1
|
Yanasegaran K, Ng JYE, Chua EW, Nawi AM, Ng PY, Abdul Manaf MR. Single nucleotide polymorphisms (SNPs) that are associated with obesity and type 2 diabetes among Asians: a systematic review and meta-analysis. Sci Rep 2024; 14:20062. [PMID: 39209904 PMCID: PMC11362307 DOI: 10.1038/s41598-024-70674-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Single nucleotide polymorphisms (SNPs) could increase the susceptibility of individuals to develop obesity and type 2 diabetes (T2DM). Obesity and T2DM are closely related pathophysiologically, thus similar SNPs could mediate both these diseases, but this is rarely reported. Furthermore, limited studies have been performed to summarize SNP data in the Asian population compared to the Western population. In this study, we aimed to summarize SNPs that are associated with the development of obesity and T2DM among Asian populations. We searched six literature databases and Review Manager (RevMan) was used for meta-analysis. The pooled odds ratios (ORs) and 95% CIs were calculated with a random effects model for the heterogeneity among studies. The pooled analysis showed that rs9939609 (FTO gene) and rs17782313 and rs571312 (MC4R gene) are associated with obesity with an odd ratio (OR) of 1.37, 1.36 and 1.29 respectively. For T2DM, five SNPs, rs7903146 and rs12255372 (TCF7L2 gene), rs13266634 and rs11558471 (SLC30A8 gene) and rs2283228 (KCNQ1 gene) have also shown strong associations with T2DM at OR of 1.64, 1.61, 1.22, 1.29 and 1.60 respectively. This data could be used to develop a gene screening panel for assessing obesity and T2DM susceptibility.
Collapse
Affiliation(s)
- Kevina Yanasegaran
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, 50300, Kuala Lumpur, Malaysia
| | - Jeremy Yung Ern Ng
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, 50300, Kuala Lumpur, Malaysia
| | - Eng Wee Chua
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, 50300, Kuala Lumpur, Malaysia
| | - Azmawati Mohammed Nawi
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Kuala Lumpur, Malaysia
| | - Pei Yuen Ng
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, 50300, Kuala Lumpur, Malaysia.
| | - Mohd Rizal Abdul Manaf
- Department of Public Health Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
2
|
Le J, Chen Y, Yang W, Chen L, Ye J. Metabolic basis of solute carrier transporters in treatment of type 2 diabetes mellitus. Acta Pharm Sin B 2024; 14:437-454. [PMID: 38322335 PMCID: PMC10840401 DOI: 10.1016/j.apsb.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/10/2023] [Accepted: 08/09/2023] [Indexed: 02/08/2024] Open
Abstract
Solute carriers (SLCs) constitute the largest superfamily of membrane transporter proteins. These transporters, present in various SLC families, play a vital role in energy metabolism by facilitating the transport of diverse substances, including glucose, fatty acids, amino acids, nucleotides, and ions. They actively participate in the regulation of glucose metabolism at various steps, such as glucose uptake (e.g., SLC2A4/GLUT4), glucose reabsorption (e.g., SLC5A2/SGLT2), thermogenesis (e.g., SLC25A7/UCP-1), and ATP production (e.g., SLC25A4/ANT1 and SLC25A5/ANT2). The activities of these transporters contribute to the pathogenesis of type 2 diabetes mellitus (T2DM). Notably, SLC5A2 has emerged as a valid drug target for T2DM due to its role in renal glucose reabsorption, leading to groundbreaking advancements in diabetes drug discovery. Alongside SLC5A2, multiple families of SLC transporters involved in the regulation of glucose homeostasis hold potential applications for T2DM therapy. SLCs also impact drug metabolism of diabetic medicines through gene polymorphisms, such as rosiglitazone (SLCO1B1/OATP1B1) and metformin (SLC22A1-3/OCT1-3 and SLC47A1, 2/MATE1, 2). By consolidating insights into the biological activities and clinical relevance of SLC transporters in T2DM, this review offers a comprehensive update on their roles in controlling glucose metabolism as potential drug targets.
Collapse
Affiliation(s)
- Jiamei Le
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yilong Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wei Yang
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Ligong Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Jianping Ye
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
- Research Center for Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
3
|
Zeng Q, Tan B, Han F, Huang X, Huang J, Wei Y, Guo R. Association of solute carrier family 30 A8 zinc transporter gene variations with gestational diabetes mellitus risk in a Chinese population. Front Endocrinol (Lausanne) 2023; 14:1159714. [PMID: 37324267 PMCID: PMC10266221 DOI: 10.3389/fendo.2023.1159714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Background The solute carrier family 30 A8 zinc transporter (SLC30A8) plays a crucial role in insulin secretion. This study aimed to investigate the impact of SLC30A8 gene polymorphisms on gestational diabetes mellitus (GDM). Methods The research objective was to select 500 patients with GDM and 502 control subjects. Rs13266634 and rs2466293 were genotyped using the SNPscan™ genotyping assay. Statistical tests, such as the chi-square test, t-test, logistic regression, ANOVA, and meta-analysis, were conducted to determine the differences in genotypes, alleles, and their associations with GDM risk. Results Statistically significant differences were observed in age, pregestational BMI, SBP, DBP, and parity between individuals with GDM and healthy subjects (P < 0.05). After adjusting for these factors, rs2466293 remained significantly associated with an increased risk of GDM in overall subjects (GG+AG vs. AA: OR = 1.310; 95% CI: 1.005-1.707; P = 0.046, GG vs. AA: OR = 1.523; 95% CI: 1.010-2.298; P = 0.045 and G vs. A: OR = 1.249; 95% CI: 1.029-1.516; P = 0.024). Rs13266634 was still found to be significantly associated with a decreased risk of GDM in individuals aged ≥ 30 years (TT vs. CT+CC: OR = 0.615; 95% CI: 0.392-0.966; P = 0.035, TT vs. CC: OR = 0.503; 95% CI: 0.294-0.861; P = 0.012 and T vs. C: OR =0.723; 95% CI: 0.557-0.937; P = 0.014). Additionally, the haplotype CG was found to be associated with a higher risk of GDM (P < 0.05). Furthermore, pregnant women with the CC or CT genotype of rs13266634 exhibited significantly higher mean blood glucose levels than those with the TT genotype (P < 0.05). Our findings were further validated by the results of a meta-analysis. Conclusion The SLC30A8 rs2466293 polymorphism was found to be associated with an increased risk of GDM, while rs13266634 was associated with a decreased risk of GDM in individuals aged ≥ 30 years. These findings provide a theoretical basis for GDM testing.
Collapse
Affiliation(s)
- Qiaoli Zeng
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Maternal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Bing Tan
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Department of Endocrinology, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengqiong Han
- Department of Obstetric, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Xiujuan Huang
- Department of Children’s Health, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Jinzhi Huang
- Department of Gynaecology, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Yue Wei
- Department of Ultrasound, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Runmin Guo
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Maternal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Department of Endocrinology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
4
|
Chi Y, Zhang X, Liang D, Wang Y, Cai X, Dong J, Li L, Chi Z. ZnT8 Exerts Anti-apoptosis of Kidney Tubular Epithelial Cell in Diabetic Kidney Disease Through TNFAIP3-NF-κB Signal Pathways. Biol Trace Elem Res 2023; 201:2442-2457. [PMID: 35871203 DOI: 10.1007/s12011-022-03361-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/13/2022] [Indexed: 02/05/2023]
Abstract
Apoptosis of kidney tubular epithelial cells contributes to diabetic kidney disease (DKD) pathophysiology, but the mechanisms are not fully understood. Zinc transporter protein member 8 (ZnT8, SLC30A8) is a susceptive gene in diabetes. Here, we aim to investigate whether ZnT8 has effects on pathophysiology of DKD. The animal groups include control, ZnT8KO mice, STZ-induced, and ZnT8-KO-STZ. STZ-induced DKD was performed in male C57BL/6 J mice and in ZnT8-KO mice. High glucose (HG)-induced apoptosis in a normal rat kidney tubular epithelial cell line (NRK-52E cells) was performed in vitro. Transfection of hZnT8-EGFP or TNFAIP3 siRNA was done in NRK-52E cells. Flow cytometry with Annexin V-FITC/PI double staining and TUNEL analysis was performed for the detection of apoptosis. Gene expression at mRNA and protein levels was examined with real-time RT-PCR and Western blot. Urine albumin to creatinine ratio, proinflammatory cytokines, and apoptosis were enhanced in kidneys of STZ and ZnT8-KO-STZ mice compared to control or ZnT8-KO mice. ZnT8 overexpression after hZnT8-EGFP transfection decreased HG-stimulated inflammatory activity and apoptosis in NRK-52E cells. Furthermore, treatment with ZnSO4 blunted HG-induced apoptosis and NF-κB activation. ZnSO4 increased the abundance of zinc-finger protein TNF-α-induced protein 3 (TNFAIP3). Also, ZnT8 over-expression after hZnT8-EGFP transfection significantly ameliorates HG-induced NF-κB-dependent transcriptional activity and apoptotic protein expressions in NRK-52E cells, but the inhibitory effect of ZnT8 was significantly abolished with TNFAIP3 siRNA. Our study provides evidence that ZnT8 has protective effects against apoptosis of renal tubular epithelial cells through induction of TNFAIP3 and subsequent suppression of the NF-κB pathway.
Collapse
Affiliation(s)
- Yinmao Chi
- Department of Physiology, China Medical University, Shenyang, Liaoning Province, 110001, People's Republic of China
| | - Xiuli Zhang
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong Province, 518000, People's Republic of China.
- Department of Nephrology, Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, 518000, People's Republic of China.
| | - Dan Liang
- Troops of 95988 Unit, Changchun, Jilin, 158000, People's Republic of China
| | - Yue Wang
- Department of Tissue Culture, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, 110001, People's Republic of China
| | - Xiaoyi Cai
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong Province, 518000, People's Republic of China
- Shantou University Medical College, Shantou, Guangdong Province, 515000, People's Republic of China
| | - Jiqiu Dong
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong Province, 518000, People's Republic of China
| | - Lingzhi Li
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong Province, 518000, People's Republic of China
| | - Zhihong Chi
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning Province, 110001, People's Republic of China
| |
Collapse
|
5
|
da Costa SKS, Almeida JA, Pires LV, Brandão-Lima PN, Rogero MM, Mendes-Netto RS. Relationship Between the Single Nucleotide Polymorphism rs11558471 in the SLC30A8/ZnT8 Gene and Cardiometabolic Markers in Postmenopausal Women. Biol Trace Elem Res 2023; 201:2183-2190. [PMID: 35793043 DOI: 10.1007/s12011-022-03337-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/19/2022] [Indexed: 11/02/2022]
Abstract
Postmenopausal women have more risk factors for metabolic syndrome, and genetic alterations in SLC30A8 (zinc transporter 8 [ZnT8]) are directly related to these factors. Our aim was to assess the relationship of the single nucleotide polymorphism (SNP) rs11558471 in the SLC30A8/ZnT8 gene with cardiometabolic markers in postmenopausal women. This cross-sectional study included 53 postmenopausal women divided into two groups according to the SNP genotype (AG + GG [n = 25] and AA [n = 28]). Anthropometric, dietary, and biochemical (glycemic, lipidic, hepatic, renal, and hormonal markers) variables were evaluated and compared between groups. No differences in glycemic, hepatic, renal, and hormonal markers were found between groups. However, the group with the polymorphic allele (AG + GG) had a better lipid profile than non-carriers (total cholesterol, p = 0.041; low-density lipoprotein cholesterol [LDL-c], p = 0.035; non-high-density lipoprotein cholesterol [non-HDL-c], p = 0.043). Logistic regression showed that the group with polymorphic allele had lower chances of increasing levels of LDL-c (odds ratio [OR] = 0.225, p = 0.012) and non-HDL-c (OR = 0.316, p = 0.045). After adjusting for age, body mass index, physical activity, and use of diabetes and dyslipidemia drugs, only LDL-c remained associated (OR = 0.218; p = 0.017). The variant allele of SNP rs11558471 in the SLC30A8 gene was associated with better LDL-c levels, which helps reduce the risks for cardiovascular diseases in postmenopausal women.
Collapse
Affiliation(s)
- Sheila Kely Santos da Costa
- Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, 49100-000, Brazil
| | - Jamylle Araújo Almeida
- Department of Nutrition, Center for Biological and Health Sciences, Nutrition Sciences Post-Graduation Program, Federal University of Sergipe, Av. Marechal Rondon, s/n - Jd. Rosa Elze, São Cristóvão, Sergipe, 49100-000, Brazil
| | - Liliane Viana Pires
- Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, 49100-000, Brazil.
- Department of Nutrition, Center for Biological and Health Sciences, Nutrition Sciences Post-Graduation Program, Federal University of Sergipe, Av. Marechal Rondon, s/n - Jd. Rosa Elze, São Cristóvão, Sergipe, 49100-000, Brazil.
| | - Paula Nascimento Brandão-Lima
- Nutrition in Public Health Post-Graduation Program, Faculty of Public Health, University of São Paulo, São Paulo, São Paulo, 01246904, Brazil
| | - Marcelo Macedo Rogero
- Nutrition in Public Health Post-Graduation Program, Faculty of Public Health, University of São Paulo, São Paulo, São Paulo, 01246904, Brazil
| | - Raquel Simões Mendes-Netto
- Department of Nutrition, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, 49100-000, Brazil
- Department of Nutrition, Center for Biological and Health Sciences, Nutrition Sciences Post-Graduation Program, Federal University of Sergipe, Av. Marechal Rondon, s/n - Jd. Rosa Elze, São Cristóvão, Sergipe, 49100-000, Brazil
| |
Collapse
|
6
|
Abu Seman N, Othman SH. Recent Progress in Genetics and Epigenetics Research on Diabetic Nephropathy in Malaysia. J Diabetes Res 2023; 2023:9053580. [PMID: 37187702 PMCID: PMC10181909 DOI: 10.1155/2023/9053580] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/15/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Diabetic nephropathy is a multifactorial disease. Gene susceptibility, as well as environmental exposure, plays an important role in disease progression. Malaysia is reported to be among the world's second-fastest-growing rates of kidney failure. Diabetic nephropathy has become the main cause of end-stage renal disease in Malaysia. This article is aimed at reviewing genetic studies conducted among diabetic nephropathy patients in the Malaysian population. This review was conducted by searching PubMed, MEDLINE, and Google Scholar databases to identify all relevant papers published in English from March 2022 to April 2022, using the following keywords: diabetes, type 2 diabetes, diabetic nephropathy, diabetic kidney disease, and Malaysia. The case-control study among diabetic patients with and without diabetic nephropathy showed a significant association with diabetic nephropathy in CNDP1, NOS3, and MnSOD genes. In the ethnic subgroup analysis, significant differences for diabetic nephropathy in terms of diabetes duration (≥10 years) were observed for CCL2 rs3917887, CCR5 rs1799987, ELMO1 rs74130, and IL8 rs4073. The IL8 rs4073 was associated only with the Indians, while the CCR5 rs1799987 was associated with the Chinese. In Malays, SLC12A3 Arg913Gln polymorphism and ICAM1 K469E (A/G) polymorphism were found to be associated with diabetic nephropathy. Studies on gene-environment interactions have suggested significant genetic and environmental factors such as smoking, waist circumference, and sex for eNOS rs2070744, PPARGC1A rs8192678, KCNQ1 rs2237895, and KCNQ1 rs2283228 with kidney disease. The genetic variants' contributions differed across ethnic groups. Therefore, a study to validate the genetic variants that are found to be associated with different ethnicities in Malaysia may be important in future studies.
Collapse
Affiliation(s)
- Norhashimah Abu Seman
- Endocrine and Metabolic Unit, Nutrition, Metabolism and Cardiovascular Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Setia Alam, 40170 Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Siti Haslina Othman
- Endocrine and Metabolic Unit, Nutrition, Metabolism and Cardiovascular Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Setia Alam, 40170 Shah Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
7
|
Goyal Y, Verma AK, Kumar S, Bhatt D, Ahmad F, Dev K. Association of SLC30A8 (rs13266634) and GLIS3 (rs7034200) gene variant in development of type 2 diabetes mellitus in Indian population: A case-control study. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
8
|
Hu F, Zhang Y, Qin P, Zhao Y, Liu D, Zhou Q, Tian G, Li Q, Guo C, Wu X, Qie R, Huang S, Han M, Li Y, Hu D, Zhang M. Integrated analysis of probability of type 2 diabetes mellitus with polymorphisms and methylation of SLC30A8 gene: a nested case-control study. J Hum Genet 2022; 67:651-660. [PMID: 35996015 DOI: 10.1038/s10038-022-01067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/16/2022] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
To estimate the associations between single-nucleotide polymorphisms (SNPs) and methylation of SLC30A8 gene and T2DM risk, and the interactions among SNPs, methylation, and environmental factors on T2DM risk. We genotyped 9 SNPs and tested methylation at 46 CpG loci of SLC30A8 in the baseline DNA of 290 T2DM cases and 290 matched controls nested in the Rural Chinese Cohort Study. A conditional logistic regression model was used to estimate the associations between SNPs and SLC30A8 methylation and T2DM risk. Multifactor Dimensionality Reduction analysis was used to estimate the effect of interactions among SNPs, methylation, and environment on T2DM risk. Probability of T2DM was decreased with rs11558471 (GG vs. AA, OR = 0.55, 95% CI 0.32, 0.96), with rs13266634 (TT vs. CC, OR = 0.55, 95% CI 0.32, 0.94), with rs3802177 (AA vs. GG, OR = 0.54, 95%CI 0.31, 0.94), and its probability was increased with rs2466293 of SLC30A8 (GA vs. AA, OR = 1.63, 95% CI 1.08-2.47). Its probability was also significantly associated with methylation of CG9 and CG45 (OR = 0.56 [95% CI 0.33-0.97] and 1.61 [95%CI 1.03--2.51]). T2DM probability was significantly associated with the interaction effect between rs2466293 and hypertension (p = 0.045). T2DM probability was also significantly associated with the combination effects of rs2466293 with BMI, hypertension, and hypertriglyceridemia, with the combination effects of hypertriglyceridemia with rs11558471, rs13266634, and methylation of CG45.
Collapse
Affiliation(s)
- Fulan Hu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yanyan Zhang
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Pei Qin
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Dechen Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qionggui Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Gang Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Quanman Li
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Chunmei Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Xiaoyan Wu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ranran Qie
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shengbing Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Minghui Han
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yang Li
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Dongsheng Hu
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China
| | - Ming Zhang
- School of Public Health, Shenzhen University Health Science Center, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
9
|
Zhang M, Gao Y, Li Q, Cao H, Yang J, Cai X, Xiao J. Downregulation of DNA methyltransferase-3a ameliorates the osteogenic differentiation ability of adipose-derived stem cells in diabetic osteoporosis via Wnt/β-catenin signaling pathway. Stem Cell Res Ther 2022; 13:397. [PMID: 35927735 PMCID: PMC9351106 DOI: 10.1186/s13287-022-03088-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/23/2022] [Indexed: 02/06/2023] Open
Abstract
Background Diabetes-related osteoporosis (DOP) is a chronic disease caused by the high glucose environment that induces a metabolic disorder of osteocytes and osteoblast-associated mesenchymal stem cells. The processes of bone defect repair and regeneration become extremely difficult with DOP. Adipose-derived stem cells (ASCs), as seed cells in bone tissue engineering technology, provide a promising therapeutic approach for bone regeneration in DOP patients. The osteogenic ability of ASCs is lower in a DOP model than that of control ASCs. DNA methylation, as a mechanism of epigenetic regulation, may be involved in DNA methylation of various genes, thereby participating in biological behaviors of various cells. Emerging evidence suggests that increased DNA methylation levels are associated with activation of Wnt/β-catenin signaling pathway. The purpose of this study was to investigate the influence of the diabetic environment on the osteogenic potential of ASCs, to explore the role of DNA methylation on osteogenic differentiation of DOP-ASCs via Wnt/β-catenin signaling pathway, and to improve the osteogenic differentiation ability of ASCs with DOP. Methods DOP-ASCs and control ASCs were isolated from DOP C57BL/6 and control mice, respectively. The multipotency of DOP-ASCs was confirmed by Alizarin Red-S, Oil Red-O, and Alcian blue staining. Real-time polymerase chain reaction (RT-PCR), immunofluorescence, and western blotting were used to analyze changes in markers of osteogenic differentiation, DNA methylation, and Wnt/β-catenin signaling. Alizarin Red-S staining was also used to confirm changes in the osteogenic ability. DNMT small interfering RNA (siRNA), shRNA-Dnmt3a, and LVRNA-Dnmt3a were used to assess the role of Dnmt3a in osteogenic differentiation of control ASCs and DOP-ASCs. Micro-computed tomography, hematoxylin and eosin staining, and Masson staining were used to analyze changes in the osteogenic capability while downregulating Dnmt3a with lentivirus in DOP mice in vivo. Results The proliferative ability of DOP-ASCs was lower than that of control ASCs. DOP-ASCs showed a decrease in osteogenic differentiation capacity, lower Wnt/β-catenin signaling pathway activity, and a higher level of Dnmt3a than control ASCs. When Dnmt3a was downregulated by siRNA and shRNA, osteogenic-related factors Runt-related transcription factor 2 and osteopontin, and activity of Wnt/β-catenin signaling pathway were increased, which rescued the poor osteogenic potential of DOP-ASCs. When Dnmt3a was upregulated by LVRNA-Dnmt3a, the osteogenic ability was inhibited. The same results were obtained in vivo. Conclusions Dnmt3a silencing rescues the negative effects of DOP on ASCs and provides a possible approach for bone tissue regeneration in patients with diabetic osteoporosis.
Collapse
Affiliation(s)
- Maorui Zhang
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yujin Gao
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qing Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Huayue Cao
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jianghua Yang
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Jingang Xiao
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China. .,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China. .,Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
10
|
Willekens J, Runnels LW. Impact of Zinc Transport Mechanisms on Embryonic and Brain Development. Nutrients 2022; 14:2526. [PMID: 35745255 PMCID: PMC9231024 DOI: 10.3390/nu14122526] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
The trace element zinc (Zn) binds to over ten percent of proteins in eukaryotic cells. Zn flexible chemistry allows it to regulate the activity of hundreds of enzymes and influence scores of metabolic processes in cells throughout the body. Deficiency of Zn in humans has a profound effect on development and in adults later in life, particularly in the brain, where Zn deficiency is linked to several neurological disorders. In this review, we will summarize the importance of Zn during development through a description of the outcomes of both genetic and early dietary Zn deficiency, focusing on the pathological consequences on the whole body and brain. The epidemiology and the symptomology of Zn deficiency in humans will be described, including the most studied inherited Zn deficiency disease, Acrodermatitis enteropathica. In addition, we will give an overview of the different forms and animal models of Zn deficiency, as well as the 24 Zn transporters, distributed into two families: the ZIPs and the ZnTs, which control the balance of Zn throughout the body. Lastly, we will describe the TRPM7 ion channel, which was recently shown to contribute to intestinal Zn absorption and has its own significant impact on early embryonic development.
Collapse
Affiliation(s)
| | - Loren W. Runnels
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
11
|
Kaimala S, Kumar CA, Allouh MZ, Ansari SA, Emerald BS. Epigenetic modifications in pancreas development, diabetes, and therapeutics. Med Res Rev 2022; 42:1343-1371. [PMID: 34984701 PMCID: PMC9306699 DOI: 10.1002/med.21878] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 11/24/2021] [Accepted: 12/18/2021] [Indexed: 12/26/2022]
Abstract
A recent International Diabetes Federation report suggests that more than 463 million people between 20 and 79 years have diabetes. Of the 20 million women affected by hyperglycemia during pregnancy, 84% have gestational diabetes. In addition, more than 1.1 million children or adolescents are affected by type 1 diabetes. Factors contributing to the increase in diabetes prevalence are complex and include contributions from genetic, environmental, and epigenetic factors. However, molecular regulatory mechanisms influencing the progression of an individual towards increased susceptibility to metabolic diseases such as diabetes are not fully understood. Recent studies suggest that the pathogenesis of diabetes involves epigenetic changes, resulting in a persistently dysregulated metabolic phenotype. This review summarizes the role of epigenetic mechanisms, mainly DNA methylation and histone modifications, in the development of the pancreas, their contribution to the development of diabetes, and the potential employment of epigenetic modulators in diabetes treatment.
Collapse
Affiliation(s)
- Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Challagandla Anil Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Mohammed Z Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE.,Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| |
Collapse
|
12
|
Padilla-Martinez F, Wojciechowska G, Szczerbinski L, Kretowski A. Circulating Nucleic Acid-Based Biomarkers of Type 2 Diabetes. Int J Mol Sci 2021; 23:ijms23010295. [PMID: 35008723 PMCID: PMC8745431 DOI: 10.3390/ijms23010295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/25/2021] [Accepted: 12/26/2021] [Indexed: 11/23/2022] Open
Abstract
Type 2 diabetes (T2D) is a deficiency in how the body regulates glucose. Uncontrolled T2D will result in chronic high blood sugar levels, eventually resulting in T2D complications. These complications, such as kidney, eye, and nerve damage, are even harder to treat. Identifying individuals at high risk of developing T2D and its complications is essential for early prevention and treatment. Numerous studies have been done to identify biomarkers for T2D diagnosis and prognosis. This review focuses on recent T2D biomarker studies based on circulating nucleic acids using different omics technologies: genomics, transcriptomics, and epigenomics. Omics studies have profiled biomarker candidates from blood, urine, and other non-invasive samples. Despite methodological differences, several candidate biomarkers were reported for the risk and diagnosis of T2D, the prognosis of T2D complications, and pharmacodynamics of T2D treatments. Future studies should be done to validate the findings in larger samples and blood-based biomarkers in non-invasive samples to support the realization of precision medicine for T2D.
Collapse
Affiliation(s)
- Felipe Padilla-Martinez
- Clinical Research Centre, Medical University of Bialystok, 15276 Białystok, Poland; (F.P.-M.); (L.S.); (A.K.)
| | - Gladys Wojciechowska
- Clinical Research Centre, Medical University of Bialystok, 15276 Białystok, Poland; (F.P.-M.); (L.S.); (A.K.)
- Correspondence:
| | - Lukasz Szczerbinski
- Clinical Research Centre, Medical University of Bialystok, 15276 Białystok, Poland; (F.P.-M.); (L.S.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15276 Białystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, 15276 Białystok, Poland; (F.P.-M.); (L.S.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15276 Białystok, Poland
| |
Collapse
|
13
|
Tan Y, Cao H, Li Q, Sun J. The role of transcription factor Ap1 in the activation of the Nrf2/ARE pathway through TET1 in diabetic nephropathy. Cell Biol Int 2021; 45:1654-1665. [PMID: 33760331 DOI: 10.1002/cbin.11599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/01/2021] [Accepted: 03/21/2021] [Indexed: 02/06/2023]
Abstract
TET1 mediates demethylation in tumors, but its role in diabetic nephropathy (DN), a prevalent diabetic complication, is unclear. We attempted to probe the possible mechanism of TET1 in DN. A DN rat model was established and verified by marker detection and histopathological observation. The in vitro model was established on human mesangial cells (HMCs) induced by high glucose (HG), and verified by evaluation of fibrosis and inflammation. The differentially expressed mRNA was screened out by microarray analysis. The most differentially expressed mRNA (TET1) was reduced in DN rats and HG-HMCs. The upstream and downstream factors of TET1 were verified, and their roles in DN were analyzed by gain- and loss-function assays. TET1 was decreased in DN rats and HG-HMCs. High expression of TET1 decreased biochemical indexes and renal injury of DN rats and hampered the activity, fibrosis, and inflammation of HG-HMCs. Ap1 lowered TET1 expression, and enhanced inflammation in HG-HMCs, and accentuated renal injury in DN rats. TET1 overexpression inhibited the effect of Ap1 on DN. TET1 promoted the transcription of Nrf2. The Ap1/TET1 axis mediated the Nrf2/ARE pathway activity. Overall, TET1 overexpression weakened the inhibitory effect of Ap1 on the Nrf2/ARE pathway, thus alleviating inflammation and renal injury in DN.
Collapse
Affiliation(s)
- Yongshun Tan
- Department of Nephrology, Jinan City People's Hospital, Jinan, Shandong, China
| | - Huaimin Cao
- Department of Endocrinology, Gaotang County People's Hospital, Liaocheng, Shandong, China
| | - Qingfei Li
- Department of Endocrinology, Linyi People's Hospital, Dezhou, Shandong, China
| | - Jianjun Sun
- Department 1 of Nephrology, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
14
|
Seddon AR, Liau Y, Pace PE, Miller AL, Das AB, Kennedy MA, Hampton MB, Stevens AJ. Genome-wide impact of hydrogen peroxide on maintenance DNA methylation in replicating cells. Epigenetics Chromatin 2021; 14:17. [PMID: 33761969 PMCID: PMC7992848 DOI: 10.1186/s13072-021-00388-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/01/2021] [Indexed: 12/22/2022] Open
Abstract
Background Environmental factors, such as oxidative stress, have the potential to modify the epigenetic landscape of cells. We have previously shown that DNA methyltransferase (DNMT) activity can be inhibited by sublethal doses of hydrogen peroxide (H2O2). However, site-specific changes in DNA methylation and the reversibility of any changes have not been explored. Using bead chip array technology, differential methylation was assessed in Jurkat T-lymphoma cells following exposure to H2O2. Results Sublethal H2O2 exposure was associated with an initial genome-wide decrease in DNA methylation in replicating cells, which was largely corrected 72 h later. However, some alterations were conserved through subsequent cycles of cell division. Significant changes to the variability of DNA methylation were also observed both globally and at the site-specific level. Conclusions This research indicates that increased exposure to H2O2 can result in long-term alterations to DNA methylation patterns, providing a mechanism for environmental factors to have prolonged impact on gene expression. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00388-6.
Collapse
Affiliation(s)
- Annika R Seddon
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand.
| | - Yusmiati Liau
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | - Paul E Pace
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | - Allison L Miller
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | - Andrew B Das
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | - Martin A Kennedy
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | - Mark B Hampton
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand
| | - Aaron J Stevens
- Department of Pathology and Biomedical Science, University of Otago, PO Box 4345, Christchurch, 8140, New Zealand.
| |
Collapse
|
15
|
Mashal S, Khanfar M, Al-Khalayfa S, Srour L, Mustafa L, Hakooz NM, Zayed AA, Khader YS, Azab B. SLC30A8 gene polymorphism rs13266634 associated with increased risk for developing type 2 diabetes mellitus in Jordanian population. Gene 2020; 768:145279. [PMID: 33161057 DOI: 10.1016/j.gene.2020.145279] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/08/2020] [Accepted: 10/23/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Several genome-wide association studies (GWAS) have identified the single nucleotide polymorphism (SNP) rs13266634 in the Solute carrier family 30 member 8 (SLC30A8) gene as a risk factor to type 2 diabetes mellitus (T2DM). Nevertheless, other studies reported controversial findings of no significant association between the rs13266634 with T2DM. In this study, we aimed to investigate the association of this SNP with T2DM among Jordanian population in addition to define its corresponding allelic and genotypic frequencies. METHOD This case-control study enrolled 358 T2DM patients and 326 healthy controls who fulfilled the inclusion criteria. Blood samples were collected from all participants and were used for the rs13266634 SNP genotyping by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique. RESULTS We demonstrated a significant association between the C/T rs13266634 SNP and T2DM among Jordanian population. A significant difference was found between the cases and controls regarding the allelic (P = 0.003) distribution. Compared to people having T allele, those with C allele had higher risk of T2DM (OR = 1.47 ; 95% CI: 1.14 - 1.89; P = 0.003). Having a CC genotype versus TT genotype was significantly associated with increased risk to T2DM (OR = 2.44; 95% CI: 1.16 - 5.12; P = 0.019) after adjusting for age, gender, and BMI. Under the recessive model, subjects with CC genotype were more likely to have T2DM compared to those with CT or TT genotypes, (OR = 1.64; 95% CI: 1.18 - 2.26; P = 0.003) after adjusting for age, gender and BMI. CONCLUSION The rs13266634 SNP is significantly associated with T2DM susceptibility among Jordanian Population.
Collapse
Affiliation(s)
- Safaa Mashal
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, University of Jordan, PO Box: 13617, Queen Rania St., Amman 11942, Jordan
| | - Mariam Khanfar
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, P.O.Box 3030, Irbid 22110, Jordan
| | - Sawsan Al-Khalayfa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, University of Jordan, PO Box: 13617, Queen Rania St., Amman 11942, Jordan
| | - Luma Srour
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, University of Jordan, PO Box: 13617, Queen Rania St., Amman 11942, Jordan
| | - Lina Mustafa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, University of Jordan, PO Box: 13617, Queen Rania St., Amman 11942, Jordan
| | - Nancy M Hakooz
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, University of Jordan, PO Box: 13617, Queen Rania St., Amman 11942, Jordan
| | - Ayman A Zayed
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, School of Medicine, The University of Jordan, Jordan University Hospital, PO Box: 13617, Queen Rania St., Amman 11942, Jordan
| | - Yousef S Khader
- Department of Community Medicine, Public Health and Family Medicine, Faculty of Medicine, Jordan University of Science and Technology, P.O.Box 3030, Irbid 22110, Jordan
| | - Bilal Azab
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, University of Jordan, PO Box: 13617, Queen Rania St., Amman 11942, Jordan; Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, United States.
| |
Collapse
|
16
|
Zhang X, Guan T, Yang B, Gu HF, Chi Z. Effects of ZnT8 on epithelial-to-mesenchymal transition and tubulointerstitial fibrosis in diabetic kidney disease. Cell Death Dis 2020; 11:544. [PMID: 32681069 PMCID: PMC7367835 DOI: 10.1038/s41419-020-2731-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/21/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022]
Abstract
Zinc transporter 8 (ZnT8) transports zinc ions for crystallization and storage of insulin in pancreatic beta-cells and ZnT8 dysfunction is involved in pathogenesis of diabetes. The current study aimed to investigate whether ZnT8 has effects in pathophysiology of diabetic kidney disease (DKD) by using animal models for diabetes, including STZ-induced diabetic, db/db, ZnT8-KO, ZnT8-KO-STZ and ZnT8-KO-db/db mice. Results demonstrated that urine albumin to creatinine ratio and epithelial-to-mesenchymal transition (EMT) were increased in kidneys of ZnT8-KO-STZ and ZnT8-KO-db/db mice compared with C57BL/6 J and ZnT8-KO mice, while serum TGF-β1, IL-6, and TNF-α levels were elevated in parallel. In kidneys of mice intercrossed between ZnT8-KO and STZ-induced diabetic or db/db mice, these three inflammatory factors, ACR and EMT were also found to be increased compared with C57BL/6J, db/db and ZnT8-KO mice. Furthermore, ZnT8 up-regulation by hZnT8-EGFP reduced the levels of high glucose (HG)-induced EMT and inflammatory factors in normal rat kidney tubular epithelial cell (NRK-52E cells). Expression of phosphorylated Smad2/Smad3 was up-regulated after HG stimulation and further enhanced by ZnT8 siRNA but down-regulated after hZnT8-EGFP gene transfection. The current study thus provides the first evidence that ZnT8 protects against EMT-tubulointerstitial fibrosis though the restrain of TGF-β1/Smads signaling activation in DKD.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Nephrology, Second People's Hospital, The First Affiliated Hospital of Shenzhen University, 518000, Shenzhen, Guangdong Province, P.R. China.,Department of Pathophysiology, China Medical University, 110001, Shenyang, Liaoning Province, P.R. China
| | - Tingwen Guan
- Department of Pathophysiology, China Medical University, 110001, Shenyang, Liaoning Province, P.R. China
| | - Boxuan Yang
- Department of Pathophysiology, China Medical University, 110001, Shenyang, Liaoning Province, P.R. China
| | - Harvest F Gu
- Center for Pathophysiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, Jiangsu Province, P.R. China.
| | - Zhihong Chi
- Department of Pathophysiology, China Medical University, 110001, Shenyang, Liaoning Province, P.R. China.
| |
Collapse
|
17
|
Ahmed SAH, Ansari SA, Mensah-Brown EPK, Emerald BS. The role of DNA methylation in the pathogenesis of type 2 diabetes mellitus. Clin Epigenetics 2020; 12:104. [PMID: 32653024 PMCID: PMC7353744 DOI: 10.1186/s13148-020-00896-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic condition characterised by β cell dysfunction and persistent hyperglycaemia. The disorder can be due to the absence of adequate pancreatic insulin production or a weak cellular response to insulin signalling. Among the three types of DM, namely, type 1 DM (T1DM), type 2 DM (T2DM), and gestational DM (GDM); T2DM accounts for almost 90% of diabetes cases worldwide. Epigenetic traits are stably heritable phenotypes that result from certain changes that affect gene function without altering the gene sequence. While epigenetic traits are considered reversible modifications, they can be inherited mitotically and meiotically. In addition, epigenetic traits can randomly arise in response to environmental factors or certain genetic mutations or lesions, such as those affecting the enzymes that catalyse the epigenetic modification. In this review, we focus on the role of DNA methylation, a type of epigenetic modification, in the pathogenesis of T2DM.
Collapse
Affiliation(s)
- Sanabil Ali Hassan Ahmed
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Eric P K Mensah-Brown
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 17666, Al Ain, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
18
|
Zhang Y, Zhang Y, Sun K, Meng Z, Chen L. The SLC transporter in nutrient and metabolic sensing, regulation, and drug development. J Mol Cell Biol 2020; 11:1-13. [PMID: 30239845 PMCID: PMC6359923 DOI: 10.1093/jmcb/mjy052] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
The prevalence of metabolic diseases is growing worldwide. Accumulating evidence suggests that solute carrier (SLC) transporters contribute to the etiology of various metabolic diseases. Consistent with metabolic characteristics, the top five organs in which SLC transporters are highly expressed are the kidney, brain, liver, gut, and heart. We aim to understand the molecular mechanisms of important SLC transporter-mediated physiological processes and their potentials as drug targets. SLC transporters serve as ‘metabolic gate’ of cells and mediate the transport of a wide range of essential nutrients and metabolites such as glucose, amino acids, vitamins, neurotransmitters, and inorganic/metal ions. Gene-modified animal models have demonstrated that SLC transporters participate in many important physiological functions including nutrient supply, metabolic transformation, energy homeostasis, tissue development, oxidative stress, host defense, and neurological regulation. Furthermore, the human genomic studies have identified that SLC transporters are susceptible or causative genes in various diseases like cancer, metabolic disease, cardiovascular disease, immunological disorders, and neurological dysfunction. Importantly, a number of SLC transporters have been successfully targeted for drug developments. This review will focus on the current understanding of SLCs in regulating physiology, nutrient sensing and uptake, and risk of diseases.
Collapse
Affiliation(s)
- Yong Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.,Advanced Biotechnology and Application Research Center, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yuping Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Kun Sun
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Ziyi Meng
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Ligong Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
19
|
Bhowmick A, Sarkar P, Baruah MP, Bodhini D, Radha V, Mohan V, Banu S. Association of SLC30A8, CDKAL1, TCF7L2 and HHEX Gene Polymorphisms with Type 2 Diabetes in the Population of North East India. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720020036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
20
|
Salameh Y, Bejaoui Y, El Hajj N. DNA Methylation Biomarkers in Aging and Age-Related Diseases. Front Genet 2020; 11:171. [PMID: 32211026 PMCID: PMC7076122 DOI: 10.3389/fgene.2020.00171] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Recent research efforts provided compelling evidence of genome-wide DNA methylation alterations in aging and age-related disease. It is currently well established that DNA methylation biomarkers can determine biological age of any tissue across the entire human lifespan, even during development. There is growing evidence suggesting epigenetic age acceleration to be strongly linked to common diseases or occurring in response to various environmental factors. DNA methylation based clocks are proposed as biomarkers of early disease risk as well as predictors of life expectancy and mortality. In this review, we will summarize key advances in epigenetic clocks and their potential application in precision health. We will also provide an overview of progresses in epigenetic biomarker discovery in Alzheimer's, type 2 diabetes, and cardiovascular disease. Furthermore, we will highlight the importance of prospective study designs to identify and confirm epigenetic biomarkers of disease.
Collapse
Affiliation(s)
| | | | - Nady El Hajj
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
21
|
Domínguez-Cruz MG, Muñoz MDL, Totomoch-Serra A, García-Escalante MG, Burgueño J, Valadez-González N, Pinto-Escalante D, Díaz-Badillo A. Maya gene variants related to the risk of type 2 diabetes in a family-based association study. Gene 2020; 730:144259. [PMID: 31759989 DOI: 10.1016/j.gene.2019.144259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/01/2022]
Abstract
Mexican Maya populations have a notably high prevalence of type 2 diabetes (T2D) as a consequence of the interaction between environmental factors and a genetic component. To assess the impact of 24 single nucleotide variants (SNVs) located in 18 T2D risk genes, we conducted a family-based association evaluation in samples from Maya communities with a high incidence of the disease. A total of four hundred individuals were recruited from three Maya communities with a high T2D incidence. Family pedigrees (100) and 49 nuclear families were included. Genotyping was performed by allelic discrimination with TaqMan probes. This study also included the family-based association test (FBAT) statistic U to assess the genetic associations with T2D, and the multivariate statistical and haplotype analyses. A positive association with TD2 risk was found for WFS1 rs6446482 (p = 0.046, Z = 1.994) under an additive model, and SIRT1 rs7896005 (p = 0.038, Z = 2.073) under the dominant model. Multivariate model analysis, including T2D status, age, and body mass index (BMI), displayed significant covariance in PPARGC-1α rs8192678; SIRT1 rs7896005; TCF7L2 rs7903146 and rs122243326; UCP3 rs3781907; and HHEX rs1111875 with a P < 0.05. This study revealed an association of SIRT1 and WFS1 with T2D risk.
Collapse
Affiliation(s)
- Miriam G Domínguez-Cruz
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - María de Lourdes Muñoz
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico.
| | - Armando Totomoch-Serra
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico; PhD Program in Medical Sciences, Universidad de La Frontera, Chile
| | - María G García-Escalante
- Laboratorios de Genética y Hematología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Juan Burgueño
- Centro Internacional de Mejoramiento de Maíz y Trigo, El Batán, Texcoco, State of Mexico, Mexico
| | - Nina Valadez-González
- Laboratorios de Genética y Hematología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Doris Pinto-Escalante
- Laboratorios de Genética y Hematología, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Alvaro Díaz-Badillo
- Maestría en Salud Publica, Universidad México Americana del Norte, Reynosa, Tamaulipas, Mexico; Department of Epidemiology, Human Genetics & Environmental Sciences, The University of Texas Health Science Center at Houston, Brownville, TX, USA
| |
Collapse
|
22
|
Contemplating the role of genetic variants of HHEX, CDKAL1, WFS1 and SLC30A8 genes of TYPE-2 diabetes in Asians ethnic groups. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100465] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Juvinao-Quintero DL, Hivert MF, Sharp GC, Relton CL, Elliott HR. DNA Methylation and Type 2 Diabetes: the Use of Mendelian Randomization to Assess Causality. CURRENT GENETIC MEDICINE REPORTS 2019; 7:191-207. [PMID: 32274260 PMCID: PMC7145450 DOI: 10.1007/s40142-019-00176-5] [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] [Indexed: 12/20/2022]
Abstract
Purpose of Review This review summarises recent advances in the field of epigenetics in order to understand the aetiology of type 2 diabetes (T2D). Recent Findings DNA methylation at a number of loci has been shown to be robustly associated with T2D, including TXNIP, ABCG1, CPT1A, and SREBF1. However, due to the cross-sectional nature of many epidemiological studies and predominant analysis in samples derived from blood rather than disease relevant tissues, inferring causality is difficult. We therefore outline the use of Mendelian randomisation (MR) as one method able to assess causality in epigenetic studies of T2D. Summary Epidemiological studies have been fruitful in identifying epigenetic markers of T2D. Triangulation of evidence including utilisation of MR is essential to delineate causal from non-causal biomarkers of disease. Understanding the causality of epigenetic markers in T2D more fully will aid prioritisation of CpG sites as early biomarkers to detect disease or in drug development to target epigenetic mechanisms in order to treat patients.
Collapse
Affiliation(s)
- Diana L Juvinao-Quintero
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, USA
| | - Gemma C Sharp
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Bristol NIHR Biomedical Research Centre, Bristol, UK
| | - Hannah R Elliott
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| |
Collapse
|
24
|
Gu HF. Genetic and Epigenetic Studies in Diabetic Kidney Disease. Front Genet 2019; 10:507. [PMID: 31231424 PMCID: PMC6566106 DOI: 10.3389/fgene.2019.00507] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/08/2019] [Indexed: 01/19/2023] Open
Abstract
Chronic kidney disease is a worldwide health crisis, while diabetic kidney disease (DKD) has become the leading cause of end-stage renal disease (ESRD). DKD is a microvascular complication and occurs in 30–40% of diabetes patients. Epidemiological investigations and clinical observations on the familial clustering and heritability in DKD have highlighted an underlying genetic susceptibility. Furthermore, DKD is a progressive and long-term diabetic complication, in which epigenetic effects and environmental factors interact with an individual’s genetic background. In recent years, researchers have undertaken genetic and epigenetic studies of DKD in order to better understand its molecular mechanisms. In this review, clinical material, research approaches and experimental designs that have been used for genetic and epigenetic studies of DKD are described. Current information from genetic and epigenetic studies of DKD and ESRD in patients with diabetes, including the approaches of genome-wide association study (GWAS) or epigenome-wide association study (EWAS) and candidate gene association analyses, are summarized. Further investigation of molecular defects in DKD with new approaches such as next generation sequencing analysis and phenome-wide association study (PheWAS) is also discussed.
Collapse
Affiliation(s)
- Harvest F Gu
- Center for Pathophysiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
25
|
Huang Q, Du J, Merriman C, Gong Z. Genetic, Functional, and Immunological Study of ZnT8 in Diabetes. Int J Endocrinol 2019; 2019:1524905. [PMID: 30936916 PMCID: PMC6413397 DOI: 10.1155/2019/1524905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/14/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022] Open
Abstract
Zinc level in the body is finely regulated to maintain cellular function. Dysregulation of zinc metabolism may induce a variety of diseases, e.g., diabetes. Zinc participates in insulin synthesis, storage, and secretion by functioning as a "cellular second messenger" in the insulin signaling pathway and glucose homeostasis. The highest zinc concentration is in the pancreas islets. Zinc accumulation in cell granules is manipulated by ZnT8, a zinc transporter expressed predominately in pancreatic α and β cells. A common ZnT8 gene (SLC30A8) polymorphism increases the risk of type 2 diabetes mellitus (T2DM), and rare mutations may present protective effects. In type 1 diabetes mellitus (T1DM), autoantibodies show specificity for binding two variants of ZnT8 (R or W at amino acid 325) dictated by a polymorphism in SLC30A8. In this review, we summarize the structure, feature, functions, and polymorphisms of ZnT8 along with its association with diabetes and explore future study directions.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Pharmacy, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jie Du
- Department of Pharmacy, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Chengfeng Merriman
- Department of Physiology, Johns Hopkins School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Zhicheng Gong
- Department of Pharmacy, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
26
|
Zhang X, Guan T, Yang B, Chi Z, Wang ZY, Gu HF. A novel role for zinc transporter 8 in the facilitation of zinc accumulation and regulation of testosterone synthesis in Leydig cells of human and mouse testicles. Metabolism 2018; 88:40-50. [PMID: 30236453 DOI: 10.1016/j.metabol.2018.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/05/2018] [Accepted: 09/11/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Zinc is intimately involved in testosterone production. Zinc transporter 8 (ZnT8) is found to be localized in insulin secretory granules as a β-cell specific Zn transporter. The effect of ZnT8 and related zinc accumulation in steroidogenesis, however, is still unknown. The present study aimed to explore whether ZnT8 plays a role in the facilitation of zinc accumulation and regulation of testosterone synthesis in testicles. METHODS Leydig cells were isolated from the testicles of human, CD-1 suckling and ZnT8-KO mice. Zn accumulation in mitochondria was induced by hCG stimulation. Transfection of hZnT8-EGFP and RNA interfere of mZnT8 were done in MLTC-1 cells. ZnT8 expression and its co-localization with steroidogenic acute regulatory (StAR) protein were analyzed with RT-PCR, Western blot and dual-fluorescent staining protocols. Serum testosterone levels in mice were determined with chemiluminescent enzyme immunoassay. RESULTS ZnT8 was found to be presented in Leydig cells and up-regulated in suckling mouse Leydig cells and MLTC-1 cells after hCG administration, by which zinc accumulation occurred in mitochondria. ZnT8 gene silencing or knockout inhibited stimulated progesterone and testosterone production, reduced stimulated zinc accumulation and down-regulated phosphorylated steroidogenic acute regulatory (StAR) expression in Leydig cells. Furthermore, an inhibitor (H89) of PKA blocked hCG-stimulated progesterone caused by ZnT8 over-expression and zinc treatment. CONCLUSION The present study provided the first evidence that ZnT8 transports Zn into Leydig cell mitochondria with gonadotropin stimulation and suggests that ZnT8 may play a role in testosterone production via the PKA signaling pathway.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Nephrology, The second People's Hospital, Shenzhen, The first Affiliated Hospital of Shenzhen University, Guangdong 518000, PR China; Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Tingwen Guan
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Boxuan Yang
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Zhihong Chi
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Zhan-You Wang
- College of Life and Health Sciences, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Harvest F Gu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
27
|
Rodriguez H, El-Osta A. Epigenetic Contribution to the Development and Progression of Vascular Diabetic Complications. Antioxid Redox Signal 2018; 29:1074-1091. [PMID: 29304555 DOI: 10.1089/ars.2017.7347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SIGNIFICANCE The number of people suffering from diabetes worldwide is steadily rising. Complications from diabetes, including cardiovascular and renal disease, contribute to the high morbidity and mortality associated with this disease. Recent Advances: Hyperglycemia promotes tissue damage through diverse mechanisms involving increased production of reactive oxygen species. Increased oxidative stress drives changes in chromatin structure that mediate gene expression changes leading to the upregulation of proinflammatory and profibrotic mediators. The epigenetic contribution to diabetes-induced changes in gene expression is increasingly recognized as a key factor in the development and progression of vascular diabetic complications. CRITICAL ISSUES The mechanisms through which stimuli from the diabetic milieu promote epigenetic changes remain poorly understood. In addition, glycemic control constitutes an important factor influencing epigenetic states in diabetes, and the phenomenon of hyperglycemic memory warrants further research. FUTURE DIRECTIONS Knowledge of the molecular mechanisms underlying epigenetic changes in diabetes may allow the design of novel therapeutic strategies to reduce the burden of diabetic complications. Furthermore, certain epigenetic markers are detected early during the onset of diabetes and its complications and may prove useful as biomarkers for disease risk prediction.
Collapse
Affiliation(s)
- Hanah Rodriguez
- 1 Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University , Melbourne, Australia
| | - Assam El-Osta
- 1 Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University , Melbourne, Australia .,2 Department of Pathology, University of Melbourne , Melbourne, Australia .,3 Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong , Hong Kong SAR, China
| |
Collapse
|
28
|
Lin Z, Wang Y, Zhang B, Jin Z. Association of type 2 diabetes susceptible genes GCKR, SLC30A8, and FTO polymorphisms with gestational diabetes mellitus risk: a meta-analysis. Endocrine 2018; 62:34-45. [PMID: 30091126 DOI: 10.1007/s12020-018-1651-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022]
Abstract
PURPOSE Current studies have detected the correlation of polymorphisms in type 2 diabetes susceptible genes GCKR, SLC30A8 and FTO with gestational diabetes mellitus (GDM) risk. However, findings of these studies were incongruous. Hence, we performed an integrated review and meta-analysis for the researches regarding the association of single nucleotide polymorphisms (SNPs) in GCKR, SLC30A8 and FTO genes and GDM risk. METHODS Eligible publications were selected on the basis of several inclusion and exclusion criteria. Correlation between each SNP and GDM risk was estimated by computing odds ratios (ORs) with 95% confidence intervals (95%CIs). RESULTS Consequently, 19 case-control studies (from 16 citations) including 3636 GDM cases and 7229 GDM-free controls were participated in a meta-analysis of seven prevalent SNPs (GCKR rs1260326 and rs780094; SLC30A8 rs13266634 and rs11558471; FTO rs8050136, rs1421085 and rs9939609). Our results demonstrated that the rs780094, rs13266634 and rs9939609 SNPs were significantly associated with GDM risk. In stratified analysis, correlations of rs780094 and rs13266634 SNPs could be observed in Asian and Caucasian subgroups. Moreover, association between rs9939609 SNP and GDM risk was detected in Caucasian subgroup. CONCLUSIONS The GCKR rs780094, SLC30A8 rs13266634 and FTO rs9939609 SNPs were demonstrated to be the potential biomarkers for GDM risk prediction.
Collapse
Affiliation(s)
- Ziqi Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110004, Liaoning, China
| | - Yue Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110004, Liaoning, China
| | - Bao Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110004, Liaoning, China
| | - Zhen Jin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
29
|
Chen Y, Hong T, Wang S, Mo J, Tian T, Zhou X. Epigenetic modification of nucleic acids: from basic studies to medical applications. Chem Soc Rev 2018; 46:2844-2872. [PMID: 28352906 DOI: 10.1039/c6cs00599c] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The epigenetic modification of nucleic acids represents one of the most significant areas of study in the field of nucleic acids because it makes gene regulation more complex and heredity more complicated, thus indicating its profound impact on aspects of heredity, growth, and diseases. The recent characterization of epigenetic modifications of DNA and RNA using chemical labelling strategies has promoted the discovery of these modifications, and the newly developed single-base or single-cell resolution mapping strategies have enabled large-scale epigenetic studies in eukaryotes. Due to these technological breakthroughs, several new epigenetic marks have been discovered that have greatly extended the scope and impact of epigenetic modifications in nucleic acids over the past few years. Because epigenetics is reversible and susceptible to environmental factors, it could potentially be a promising direction for clinical medicine research. In this review, we have comprehensively discussed how these epigenetic marks are involved in disease, including the pathogenesis, prevention, diagnosis and treatment of disease. These findings have revealed that the epigenetic modification of nucleic acids has considerable significance in various areas from methodology to clinical medicine and even in biomedical applications.
Collapse
Affiliation(s)
- Yuqi Chen
- College of Chemistry and Molecular Sciences, Institute of Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Hubei, Wuhan 430072, P. R. China.
| | | | | | | | | | | |
Collapse
|
30
|
Willmer T, Johnson R, Louw J, Pheiffer C. Blood-Based DNA Methylation Biomarkers for Type 2 Diabetes: Potential for Clinical Applications. Front Endocrinol (Lausanne) 2018; 9:744. [PMID: 30564199 PMCID: PMC6288427 DOI: 10.3389/fendo.2018.00744] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022] Open
Abstract
Type 2 diabetes (T2D) is a leading cause of death and disability worldwide. It is a chronic metabolic disorder that develops due to an interplay of genetic, lifestyle, and environmental factors. The biological onset of the disease occurs long before clinical symptoms develop, thus the search for early diagnostic and prognostic biomarkers, which could facilitate intervention strategies to prevent or delay disease progression, has increased considerably in recent years. Epigenetic modifications represent important links between genetic, environmental and lifestyle cues and increasing evidence implicate altered epigenetic marks such as DNA methylation, the most characterized and widely studied epigenetic mechanism, in the pathogenesis of T2D. This review provides an update of the current status of DNA methylation as a biomarker for T2D. Four databases, Scopus, Pubmed, Cochrane Central, and Google Scholar were searched for studies investigating DNA methylation in blood. Thirty-seven studies were identified, and are summarized with respect to population characteristics, biological source, and method of DNA methylation quantification (global, candidate gene or genome-wide). We highlight that differential methylation of the TCF7L2, KCNQ1, ABCG1, TXNIP, PHOSPHO1, SREBF1, SLC30A8, and FTO genes in blood are reproducibly associated with T2D in different population groups. These genes should be prioritized and replicated in longitudinal studies across more populations in future studies. Finally, we discuss the limitations faced by DNA methylation studies, which include including interpatient variability, cellular heterogeneity, and lack of accounting for study confounders. These limitations and challenges must be overcome before the implementation of blood-based DNA methylation biomarkers into a clinical setting. We emphasize the need for longitudinal prospective studies to support the robustness of the current findings of this review.
Collapse
Affiliation(s)
- Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- *Correspondence: Tarryn Willmer
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
31
|
Beaver LM, Nkrumah-Elie YM, Truong L, Barton CL, Knecht AL, Gonnerman GD, Wong CP, Tanguay RL, Ho E. Adverse effects of parental zinc deficiency on metal homeostasis and embryonic development in a zebrafish model. J Nutr Biochem 2017; 43:78-87. [PMID: 28268202 PMCID: PMC5406264 DOI: 10.1016/j.jnutbio.2017.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 12/09/2016] [Accepted: 02/08/2017] [Indexed: 10/20/2022]
Abstract
The high prevalence of zinc deficiency is a global public health concern, and suboptimal maternal zinc consumption has been associated with adverse effects ranging from impaired glucose tolerance to low birthweights. The mechanisms that contribute to altered development and poor health in zinc deficient offspring are not completely understood. To address this gap, we utilized the Danio rerio model and investigated the impact of dietary zinc deficiency on adults and their developing progeny. Zinc deficient adult fish were significantly smaller in size, and had decreases in learning and fitness. We hypothesized that parental zinc deficiency would have an impact on their offspring's mineral homeostasis and embryonic development. Results from mineral analysis showed that parental zinc deficiency caused their progeny to be zinc deficient. Furthermore, parental dietary zinc deficiency had adverse consequences for their offspring including a significant increase in mortality and decreased physical activity. Zinc deficient embryos had altered expression of genes that regulate metal homeostasis including several zinc transporters (ZnT8, ZnT9) and the metal-regulatory transcription factor 1 (MTF-1). Zinc deficiency was also associated with decreased expression of genes related to diabetes and pancreatic development in the embryo (Insa, Pax4, Pdx1). Decreased expression of DNA methyltransferases (Dnmt4, Dnmt6) was also found in zinc deficient offspring, which suggests that zinc deficiency in parents may cause altered epigenetic profiles for their progeny. These data should inform future studies regarding zinc deficiency and pregnancy and suggest that supplementation of zinc deficient mothers prior to pregnancy may be beneficial.
Collapse
Affiliation(s)
- Laura M Beaver
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331, United States; Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331, United States.
| | - Yasmeen M Nkrumah-Elie
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331, United States; Department of Environmental and Molecular Toxicology, Oregon State University, Sinnhuber Aquatic Research Laboratory, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States.
| | - Lisa Truong
- Department of Environmental and Molecular Toxicology, Oregon State University, Sinnhuber Aquatic Research Laboratory, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States.
| | - Carrie L Barton
- Department of Environmental and Molecular Toxicology, Oregon State University, Sinnhuber Aquatic Research Laboratory, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States.
| | - Andrea L Knecht
- Department of Environmental and Molecular Toxicology, Oregon State University, Sinnhuber Aquatic Research Laboratory, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States.
| | - Greg D Gonnerman
- Department of Environmental and Molecular Toxicology, Oregon State University, Sinnhuber Aquatic Research Laboratory, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States.
| | - Carmen P Wong
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331, United States; Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331, United States.
| | - Robert L Tanguay
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331, United States; Department of Environmental and Molecular Toxicology, Oregon State University, Sinnhuber Aquatic Research Laboratory, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States; Center for Genome Research and Biocomputing, Oregon State University, 3021 Agriculture and Life Sciences Building, Corvallis, OR 97331, United States.
| | - Emily Ho
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331, United States; Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331, United States; The Environmental Health Sciences Center, Oregon State University, 1011 Agriculture & Life Sciences Building, Corvallis, Oregon 97331, United States; Center for Genome Research and Biocomputing, Oregon State University, 3021 Agriculture and Life Sciences Building, Corvallis, OR 97331, United States; Moore Family Center for Whole Grain Foods, Nutrition and Preventive Health, Oregon State University, 212 Milam Hall, Corvallis, OR 97331, United States.
| |
Collapse
|
32
|
Liu Z, Chen T, Sun W, Yuan Z, Yu M, Chen G, Guo W, Xiao J, Tian W. DNA Demethylation Rescues the Impaired Osteogenic Differentiation Ability of Human Periodontal Ligament Stem Cells in High Glucose. Sci Rep 2016; 6:27447. [PMID: 27273319 PMCID: PMC4897703 DOI: 10.1038/srep27447] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/16/2016] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus, characterized by abnormally high blood glucose levels, gives rise to impaired bone remodeling. In response to high glucose (HG), the attenuated osteogenic differentiation capacity of human periodontal ligament stem cells (hPDLSCs) is associated with the loss of alveolar bone. Recently, DNA methylation was reported to affect osteogenic differentiation of stem cells in pathological states. However, the intrinsic mechanism linking DNA methylation to osteogenic differentiation ability in the presence of HG is still unclear. In this study, we found that diabetic rats with increased DNA methylation levels in periodontal ligaments exhibited reduced bone mass and density. In vitro application of 5-aza-2′-deoxycytidine (5-aza-dC), a DNA methyltransferase inhibitor, to decrease DNA methylation levels in hPDLSCs, rescued the osteogenic differentiation capacity of hPDLSCs under HG conditions. Moreover, we demonstrated that the canonical Wnt signaling pathway was activated during this process and, under HG circumstances, the 5-aza-dC-rescued osteogenic differentiation capacity was blocked by Dickkopf-1, an effective antagonist of the canonical Wnt signaling pathway. Taken together, these results demonstrate for the first time that suppression of DNA methylation is able to facilitate the osteogenic differentiation capacity of hPDLSCs exposed to HG, through activation of the canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Southwest Medical University, Luzhou 646000, P.R. China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Tian Chen
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Wenhua Sun
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Zongyi Yuan
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Southwest Medical University, Luzhou 646000, P.R. China
| | - Mei Yu
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Guoqing Chen
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Weihua Guo
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| | - Jingang Xiao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Southwest Medical University, Luzhou 646000, P.R. China.,Orofacial Reconstruction and Regeneration Laboratory, Hospital of Stomatology, Southwest Medical University, Luzhou 646000, P.R. China
| | - Weidong Tian
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P.R. China.,Department of Oral and Maxillofacial Surgery, West China School of Stomatology, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|