nutrition, P.O. Box 17, 6700 AA Wageningen, The Netherlands
| | - Karin R Conradie
- Centre for Excellence in Nutrition, North-West University, Potchefstroom, North West Province 2520, South Africa
| |
Collapse
|
100
|
Lockridge AD, Baumann DC, Akhaphong B, Abrenica A, Miller RF, Alejandro EU. Serine racemase is expressed in islets and contributes to the regulation of glucose homeostasis. Islets 2016; 8:195-206. [PMID: 27880078 PMCID: PMC5161142 DOI: 10.1080/19382014.2016.1260797] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
NMDA receptors (NMDARs) have recently been discovered as functional regulators of pancreatic β-cell insulin secretion. While these excitatory receptor channels have been extensively studied in the brain for their role in synaptic plasticity and development, little is known about how they work in β-cells. In neuronal cells, NMDAR activation requires the simultaneous binding of glutamate and a rate-limiting co-agonist, such as D-serine. D-serine levels and availability in most of the brain rely on endogenous synthesis by the enzyme serine racemase (Srr). Srr transcripts have been reported in human and mouse islets but it is not clear whether Srr is functionally expressed in β-cells or what its role in the pancreas might be. In this investigation, we reveal that Srr protein is highly expressed in primary human and mouse β-cells. Mice with whole body deletion of Srr (Srr KO) show improved glucose tolerance through enhanced insulin secretory capacity, possibly through Srr-mediated alterations in islet NMDAR expression and function. We observed elevated insulin sensitivity in some animals, suggesting Srr metabolic regulation in other peripheral organs as well. Srr expression in neonatal and embryonic islets, and adult deficits in Srr KO pancreas weight and islet insulin content, point toward a potential role for Srr in pancreatic development. These data reveal the first evidence that Srr may regulate glucose homeostasis in peripheral tissues and provide circumstantial evidence that D-serine may be an endogenous islet NMDAR co-agonist in β-cells.
Collapse
Affiliation(s)
| | - Daniel C. Baumann
- Department of Integrative Biology & Physiology, Minneapolis, MN, USA
| | - Brian Akhaphong
- Department of Integrative Biology & Physiology, Minneapolis, MN, USA
| | - Alleah Abrenica
- Department of Integrative Biology & Physiology, Minneapolis, MN, USA
| | - Robert F. Miller
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Emilyn U. Alejandro
- Department of Integrative Biology & Physiology, Minneapolis, MN, USA
- CONTACT Emilyn U. Alejandro Department of Integrative Biology & Physiology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|