Bioinformatics analysis of abnormal DNA methylation in muscle samples from monozygotic twins discordant for type 2 diabetes

  • Authors:
    • Fei Liu
    • Qianqian Sun
    • Lingxiao Wang
    • Shuangshuang Nie
    • Jun Li
  • View Affiliations

  • Published online on: March 6, 2015     https://doi.org/10.3892/mmr.2015.3452
  • Pages: 351-356
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to examine the changes in DNA methylation of gene promoters associated with type 2 diabetes (T2D). The DNA methylation profile dataset GSE38291 was downloaded from the Gene Expression Omnibus database. A paired t‑test was used to analyze differences in the DNA methylation of gene promoters between T2D and normal muscle samples. Gene Ontology (GO) enrichment analysis was performed using online tool, The Database for Annotation, Visualization and Integrated Discovery. Whole‑Genome rVISTA was used to analyze the enriched transcription factor (TF) binding sites upstream of the transcription start site in the differentially methylated genes. A total of 38 genes, including Sirtuin 1, N‑acetyltransferase 6, phospholipase A2 group XIIB and nuclear factor of activated T cells calcineurin‑dependent 1, were identified to be differentially methylated between these two groups. One GO term, DNA geometric change (GO:0032392), was significantly enriched (P<0.05) by the hyper‑methylated genes. In addition, the binding sites of one gene, zinc finger E‑box binding homeobox 1, and three TFs, methyl CpG binding protein 2, TFEB and TFAP4, were significantly enriched in the hyper‑ and hypo‑methylated genes, respectively. The resulting T2D‑associated genes and potential TFs provided a novel insight into the molecular mechanisms underlying the pathology of T2D. These genes may become promising target genes for the development of treatments for T2D.

Introduction

In recent decades, the incidence and prevalence of type 2 diabetes (T2D) has significantly increased worldwide, particularly among those aged between 35 and 40 years old (1). Long-term diabetes frequently induces vascular changes and dysfunction, and diabetic vascular complications are a major cause of morbidity and mortality amongst patients with diabetes (2).

Dysfunction of the β cells of the Islets of Langerhans and insulin-resistance are the main factors underlying the development of T2D (3). In addition, other factors, including inheritance, ethnicity and age, are all associated with the incidence of T2D (4). A previous study identified that the metabolism of glucose and fatty acids by skeletal muscle is impaired in T2D (5). Reduced glucose transport and phosphorylation, as well as reduced rates of glycogen synthesis are common manifestations of insulin-resistant glucose metabolism (6). However, abnormal fatty acid metabolism involves the increased accumulation of triglycerides and other lipids (7), as well as the dysregulation of lipid oxidation under fasting and insulin-stimulated conditions (8). Consequently, the dysfunction of metabolism in muscles may potentially indicate T2D.

DNA methylation is a key epigenetic modification of the genome, which is involved in numerous cellular processes, including cell differentiation and metabolism (9,10). Consistent with these significant roles, associations between aberrant DNA methylation and a growing number of human diseases have been identified (11). In addition, abnormal methylation of several genes, including cytochrome c oxidase polypeptide 7A1, nicotinamide adenine dinucleotide dehydrogenase (ubiquinone) 1 β subcomplex subunit 6, peroxisome proliferator-activated receptor γ coactivator (PGC) 1-α and PGC-1β, have been observed in tissues from the skeletal muscles of individuals with type 2 diabetes (1215).

The development of methylation chips has provided a more efficient approach for the detection of global changes in DNA methylation. Consequently, the current study utilized microarray data derived from the muscle samples of eleven monozygotic twin pairs, discordant for T2D, to detect the global changes in DNA methylation of gene promoters associated with T2D. Gene Ontology (GO) enrichment analysis and prediction analysis of binding sites for potential transcription factors (TFs) were performed in order to investigate the molecular mechanisms involved in T2D.

Materials and methods

Data acquisition

The DNA methylation dataset, GSE38291 (16), comprising data regarding 11 pairs of monozygotic twins discordant for type 2 diabetes, was downloaded from the Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/) database. Each pair of monozygotic twins included one individual with T2D and one normal individual (GSM938065-GSM938086). The Illumina HumanMethylation27 BeadChips (Illumina Inc., San Diego, CA, USA), containing a pair of methylated and unmethylated probes designed for each CpG site, were used to detect the DNA methylation in the gene promoter region.

Quantification of DNA methylation signals

The raw intensities of methylated and unmethylated probes for each sample were extracted based on the Illumina HumanMethylation27 BeadChip. The intensity data were then normalized using quantile normalization. The M-value of each CpG site was calculated as the log2 ratio of the normalized intensities of methylated probe versus unmethylated probe (17). The M-values of all probes in the promoter of a single gene were calculated and the mid-value was considered to be the methylation level of this gene promoter.

Analysis of differentially methylated genes

A paired samples t-test was employed to analyze the differentially methylated levels of gene promoter between T2D and normal muscle samples. P<0.05 and a fold change of >1.3 were considered to indicate the cutoff criteria.

GO enrichment analysis

GO enrichment analysis was performed on the differentially methylated genes for biological processes and cellular components using the online bioinformatics resource: The Database for Annotation, Visualization and Integrated Discovery version 6.7 (http://david.abcc.ncifcrf.gov/) (18). GO terms where P<0.1, were considered to indicate significantly enriched GO terms.

Screening for potential TFs

The Whole-Genome rVISTA tool (19) was used to analyze the enriched TF binding sites in the upstream region (200 bp, 500 bp and 1 kb) of transcription start sites (TSS) of the abnormal hyper- and hypo-methylated genes. The TFs with a P<0.01 were considered to be significant. Subsequently, the significant TFs obtained from the three regions were compared, and the most common TFs were considered as the final TFs of the target genes.

Results

Differentially methylated genes

A total of 38 genes were identified to possess differentially methylated regions between T2D and normal monozygotic twins. Specifically, 22 genes, including SIRT1 (Sirtuin1), NAT6 (N-acetyltransferase 6) and PLA2G12B (phospholipase A2 group XIIB), were hypermethylated in the T2D group; while 16 genes, including nuclear factor of activated T cells calcineurin-dependent 1 (NFATC1) and microtubule associated serine/threonine kinase 1, were hypo-methylated (Table I, Fig. 1). In addition, two TFs were identified in the hyper- and hypo- methylated genes, respectively, based on the public TF database (Table I) (20). These two TFs were Runt-related transcription factor 1 and NFATC1, which possessed significant hyper- and hypo-methylated regions in T2D muscles.

Table I

Significant hyper- and hypo-methylated genes in T2D.

Table I

Significant hyper- and hypo-methylated genes in T2D.

Gene Log2FCP-valueTranscription factor
Hypermethylation in T2D
 GPATC10.7700.006No
 SMCX0.6100.021No
 MUC130.5880.002No
 PMVK0.5690.042No
 RPL18A0.5030.019No
 IL20RA0.4860.025No
 NAT60.4810.006No
 OIT30.4720.011No
 ARHGAP270.4690.009No
 RUNX10.4680.015Yes
 PFS20.4660.038No
 BRUNOL50.4620.005No
 CIAPIN10.4430.012No
 SPCS20.4360.016No
 MICB0.4320.022No
 MGAT30.4310.039No
 HSPA140.4300.024No
 LOC1590900.4260.049No
 SIRT10.4190.011No
 C22orf90.4130.039No
 PLA2G12B0.4090.029No
 PPWD10.4010.047No
Hypomethylation in T2D
 RECQL4−0.4040.037No
 SYT12−0.4050.030No
 GTF3C1−0.4050.043No
 NT5DC3−0.4060.039No
 NFATC1−0.4060.002Yes
 SETMAR−0.4120.025No
 NOL7−0.4210.031No
 BICD2−0.4240.032No
 TCL6−0.4300.031No
 KCNE4−0.4310.045No
 DDOST−0.4480.004No
 MAST1−0.4500.005No
 TYR−0.4510.020No
 PURA−0.4680.038No
 GNPDA1−0.4710.022No
 NEDD1−0.4720.037No

[i] T2D, type 2 diabetes.

GO enrichment analysis

According to the GO enrichment analysis, hyper-methylated genes were enriched in three GO terms (P<0.1, Table II). The negative regulation of response to stimulus (GO: 0048585), which was associated with a process that stops, prevents or reduces the frequency, rate or extent of a response to a stimulus, was enriched by three genes, which may be significant in T2D pathology. The terms cell death (GO: 0008219) and death (GO: 0016265), refer to the biological processes that result in permanent cessation of all vital functions of a cell. By contrast, the hypomethylated genes were significantly enriched in three classes of GO terms. DNA geometric change (GO: 0032392) is a term regarding the process in which a transformation is induced in the geometry of a DNA double helix, resulting in a change in twist and/or writhe. There were three genes enriched for DNA metabolic process (GO: 0006259), which is associated with cellular metabolic processes involving deoxyribonucleic acid. Finally, two genes were highly enriched for DNA recombination (GO: 0006310), involving the process in which a new genotype is formed via a reassortment of genes resulting in novel gene combinations.

Table II

Significantly enriched GO terms of differentially methylated genes in T2D.

Table II

Significantly enriched GO terms of differentially methylated genes in T2D.

Methylation statusGO termGene countsP-valueGenes
Hypermethylated genes in T2DGO: 0048585-negative regulation of response to stimulus20.072MICB, SIRT1
GO: 0008219-cell death30.096MICB, CIAPIN1, SIRT1
GO: 0016265-death30.097MICB, CIAPIN1, SIRT1
Hypomethylated genes in T2DGO: 0032392-DNA geometric change20.015RECQL4, PURA
GO: 0006259-DNA metabolic process30.061RECQL4, SETMAR, PURA
GO: 0006310-DNA recombination20.082RECQL4, SETMAR

[i] T2D, type 2 diabetes; GO, gene ontology.

Screening for potential TFs

The enrichment of TF binding sites in the upstream region of the abnormal hyper- and hypomethylated genes was analyzed. Multiple TF binding sites were significantly enriched among the differentially methylated genes, and four corresponding TFs were subsequently identified. As shown in Fig. 2, only the TF zinc finger E-box binding homeobox 1 (ZEB1) was identified as possessing a large probability to bind and regulate the hyper-methylated genes. While the binding sites of three TFs, methyl CpG binding protein 2 (MECP2), TFEB and TFAP4, were enriched in the hypo-methylated genes.

Discussion

The differences in global DNA methylation in muscle tissues from 11 monozygotic twin pairs discordant for T2D were detected using microarray data. The utilization of mono-zygotic twins facilitates investigation of the abnormal DNA methylation induced by environmental factors, but not hereditary factors, in T2D patients.

The significantly hyper- and hypo-methylated genes identified in the current study may provide several novel candidate genes associated with the occurrence of T2D. SIRT1, an NAD(+)-dependent histone deacetylase, which was hyper-methylated according to the present results, was observed to regulate glucose/lipid metabolism through its deacetylase activity on numerous substrates (21,22). In addition, SIRT1 positively regulates insulin secretion in pancreatic β cells (23), and protects cells from oxidative stress and inflammation. Consequently, the methylation of the SIRT1 gene may be a significant gene modification involved in the occurrence of T2D. The other two genes, NAT6 and PLA2G12B, which were also hyper-methylated in T2D muscle, are involved in the metabolism of lipids (24). PLA2G12B belongs to the PLA2 family, and catalyzes the hydrolysis of glycolipids to release free fatty acids and lysophospholipids (25). NAT6 encodes a cytoplasmic N-acetyltransferase protein, with substrate specificity for proteins with an N-terminal methionine (26). Additionally, these two genes are involved in the pathway of glycerophospholipid metabolism according to the Kyoto Encyclopedia of Genes and Genomes pathway analysis (http://www.genome.jp/kegg/). All of this cumulative evidence has suggested that the hyper-methylation of PLA2G12B and NAT6 may result in the dysfunction of glycerophospholipid metabolism leading to hyperlipidemia in T2D patients.

Of the hypo-methylated genes, NFATC1 is particularly notable for its potential association with T2D. A previous study revealed that mice with a β-cell-specific deletion of the calci-neurin b1 (CNB1) subunit developed age-dependent diabetes characterized by decreased β-cell proliferation and mass, reduced pancreatic insulin content and hypo-insulinemia. The expression of active NFATC1 in CNB1-deficient β cells may rescue these defects and prevent diabetes (27). This evidence indicated the positive role of NFATC1 in the pathogenesis or the potential therapeutic role in diabetes.

According to the enrichment of TF binding sites in the upstream region of the differentially methylated genes, it was demonstrated that the transcription factor ZEB1 was significantly enriched for its binding to the hyper-methylated genes identified in the current study. Papadopoulou et al (28) hypothesized that transfection of ZEB1 cDNA may induce a reduction in B cell lymphoma 6 (BCL6) expression, and that siRNA-mediated knockdown of ZEB1 may lead to an increase in BCL6 mRNA expression (28). BCL-6 represses menin, a product of the Men1 gene, expression. Transgenic menin overexpression in β cells prevents islet expansion, which leads to maternal hyperglycemia (29). Therefore, transgenic menin mice exhibit features of human gestational diabetes (29). For these reasons, ZEB1, as a transcription factor, may be involved in the pathogenesis of T2D. The binding site of MECP2 was significantly enriched by the hypo-methylated genes in the present study. MECP2 is capable of binding specifically to methylated DNA to suppress transcription from methylated gene promoters (30). The DNA methylation of genes, as the major modification of eukaryotic genomes (31), was repressed by MECP2, indicating a key function for MECP2 in the regulation of methylated genes. It was therefore hypothesized that MECP2 may be involved in the process of T2D via regulation of the hypo-methylated genes; however, this hypothesis requires further investigation for confirmation. The present results also identified two candidate TFs (TFEB and TFAP4), which may regulate the hypo-methylated genes in T2D. To the best of our knowledge, few studies have investigated the association between these two TFs and T2D, and therefore future studies should focus on elucidating this association.

In conclusion, a bioinformatics analysis of the differentially methylated genes in muscle tissues from 11 monozygotic twin pairs discordant for T2D was performed. The identified diabetes-associated genes (SIRT1, NAT6, PLA2G12B and NFATC1) and potential TFs provided a novel insight into the molecular mechanism underlying the occurrence of T2D. These genes may become promising target genes in the development of novel treatments for T2D.

Acknowledgments

The present study was supported by a grant from a project supported by the China National Natural Science Foundation (grant no. 81300260) and the Foundation of Science and Technology Department of Sichuan Province (grant no. 14ZC1146).

References

1 

Parving H-H, Brenner BM, McMurray JJ, et al ALTITUDE Investigators: Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med. 367:2204–2213. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Kitada M and Koya D: SIRT1 in type 2 diabetes: Mechanisms and therapeutic potential. Diabetes Metab J. 37:315–325. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Kahn SE: The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of Type 2 diabetes. Diabetologia. 46:3–19. 2003.PubMed/NCBI

4 

Spranger J, Kroke A, Möhlig M, et al: Adiponectin and protection against type 2 diabetes mellitus. Lancet. 361:226–228. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Hofsø D, Jenssen T, Hager H, Røislien J and Hjelmesaeth J: Fasting plasma glucose in the screening for type 2 diabetes in morbidly obese subjects. Obes Surg. 20:302–307. 2010. View Article : Google Scholar

6 

Zhang P, Zhang X, Brown J, et al: Global healthcare expenditure on diabetes for 2010 and 2030. Diabetes Res Clin Pract. 87:293–301. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Liebl A, Mata M and Eschwège E; ODE-2 Advisory Board: Evaluation of risk factors for development of complications in Type II diabetes in Europe. Diabetologia. 45:S23–S28. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Sjöström L, Lindroos A-K, Peltonen M, et al Swedish Obese Subjects Study Scientific Group: Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 351:2683–2693. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Barres R and Zierath JR: DNA methylation in metabolic disorders. Am J Clin Nutr. 93:897S–900S. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Takizawa T, Nakashima K, Namihira M, et al: DNA methylation is a critical cell-intrinsic determinant of astrocyte differentiation in the fetal brain. Dev Cell. 1:749–758. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Robertson KD: DNA methylation and human disease. Nat Rev Genet. 6:597–610. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Rönn T, Poulsen P, Hansson O, et al: Age influences DNA methylation and gene expression of COX7A1 in human skeletal muscle. Diabetologia. 51:1159–1168. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Ling C, Poulsen P, Carlsson E, et al: Multiple environmental and genetic factors influence skeletal muscle PGC-1α and PGC-1β gene expression in twins. J Clin Invest. 114:1518–1526. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Ling C, Poulsen P, Simonsson S, et al: Genetic and epigenetic factors are associated with expression of respiratory chain component NDUFB6 in human skeletal muscle. J Clin Invest. 117:3427–3435. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Jensen CB, Storgaard H, Madsbad S, Richter EA and Vaag AA: Altered skeletal muscle fiber composition and size precede whole-body insulin resistance in young men with low birth weight. J Clin Endocrinol Metab. 92:1530–1534. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Barrett T, Wilhite SE, Ledoux P, et al: NCBI GEO: archive for functional genomics data sets - update. Nucleic Acids Res. 41:D991–D995. 2013. View Article : Google Scholar

17 

Du P, Zhang X, Huang CC, et al: Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics. 11:5872010. View Article : Google Scholar : PubMed/NCBI

18 

Da Wei Huang BTS, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Dubchak I, Munoz M, Poliakov A, et al: Whole-Genome rVISTA: a tool to determine enrichment of transcription factor binding sites in gene promoters from transcriptomic data. Bioinformatics. 29:2059–2061. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Wingender E, Dietze P, Karas H and Knüppel R: TRANSFAC: A database on transcription factors and their DNA binding sites. Nucleic Acids Res. 24:238–241. 1996. View Article : Google Scholar : PubMed/NCBI

21 

Kume S, Uzu T, Kashiwagi A and Koya D: SIRT1, a calorie restriction mimetic, in a new therapeutic approach for type 2 diabetes mellitus and diabetic vascular complications. Endocr Metab Immune Disord Drug Targets. 10:16–24. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Sharma S, Misra CS, Arumugam S, et al: Antidiabetic activity of resveratrol, a known SIRT1 activator in a genetic model for type 2 diabetes. Phytother Res. 25:67–73. 2011. View Article : Google Scholar

23 

Bordone L, Motta MC, Picard F, et al: Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic β cells. PLoS Biol. 4:e312006. View Article : Google Scholar

24 

Bhakta S, Besra GS, Upton AM, et al: Arylamine N-acetyltransferase is required for synthesis of mycolic acids and complex lipids in Mycobacterium bovis BCG and represents a novel drug target. J Exp Med. 199:1191–1199. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Aljakna A, Choi S, Savage H, et al: Pla2g12b and Hpn are genes identified by mouse ENU mutagenesis that affect HDL cholesterol. PLoS One. 7:e431392012. View Article : Google Scholar : PubMed/NCBI

26 

Blum M, Grant DM, Mcbride W, Heim M and Meyer UA: Human arylamine N-acetyltransferase genes: isolation, chromosomal localization, and functional expression. DNA Cell Biol. 9:193–203. 1990. View Article : Google Scholar : PubMed/NCBI

27 

Heit JJ, Apelqvist AA, Gu X, et al: Calcineurin/NFAT signalling regulates pancreatic β-cell growth and function. Nature. 443:345–349. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Papadopoulou V, Postigo A, Sánchez-Tilló E, Porter AC and Wagner SD: ZEB1 and CtBP form a repressive complex at a distal promoter element of the BCL6 locus. Biochem J. 427:541–550. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Karnik SK, Chen H, McLean GW, et al: Menin controls growth of pancreatic β-cells in pregnant mice and promotes gestational diabetes mellitus. Science. 318:806–809. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Nan X, Ng HH, Johnson CA, et al: Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature. 393:386–389. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Li E, Beard C and Jaenisch R: Role for DNA methylation in genomic imprinting. Nature. 366:362–365. 1993. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 12 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu F, Sun Q, Wang L, Nie S and Li J: Bioinformatics analysis of abnormal DNA methylation in muscle samples from monozygotic twins discordant for type 2 diabetes. Mol Med Rep 12: 351-356, 2015
APA
Liu, F., Sun, Q., Wang, L., Nie, S., & Li, J. (2015). Bioinformatics analysis of abnormal DNA methylation in muscle samples from monozygotic twins discordant for type 2 diabetes. Molecular Medicine Reports, 12, 351-356. https://doi.org/10.3892/mmr.2015.3452
MLA
Liu, F., Sun, Q., Wang, L., Nie, S., Li, J."Bioinformatics analysis of abnormal DNA methylation in muscle samples from monozygotic twins discordant for type 2 diabetes". Molecular Medicine Reports 12.1 (2015): 351-356.
Chicago
Liu, F., Sun, Q., Wang, L., Nie, S., Li, J."Bioinformatics analysis of abnormal DNA methylation in muscle samples from monozygotic twins discordant for type 2 diabetes". Molecular Medicine Reports 12, no. 1 (2015): 351-356. https://doi.org/10.3892/mmr.2015.3452