Skip to main content

ORIGINAL RESEARCH article

Front. Endocrinol., 16 February 2023
Sec. Clinical Diabetes
This article is part of the Research Topic Clinical and Genetic Determinants of Diabetes and Complications View all 31 articles

The transcriptomic and epigenetic alterations in type 2 diabetes mellitus patients of Chinese Tibetan and Han populations

  • 1School of Biological and Behavioral Sciences, Queen Mary University of London, London, United Kingdom
  • 2Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
  • 3Department of Endocrinology, Kunming Municipal Hospital of Traditional Chinese Medicine, Kumning, China
  • 4Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China

Background: Due to the distinctive living environment, lifestyle, and diet, the Tibetan community in China has the lowest prevalence of T2DM and prediabetes among numerous ethnic groups, while Han community shows the highest statistic. In this study, we aim to conclude the clinical manifestations of both Tibetan and Han T2DM patients and their association with transcriptomic and epigenetic alterations.

Methods: A cross-sectional study including 120 T2DM patients from Han and Tibetan ethnic groups were conducted between 2019 to 2021 at the Hospital of Chengdu University of Traditional Chinese Medicine. The various clinical features and laboratory tests were recorded and analyzed between the two groups. The genome-wide methylation pattern and RNA expression were determined by Reduced Representation Bisulfite Sequencing (RBBS) and Poly (A) RNA sequencing (RNA-seq) from leucocytes of peripheral blood samples in 6 Han and 6 Tibetan patients. GO analysis and KEGG analysis were conducted in differentially expressed genes and those with differentially methylated regions.

Results: Compared to Han, Tibetan T2DM individuals intake more coarse grains, meat and yak butter, but less refined grains, vegetables and fruit. They also showed increased BMI, Hb, HbA1c, LDL, ALT, GGT and eGFR, and decreased level of BUN. Among the 12 patients in the exploratory cohort, we identified 5178 hypomethylated and 4787 hypermethylated regions involving 1613 genes in the Tibetan group. RNA-seq showed a total of 947 differentially expressed genes (DEGs) between the two groups, with 523 up-regulated and 424 down-regulated in Tibetan patients. By integrating DNA methylation and RNA expression data, we identified 112 DEGs with differentially methylated regions (overlapping genes) and 14 DEGs with promoter-related DMRs. The functional enrichment analysis demonstrated that the overlapping genes were primarily involved in metabolic pathways, PI3K-Akt signaling pathway, MAPK signaling pathway, pathways in cancer and Rap1 signaling pathway.

Conclusion: Our study demonstrates the clinical characteristics of T2DM differ subtly between various ethnic groups that may be related to epigenetic modifications, thus providing evidence and ideas for additional research on the genetic pattern of T2DM.

1. Introduction

The main features of type 2 diabetes mellitus (T2DM) include hyperinsulinemia, insulin resistance (IR) and islet cell damage, which can reach 50% at the time of diagnosis (1). With a high-energy diet, decreased physical activity, and an increase in obesity, the incidence of diabetes is rising globally, along with the rate of disability and mortality. People who have T2DM experience vascular and neurological consequences, as well as life, psychological, and financial stress. The diabetic population will predictably reach 147 million by 2045 (2). Most diabetes is a complex disease caused by a combination of multiple genes and environmental factors. Genetic factors are present in approximately 25% to 69% of people with T2DM worldwide (3) and over 560 genetic loci are identified to be relevant (4).

Epigenetics, including DNA methylation, histone modifications and microRNAs, lead to changes in gene function based on mitosis and meiosis without alteration in DNA sequence (5), in which DNA methylation has been recognized to be an important genetic factor contributing to T2DM (6). DNA methylation refers to the S-adenosyl methionine (SAM), as the methyl donor, transfers the activated methyl group to carbon 5 of the cytosine-phosphate-guanine (CpG) by the catalyzation of DNA methyltransferases (DNMTs). In general, gene expression is opposite to the level of methylation in the promoter region, which means low methylation levels result in up-regulation of gene expression, whereas high methylation results in down-regulation of expression (7, 8). As DNA methylation is reversible and can be interfered with, some chemicals can be used as targets to modify DNA methylation (9), providing a new perspective for T2DM treatment. Previous studies have shown that many genes are related to islet function, such as PDX1 (10), PPARGC1A (11), INS (12), GLP1R (13) and KCNQ1 (14), have been associated with the development of T2DM. Meanwhile, methylome-wide association studies (MWAS) for T2DM have identified differentially methylated sites (DMSs) in TXNIP (15), PHOSPHO1 (16), SREBF1 (17), ABCG1 (17), SOCS3 (18), and CPTA1 (19).

Environmental factors such as diet, exercise and obesity can also alter the epigenome. Tibetans are a distinct ethnic group in China that have historically lived at high altitudes. They primarily reside in the Tibetan Autonomous Region (TAR), as well as the provinces of Qinghai, Sichuan, Yunnan, and Gansu in China. Although highlanders had a lower incidence of diabetes, the number has quickly risen as a result of greater longevity and lifestyle changes (20, 21). According to nationwide research, the Han Chinese population had a 14.7% prevalence of diabetes and a 38.8% prevalence of prediabetes, whereas the Tibetan community had the lowest prevalence of both conditions at 4.3% and 31.3%, respectively (22). Lifestyle changes, particularly in calorie intake, are associated with the development of diabetes, possibly through epigenetic mechanisms (23, 24). This study aimed to demonstrate the differences in clinical characteristics between Tibetan and Han T2DM patients and to explore the transcriptomic and epigenetic alterations in the two groups.

2. Materials and methods

2.1. Study population

2.1.1. Cross-sectional cohort

A total of 60 Tibetan and 60 Han patients with T2DM were recruited at the Hospital of Chengdu University of Traditional Chinese Medicine from 2019 to 2021. All the patients were diagnosed with T2DM according to the 1999 WHO criteria (25). There was no kinship between the included study subjects and three consecutive generations for each patient are the same ethnic group. The exclusion criteria include 1) other types of diabetes; 2) having immune system diseases; 3) any types of tumors; 4) acute and chronic infections; 5) psychoneurological disorders; 6) recent use of drugs that affect lipid metabolism; 7) having liver or kidney failure or severe heart diseases; 8) disagreeing to participate in the study.

2.1.2. Subjects enrolled for exploratory cohort

Among the cross-sectional cohort, 6 Tibetan and 6 matched Han T2DM patients were selected for the exploratory cohort of RNA expression and DNA methylation. These two groups were selected by a matched pairs design based on shared characteristics including age, gender, weight, height and duration of T2DM to control lurking variables.

2.2. Clinical data collection

The general information for all patients included age, gender, body mass index (BMI), drinking and smoking history, family history of diabetes, the duration of T2DM, food intake, systolic blood pressure (SBP), diastolic blood pressure (DBP) and hemoglobin (Hb). In addition to HbA1c test, a standard 2-h OGTT test was performed by using a 75g glucose load to assess the patient’s islet function and glycemic control. Plasma glucose and insulin level at 0 (fasting), 1, 2 and 3-hour postprandial blood glucose (PBG) were measured, and blood C-peptide was measured at 0 and 2-hour postprandial. The biochemical analysis includes the total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), γ-glutamyltransferase (GGT), total bile acid (TBA), direct bilirubin (DBIL), blood creatinine (BCr), blood urea nitrogen (BUN), blood uric acid (BUA), were detected to estimate blood lipids, liver function, and kidney function. The Cockcroft-Gault equation was used to determine the estimated glomerular filtration rate (eGFR) (26).

2.3. Blood sample and DNA extraction

In the exploratory cohort, 3ml EDTA-treated peripheral blood sample of each participant was collected and stored in -80°C. Genomic DNA was extracted from peripheral blood using magnetic universal genomic DNA kit (TIANGEN Biotech (Beijing) co., Ltd). DNA concentration and quality were measured by Nanodrop.

2.4. Reduced representation bisulfite sequencing

1µg genomic DNA was digested using MspI enzyme for 16 hours at 37°C. After digestion, libraries were constructed as the Illumina Pair-End protocol with some modifications. Briefly, purified digested DNA was subsequently treated with a mix of T4 DNA polymerase, Klenow Fragment and T4 polynucleotide kinase to repair, blunt and phosphorylate ends. The DNA fragments were subsequently 3’ adenylated using Klenow Fragment (3’-5’ exo-) and following with ligation to adaptors synthesized with 5’-methylcytosine instead of cytosine using T4 DNA Ligase. the DNA was purified using QIAquick PCR purification kit (Qiagen) after reaction of each step. After purification, the library was subjected to 40°C for 30 min treatment in a thermo cycler with the lid heated at 57°C. After that, centrifuged the reaction mixture at 14,000 X g for 10 min and then transferred the supernatant into a new 0.2 ml PCR tube for the further bisulfite treatment, respectively. Bisulfite conversion treatment was performed using a ZYMO EZ DNA Methylation-Gold Kit (Zymo research, Irvine, CA, USA) according to the manufacturer’s instructions. The final RRBS libraries were generated by PCR amplification using adapter compatible barcode primers, quantified by an Agilent 2100 Bioanalyzer (Agilent Technologies) and real-time PCR assay and then sequenced by Illumina Hiseq.

2.5. Methylation calculation and identification of DMRs

Low-quality reads that contained more than 5 ‘N’s or had a low-quality value for over 50% of the sequence (Phred score< 5) were filtered. The sequencing reads of the samples were aligned to the human reference genome (hg19) using BSMAP (Version 2.74) (27). The methylated CpG (mCG) sites were identified following a previously described algorithm (28). The methylation levels for each sample were calculated using in-house Perl scripts. Differentially methylated regions (DMRs) were identified using metilene (Version 0.2-6) within a 500 bp sliding window at 250 bp steps with at least 10 CpGs covered by over 10× sequence reads, applying the thresholds of differential methylation β ≥ 15%, FDR for two-dimensional Kolmogorov-Smirnov-Test p-value< 0.05 (29). The enrichment analyses were conducted using WebGestalt (WEB-based Gene SeT Analysis Toolkit) (30).

2.6. RNA library construction and sequencing

Total RNA was extracted from cells using Trizol (Invitrogen) according to the manufacturer’s protocol, and ribosomal RNA was removed using the Ribo-Zero™ kit (Epicentre, Madison, WI, USA). Fragmented RNA (the average length was approximately 200 bp) was subjected to first-strand and second-strand cDNA synthesis followed by adaptor ligation and enrichment with a low cycle according to instructions of NEBNext® Ultra™ RNA Library Prep Kit for Illumina (NEB, USA). The purified library products were evaluated using the Agilent 2200 TapeStation and Qubit®2.0 (Life Technologies, USA). The libraries were paired-end sequenced (PE150, Sequencing reads were 150 bp) at Guangzhou MethylGene Co., Ltd. (Guangzhou, China) using the Illumina Xten platform.

2.7. Pre-processing of sequencing reads/quality control

Raw fastq sequences were treated with Trimmomatic tools (v 0.36) using the following options: TRAILING: 20, MINLEN:235 and CROP:235, to remove trailing sequences below a Phred quality score of 20 and to achieve uniform sequence lengths for downstream clustering processes. Sequencing read quality was inspected using the FastQC software. Adapter removal and read trimming were performed using Trimmomatic. Sequencing reads were trimmed from the end (base quality less than Q20) and filtered by length (less than 25).

2.8. Quantification of gene expression level

Paired-end reads were aligned to the human reference genome (hg19) with HISAT2. HTSeq v0. 6.0 was used to count the numbers of reads mapped to each gene. The whole sample expression levels were presented as RPKM (expected number of Reads Per Kilobase of transcript sequence per Million base pairs sequenced), which is the recommended and most common method to estimate the level of gene expression.

2.9. Differential expression analysis

The statistically significant DE genes were obtained by an adjusted P-value threshold of<0.05 and |log2(fold change) | > 1 using the DEGseq software. Finally, a hierarchical clustering analysis was performed using the R language package gplots according to the RPKM values of differential genes in different groups. And colors represent different clustering information, such as the similar expression pattern in the same group, including similar functions or participating in the same biological process.

2.10. GO terms and KEGG pathway enrichment analysis

All differentially expressed mRNAs were selected for GO and KEGG pathway analyses. GO was performed with KOBAS3.0 software, including cellular component (CC), molecular function (MF) and biological process (BP). GO provides label classification of gene function and gene product attributes (http://www.geneontology.org). GO analysis covers three domains: cellular component (CC), molecular function (MF) and biological process (BP). The differentially expressed mRNAs and the enrichment of different pathways were mapped using the KEGG pathways with KOBAS3.0 software (http://www.genome.jp/kegg).

2.11. Statistical analysis

The median and quartile were used for the statistical and data description of the normally distributed measures, and the number of cases (n) and percentages (%) were used for the statistical and data description of the categorical counts. Normality and homogeneity of all data were evaluated by Kolmogorov-Smirnov test. Student T-test or Mann-Whitney U test was applied to compare the differences of continuous variables. Pearson Chi-square test was employed to evaluate statistical differences of categorical variables. The Wilcoxon test was used to compare the continuous non-normally distributed variables between 6 Tibetans and 6 Hans in the exploratory cohort. Pearson correlation was used to identify the 14 overlapping genes and clinical characteristics with significant differences. All data were statistically analyzed by SPSS 23.0 software (SPSS Inc., Chicago, IL, USA). Graphs were generated using Graphpad 7.0 software (GraphPad Software, Inc., San Diego, USA).

3. Results

3.1. The demographical and clinical characteristics between Tibetan and Han T2DM populations

A total of 120 participants were enrolled for the final analysis, including 60 Tibetans and 60 Hans. The patient flow chart is demonstrated in Figure 1. The basic and biochemical characteristics are shown in Table 1. Although no difference was observed in age, gender, and duration of T2DM, the BMI of Tibetans was significantly higher than Hans (26.08 vs 23.3, P = 0.017). Tibetans consume fewer refined grains (141.5 g/day vs 193.5 g/day, P< 0.001), vegetables and fruit (91 g/day vs 296.5 g/day, P< 0.001) than Han people, but they consume more coarse grains (171 g/day vs 63.5 g/day, P< 0.001), meat (181.5 g/day vs 100.5 g/day, P< 0.001), and yak butter (98.5 g/day vs 0 g/day, P< 0.001). Not surprisingly, the Hb level of Tibetans is higher than Hans (146.5 g/L vs 138.5 g/L, P< 0.001) due to the high-altitude, low-oxygen environment of Tibetan settlements. Despite similar BG, insulin, and C-peptide level, HbA1c of Tibetan T2DM patients was higher than Han patients (9.75% vs 8.65%, P = 0.001). Similarly, LDL level is significantly higher in Tibetan group compared to Han group (3.12 mmol/L vs 2.53 mmol/L, P = 0.002). Regarding the liver function, the blood tests also showed higher levels of ALT (30.5 IU/L vs 21.5 IU/L, P = 0.013), and GGT (38 IU/L vs 21 IU/L, P< 0.001). The level of BUN was lower (4.95 mmol/L vs 5.61 mmol/L, P = 0.002) and eGFR of Tibetan T2DM patients was statistically higher than Han patients (129.77 mL/min vs 96.5 mL/min, P< 0.001). There were no significant differences in other parameters of biochemical tests between the two groups.

FIGURE 1
www.frontiersin.org

Figure 1 Flow chart of patient collection. COPD: chronic obstructive pulmonary disease.

TABLE 1
www.frontiersin.org

Table 1 Demographical and biochemical characteristics between Tibetan and Han T2DM patients in cross-sectional cohort.

A total of 12 patients with 6 in each group were selected by paired design for the exploratory cohort. The age ranged from 33 to 54 years old and the duration of T2DM ranged from 2 to 7.1 years. As shown in Supplementary Table S1, there were no significant differences in basic and biochemical parameters except HbA1c (9.9% vs 9%, P = 0.046), FBG (8.48 mmol/L vs 9.99 mmol/L, P = 0.028), 3-hr Insulin (12.11 mIU/L vs 21.07 mIU/L, P = 0.046), HDL (0.94 mmol/L vs 1.17 mmol/L, P = 0.046) and eGFR (143.7 mL/min vs 97 mL/min, P = 0.028).

3.2. Differentially methylated positions and regions

The whole-genome DNA methylation was detected by RRBS using peripheral blood samples from 6 Tibetan and 6 Han T2DM patients. After sulphite treatment, the conversion efficiency of all samples ranged from 98.82% to 99.27%. About 80% to 90% mCs were CG dinucleotides while about 10% to 20% were at CHG and CHH sites (G = A, C or T) (Supplementary Figure S1). Additionally, the methylation level of mC was around 80% to 100% while mCHG and mCHH were around 0% to 20%, with 20% as an interval (Supplementary Figure S2). We also explored the methylation levels in different genome regions. The level of methylation decreased in the 2kb upstream of transcription initiation but rose sharply in the exon region and reaches a maximum in the intron and 2kb downstream of genes (Supplementary Figure S3). The DMRs were mainly located in the intergenic region, accounting for 38.83%, followed by intron (32.15%) and exon regions (10.38%), respectively, in addition to 6.7% of DMRs within the gene promoter region (upstream 2kb) (Figure 2A). PCA found distinct clusters for study subjects (Figure 2B). The heatmap (Figure 2C) and volcano map (Figure 2D) have demonstrated the methylation difference between the two groups. Compared with Han group, we identified 5178 hypomethylated regions and 4787 hypermethylated regions in Tibetans (Table 2).

FIGURE 2
www.frontiersin.org

Figure 2 Summary of DMRs between Han and Tibetan T2D patients (A) The overall distribution of DMRs. (B) The principal component analysis plot using the differential methylated CpG sites between Han and Tibetans. (C) Volcano plot of methylation difference between Han and Tibetans. A total of 4787 CpG sites hypermethylated in Tibetans was represented by red point in the right side. A total of 5178 CpG sites hypomethylated in Tibetans was represented by red point in the left side. (D) Heatmap clustering analysis of DMRs of different gene functional regions. Highly methylated sites are shown in red and sparsely methylated sites are shown in blue. In addition, the pink clusters represent Tibetans and the blue clusters represent Han Chinese. H: Han, Z: Tibetan.

TABLE 2
www.frontiersin.org

Table 2 The numbers and length of differentially methylated regions.

We performed GO functional analysis according to DMR-related genes, which were mostly enriched in protein binding (BP), nucleus (CC), cytoplasm (CC) and membrane (CC) (Figure 3A). KEGG analysis showed that DMR-related genes are mainly involved in metabolic pathway, pathways in cancer, cAMP signaling pathway, HTLV-I infection, cytokine-cytokine receptor interaction, calcium signaling pathway, alcoholism, regulation of actin cytoskeleton, hippo signaling pathway, Wnt signaling pathway, non-alcoholic fatty liver disease (NAFLD), insulin secretion, glycerophospholipid metabolism and type 2 diabetes mellitus (Figure 3B).

FIGURE 3
www.frontiersin.org

Figure 3 Pathway Analysis on DMR-related genes. (A), GO analysis of DMR-related genes. (B), KEGG analysis of DMR-related genes.

3.3. Transcriptome analysis

We conducted RNA-seq on peripheral blood samples from Han and Tibetan T2DM patients in order to investigate the relationship between DNA methylation and gene expression. Each sample produced about 8 giga bases (Gb) of filtered data. Additionally, using HISAT2 software, sequencing data were compared to the human reference genome with an average match rate of 90.2% per sample and an average unique mapping rate of 86.95% (Supplementary Table S2). Gene expression levels are calculated by RPKM as the number of reads per kilobase length from a given gene per million reads, and are calculated as  RPKM=Total exon readsMapped reads×exon length  (31).

A volcano map of significantly differentially expressed genes (DEGs) was created by differential gene expression analysis using the DESeq program, with 523 genes significantly up-regulated and 424 genes significantly down-regulated in the Tibetan group compared to the Han group (Figure 4A). The heat map revealed distinct gene expression patterns in the Tibetan and Han populations (Figure 4B).

FIGURE 4
www.frontiersin.org

Figure 4 The volcano plot and heatmap of DEGs in Han and Tibetan T2D patients. (A) Volcano plot of DEGs. The x-axis represents the log2 fold change and the y-axis represents the log10 (P-value). The green dots represent downregulated genes and red dots represent upregulated genes. (B) Heat map of DEGs following clustering analysis. The vertical axis represents the sample, and the horizontal axis represents DEGs. Up: the number of up-regulated genes, down: the number of down-regulated genes, H: Han, Z: Tibetan.

Functional annotation showed that the most represented GO categories for DEGs were extracellular (CC), receptor-mediated endocytosis (BP), xenobiotic metabolic process (BP), negative regulation of endopeptidase activity (BP) and cellular response to hormone stimulus (BP) (Figure 5A), while KEGG enrichment analysis showed that the upregulated DEGs were mainly involved in steroid hormone biosynthesis, retinol metabolism, drug metabolism-cytochrome P450, PI3K-AkT signaling pathway, pentose and glucuronate interconversions, starch and sucrose metabolism, ascorbate and aldarate metabolism (Figure 5B).

FIGURE 5
www.frontiersin.org

Figure 5 GO and KEGG enrichment analysis of DEGs. (A) GO analysis of differentially expressed genes, (B) KEGG analysis of differentially expressed genes.

3.4. Integrative analysis of transcriptome and DNA methylation

In general, gene expression is negatively correlated with DNA methylation. We divided each sample into four categories, including silence, low expression, medium expression, and high expression, according to the amount of gene expression and counted the methylation levels in the gene regions of each of the four categories of genes in a single sample. Our results showed DNA methylation was negatively correlated with gene expression in regions within 1k upstream of the gene, and genes with high methylation status were not expressed or were under-expressed (Figure 6).

FIGURE 6
www.frontiersin.org

Figure 6 Relationship between DNA methylation and gene expression. Silence (RPKM=0), low: low expression (0<RPKM ≤ 1), medium: medium expression (1<RPKM ≤ 10), high: high expression (RPKM>10). H: Han, Z: Tibetan.

We observed 112 overlapping DEGs and DMR genes, of which 14 were promoter-related genes (Table 3). The GO enrichment analysis showed that the most significant enriched GO terms of overlapped genes are integral component of membrane (CC), plasma membrane (CC), homophilic cell adhesion via plasma membrane adhesion molecules (BP), calcium ion binding (BP) (Figure 7A).

TABLE 3
www.frontiersin.org

Table 3 Overlapped genes of DEGs and promoter related DMR genes.

FIGURE 7
www.frontiersin.org

Figure 7 GO and KEGG enrichment analysis of DMR-related DEGs. (A) GO analysis of DMR-related DEGs. (B) KEGG analysis of DMR-related DEGs.

According to KEGG enrichment analysis, these overlapping genes were primarily involved in metabolic pathway, including metabolism of xenobiotics by cytochrome P450, steroid hormone biosynthesis, retinol metabolism, ascorbate and aldarate metabolism, pentose and glucuronate interconversions, porphyrin and chlorophyll metabolism, drug metabolism, starch and sucrose metabolism, chemical carcinogenesis, PI3K-Akt signaling pathway, MAPK signaling pathway, pathways in cancer and Rap1 signaling pathway (Figure 7B). The relationship between overlapping genes and significant clinical characteristics was analyzed by Pearson correlation analysis. We found that the HbA1c was associated with the expression of RHOD (R = 0.697, P< 0.05), LOC100134868 (R = -0.697, P< 0.01) and LOC102723828 (R = -0.661, P< 0.05); FBG was negatively associated with APOB (R = -0.631, P< 0.05); HDL was positively associated with PAX8-AS1 (R = 0.615, P< 0.05); and eGFR was related with FOXA (R = 0.794, P< 0.01) and UMODL1-AS1 (R = 0.662, P< 0.05). In addition, insulin levels at three hours after 75g glucose load test showed positive association with the expression of MIXL1, OXCT2, LAMA5-AS1, LOC100134868 and LOC102723672 while negatively related to AJAP1, as shown in Supplementary table S3.

4. Discussion

Our study reported the clinical characteristics of Han and Tibetan T2DM patients, indicating that the same disease has clinical differences between various ethnic groups and providing evidence for clinical individualization of T2DM treatment. We also revealed for the first time the differences in DNA methylation and RNA expression between Tibetan and Han T2DM patients, and synthesized the relationship between them, which provides a basis for further exploration of T2DM development mechanisms and identification of therapeutic targets.

Tibetans live in a high altitude, low oxygen, low temperature environment. Previous studies have shown that in a healthy population of Han Chinese and Tibetans living at the same altitude, the hemoglobin concentration of highland Han is higher than that of Tibetans (32). However, Han Chinese living at lower altitudes have lower hemoglobin concentrations (33), which is consistent with our results. The diet of Tibetans consists mainly of coarse grains, meat, yak butter and other high-fat, high-calorie, high-protein foods, thus have a higher BMI. However, no differences were shown in other lipid indicators in our cross-sectional cohort, except for higher LDL level in the Tibetan T2DM group, which indicates that Tibetans may have higher metabolism.

The Han and Tibetan populations also showed slight differences in liver function, with Tibetans having higher ALT and GGT levels, but both in the normal range. Aminotransferases are considered indicators of hepatocyte health, and GGT also reflects biliary tract function. Elevated ALT is associated with age, obesity, elevated triglyceride levels, and low HDL cholesterol levels, but not with glycemic control (34). However, independent of common risk factors, ALT (35, 36) and GGT (37) are linked to an increase in the risk of T2DM. Although the eGFR level of Tibetan T2DM patients was higher and the BUN was lower than that of Han Chinese, both were at normal levels.

In the exploratory cohort, we further investigated the differences in DNA methylation and transcriptome between Han and Tibetan populations to interpret the differences in the development of T2DM between the two groups through a genetic perspective. The CpG island is a region of the DNA sequence rich in CpG sites, usually located in the promoters with an unmethylated state. When CpG islands are methylated, transcription factors become impaired in binding to promoters or bind to transcriptional repressors, altering the structure of chromatin. As a result, gene expression is altered without the changes of DNA sequence, affecting biological processes and leading to diseases (38, 39).

In our study, a total of 1613 genes with DMRs were found between Han Chinese and Tibetan T2DM patients. After GO and KEGG functional enrichment analysis, we identified signaling pathways that affect metabolism and other pathways that may play a key role in the development of T2DM, such as insulin secretion. Among them, cAMP signaling pathway, Wnt signaling pathway, and Hippo signaling pathway were more significant and relevant. cAMP is an intracellular mediator of insulin and adrenal glycogen catabolism in the liver (40). In mammals, cAMP activates cAMP-dependent protein kinase (PKA), which phosphorylates downstream protein targets and then regulates the function of ion channels, transcription factors and enzymes. Meanwhile, the cAMP signaling pathway regulates glucose homeostasis due to insulin secretion, glucose utilization, and glycogen synthesis and catabolism (41). The Wnt signaling plays an important role as an evolutionary pathway in regulating cellular homeostasis and energy homeostasis from the hypothalamus to the metabolic organs. The classical Wnt as well as non-classical Wnt pathways inhibit metabolism and lead to increased adipose tissue, resulting in metabolic stress and metabolic inflammation and obesity (42). The Hippo signaling pathway plays a role in pancreatic, hepatic, adipose and cardiac cells as well as in systemic metabolism, regulating glucolipid metabolism. Activation of the Hippo signaling pathway in hyperglycemic states induces proliferation and differentiation of pancreatic β-cells, increasing glucose uptake and utilization, thereby reducing insulin resistance, and improving insulin secretion (43).

In general, gene expression follows an opposite trend to the level of methylation in the promoter region. In the present study, we identified 947 differentially expressed genes, of which 112 overlapping genes had differential methylation levels, and a total of 14 genes with differentially methylated regions in the promoter region. Among the differentially expressed genes found to be differentially methylated in promoter regions, APOB encodes apolipoprotein B and is associated with LDL, celiac and LDL structural integrity, in lipid digestion, mobilization as well as transport (44). A study on the amount of non-insulin-dependent diabetic patients showed that APOB polymorphisms were effective in improving blood glucose and lipid levels of T2DM patients (45). PAX8-AS1 is a non-coding RNA that is involved in the pathology of the disease despite its inability to encode protein synthesis. In a study examining non-coding RNA in leukocytes from patients with gestational diabetes mellitus (GDM), PAX8-AS1 expression levels were significantly lower in GDM patients compared to healthy pregnant women and could be used as a diagnostic biomarker for GDM (46). The rest of key genes need to be further studied in the future.

Similar to previously described, KEGG analysis was mainly enriched in metabolic pathways that are related to IR or diabetes, including metabolism of xenobiotics by cytochrome P450 (47), steroid hormone biosynthesis (48), retinol metabolism (49), ascorbate and aldarate metabolism (50), pentose and glucuronate interconversions (51), starch and sucrose metabolism. Several canonical pathways outstood among the statistics, including PI3K-Akt pathway, MAPK pathways and Rap1 signaling pathway. Insulin secretion activates PI3K-Akt signaling pathway throughout the body to increase glucose utilization, reduce glucose metabolism in the liver and muscle, and regulate the balance of lipid and glucose metabolism. However, impairment of this pathway leads to insulin resistance, which in turn worsens this pathway, leading to T2DM (52). In addition, insulin can also activate MAPK pathways but inappropriate MAPK signaling contributes to the development of metabolic syndrome and T2DM (53). An in vitro study showed that activated Rap1 is a key regulator of β-cell function, as evidenced by the promotion of glucose-stimulated insulin production, islet cell hypertrophy, and islet cell proliferation by activated Rap1A (54).

The overlapping genes primarily are associated with metabolism and insulin-related pathways, suggesting that the environment and lifestyle, such as diet, may play a role in altering DNA methylations levels, therefore affecting metabolism and insulin secretion and utilization in T2DM patients.

Conclusion

As the prevalence of T2DM varies in different ethnic groups in China, our study revealed the diverse clinical features of Tibetan and Han T2DM patients. The epigenetic and transcriptional patterns have provided a perspective on the mechanisms of T2DM in different ethnic groups, and the key genes are worthy to be further studied to reveal the importance of DNA methylation for the development of T2DM.

Data availability statement

The datasets presented in this study can be found in online repositories. The name of the repository/repositories and accession number(s) can be found below: https://www.ncbi.nlm.nih.gov/, accession number: PRJNA911064.

Ethics statement

The studies involving human participants were reviewed and approved by Hospital of Chengdu University of Traditional Chinese Medicine. The patients/participants provided their written informed consent to participate in this study.

Author contributions

JL and XW conceived of the presented idea, conducted the study of cross-sectional cohort. XW carried out the exploratory study of epigenome and transcriptome. QC and QW supervised the project. All the authors contributed to the final version of the manuscript.

Funding

The study was supported by Sichuan Provincial Science and Technology Plan Project (19YYJC1515) and Special Training Program for Outstanding Young Scientific and Technological Talents (Innovation) of Chinese Medical Sciences (Grant No. ZZ14-YQ-010).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fendo.2023.1122047/full#supplementary-material

Supplementary Figure 1 | The proportion of different types of methylated cytosines in 12 samples In the pie chart, red, green and blue indicate mCG, mCHH and mCHG, respectively. H: Han, Z: Tibetan

Supplementary Figure 2 | Distribution of mC levels in mCG, mCH and mCHH. The figure showed the distribution of mC levels in 12 samples. The X axis showed the methylation level, and the Y axis showed the percentage of mC. Red, blue and green lines represent CG, CHG and CHH, respectively. H: Han, Z: Tibetan

Supplementary Figure 3 | Distribution of DNA methylation levels in each gene elements. Red, blue and green lines represent CG, CHG and CHH, respectively. H: Han, Z: Tibetan

References

1. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HAW. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med (2008) 359:1577–89. doi: 10.1056/NEJMOA0806470

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Sinclair A, Saeedi P, Kaundal A, Karuranga S, Malanda B, Williams R. Diabetes and global ageing among 65-99-year-old adults: Findings from the international diabetes federation diabetes atlas, 9th edition. Diabetes Res Clin Pract (2020) 162. doi: 10.1016/J.DIABRES.2020.108078

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Almgren P, Lehtovirta M, Isomaa B, Sarelin L, Taskinen MR, Lyssenko V, et al. Heritability and familiality of type 2 diabetes and related quantitative traits in the botnia study. Diabetologia (2011) 54:2811–9. doi: 10.1007/S00125-011-2267-5

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Vujkovic M, Keaton JM, Lynch JA, Miller DR, Zhou J, Tcheandjieu C, et al. Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat Genet (2020) 52:680–91. doi: 10.1038/S41588-020-0637-Y

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Wu CT, Morris JR. Genes, genetics, and epigenetics: a correspondence. Science (2001) 293:1103–5. doi: 10.1126/SCIENCE.293.5532.1103

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Raciti GA, Desiderio A, Longo M, Leone A, Zatterale F, Prevenzano I, et al. And type 2 diabetes: Novel biomarkers for risk assessment? Int J Mol Sci (2021) 22. doi: 10.3390/IJMS222111652

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet (2012) 13:484–92. doi: 10.1038/NRG3230

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Klose RJ, Bird AP. Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci (2006) 31:89–97. doi: 10.1016/J.TIBS.2005.12.008

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Duthie SJ. Epigenetic modifications and human pathologies: cancer and CVD. Proc Nutr Soc (2011) 70:47–56. doi: 10.1017/S0029665110003952

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Yang BT, Dayeh TA, Volkov PA, Kirkpatrick CL, Malmgren S, Jing X, et al. Increased DNA methylation and decreased expression of PDX-1 in pancreatic islets from patients with type 2 diabetes. Mol Endocrinol (2012) 26:1203–12. doi: 10.1210/ME.2012-1004

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Ling C, del Guerra S, Lupi R, Rönn T, Granhall C, Luthman H, et al. Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. Diabetologia (2008) 51:615–22. doi: 10.1007/S00125-007-0916-5

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Yang BT, Dayeh TA, Kirkpatrick CL, Taneera J, Kumar R, Groop L, et al. Insulin promoter DNA methylation correlates negatively with insulin gene expression and positively with HbA(1c) levels in human pancreatic islets. Diabetologia (2011) 54:360–7. doi: 10.1007/S00125-010-1967-6

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Hall E, Dayeh T, Kirkpatrick CL, Wollheim CB, Dekker Nitert M, Ling C. DNA Methylation of the glucagon-like peptide 1 receptor (GLP1R) in human pancreatic islets. BMC Med Genet (2013) 14. doi: 10.1186/1471-2350-14-76

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Travers ME, Mackay DJG, Nitert MD, Morris AP, Lindgren CM, Berry A, et al. Insights into the molecular mechanism for type 2 diabetes susceptibility at the KCNQ1 locus from temporal changes in imprinting status in human islets. Diabetes (2013) 62:987–92. doi: 10.2337/DB12-0819

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Wang Z, Peng H, Gao W, Cao W, Lv J, Yu C, et al. Blood DNA methylation markers associated with type 2 diabetes, fasting glucose, and HbA1c levels: An epigenome-wide association study in 316 adult twin pairs. Genomics (2021) 113:4206–13. doi: 10.1016/J.YGENO.2021.11.005

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Kulkarni H, Kos MZ, Neary J, Dyer TD, Kent JW, Göring HHH, et al. Novel epigenetic determinants of type 2 diabetes in Mexican-American families. Hum Mol Genet (2015) 24:5330–44. doi: 10.1093/HMG/DDV232

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Chambers JC, Loh M, Lehne B, Drong A, Kriebel J, Motta V, et al. Epigenome-wide association of DNA methylation markers in peripheral blood from Indian asians and europeans with incident type 2 diabetes: a nested case-control study. Lancet Diabetes Endocrinol (2015) 3:526–34. doi: 10.1016/S2213-8587(15)00127-8

PubMed Abstract | CrossRef Full Text | Google Scholar

18. al Muftah WA, Al-Shafai M, Zaghlool SB, Visconti A, Tsai PC, Kumar P, et al. Epigenetic associations of type 2 diabetes and BMI in an Arab population. Clin Epigenet (2016) 8. doi: 10.1186/S13148-016-0177-6

CrossRef Full Text | Google Scholar

19. Cardona A, Day FR, Perry JRB, Loh M, Chu AY, Lehne B, et al. Epigenome-wide association study of incident type 2 diabetes in a British population: EPIC-Norfolk study. Diabetes (2019) 68:2315–26. doi: 10.2337/DB18-0290/-/DC1

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Schulz LO, Bennett PH, Ravussin E, Kidd JR, Kidd KK, Esparza J, et al. Effects of traditional and western environments on prevalence of type 2 diabetes in pima indians in Mexico and the U. S Diabetes Care (2006) 29:1866–71. doi: 10.2337/DC06-0138

CrossRef Full Text | Google Scholar

21. Centers for Disease Control and Prevention (CDC). Prevalence of diagnosed diabetes among American Indians/Alaskan natives–united states, 1996. MMWR Morb Mortal Wkly Rep (1998) 47:901–4.

PubMed Abstract | Google Scholar

22. Wang L, Gao P, Zhang M, Huang Z, Zhang D, Deng Q, et al. Prevalence and ethnic pattern of diabetes and prediabetes in China in 2013. JAMA (2017) 317:2515–23. doi: 10.1001/JAMA.2017.7596

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Gillberg L, Ling C. The potential use of DNA methylation biomarkers to identify risk and progression of type 2 diabetes. Front Endocrinol (Lausanne) (2015) 6:43. doi: 10.3389/FENDO.2015.00043

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Ling C, Groop L. Epigenetics: a molecular link between environmental factors and type 2 diabetes. Diabetes (2009) 58:2718–25. doi: 10.2337/DB09-1003

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Alberti KGMM, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabetes Med (1998) 15:539–53. doi: 10.1002/(sici)1096-9136(199807)15:7<539::aid-dia668>3.0.co;2-s

CrossRef Full Text | Google Scholar

26. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron (1976) 16:31–41. doi: 10.1159/000180580

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Xi Y, Li W. BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinf (2009) 10. doi: 10.1186/1471-2105-10-232

CrossRef Full Text | Google Scholar

28. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature (2009) 462:315–22. doi: 10.1038/NATURE08514

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Jühling F, Kretzmer H, Bernhart SH, Otto C, Stadler PF, Hoffmann S. Metilene: fast and sensitive calling of differentially methylated regions from bisulfite sequencing data. Genome Res (2016) 26:256–62. doi: 10.1101/GR.196394.115

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Wang J, Duncan D, Shi Z, Zhang B. WEB-based GEne SeT AnaLysis toolkit (WebGestalt): update 2013. Nucleic Acids Res (2013) 41. doi: 10.1093/NAR/GKT439

CrossRef Full Text | Google Scholar

31. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B. Mapping and quantifying mammalian transcriptomes by RNA-seq. Nat Methods (2008) 5:621–8. doi: 10.1038/NMETH.1226

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Simonson TS, Wei G, Wagner HE, Wuren T, Bui A, Fine JM, et al. Increased blood-oxygen binding affinity in Tibetan and han Chinese residents at 4200 m. Exp Physiol (2014) 99:1624. doi: 10.1113/EXPPHYSIOL.2014.080820

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Sun L, Fan F, Li R, Niu B, Zhu L, Yu S, et al. Different erythrocyte MicroRNA profiles in low- and high-altitude individuals. Front Physiol (2018) 9:1099. doi: 10.3389/FPHYS.2018.01099

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Saligram S, Williams EJ, Masding MG. Raised liver enzymes in newly diagnosed type 2 diabetes are associated with weight and lipids, but not glycaemic control. Indian J Endocrinol Metab (2012) 16:1012. doi: 10.4103/2230-8210.103027

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Vozarova B, Stefan N, Lindsay RS, Saremi A, Pratley RE, Bogardus C, et al. High alanine aminotransferase is associated with decreased hepatic insulin sensitivity and predicts the development of type 2 diabetes. Diabetes (2002) 51:1889–95. doi: 10.2337/DIABETES.51.6.1889

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Cho NH, Jang HC, Sung HC, Kim HR, Hong KL, Chan JCN, et al. Abnormal liver function test predicts type 2 DiabetesA community-based prospective study. Diabetes Care (2007) 30:2566–8. doi: 10.2337/DC07-0106

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Perry IJ, Wannamethee SG, Shaper AG. Prospective study of serum gamma-glutamyltransferase and risk of NIDDM. Diabetes Care (1998) 21:732–7. doi: 10.2337/DIACARE.21.5.732

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell (2007) 128:669–81. doi: 10.1016/J.CELL.2007.01.033

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Clouaire T, Stancheva I. Methyl-CpG binding proteins: specialized transcriptional repressors or structural components of chromatin? Cell Mol Life Sci (2008) 65:1509–22. doi: 10.1007/S00018-008-7324-Y

PubMed Abstract | CrossRef Full Text | Google Scholar

40. BERTHET J, TW RALL, SUTHERLAND EW. The relationship of epinephrine and glucagon to liver phosphorylase. IV. effect of epinephrine and glucagon on the reactivation of phosphorylase in liver homogenates. J Biol Chem (1957) 224:463–75. doi: 10.1016/S0021-9258(18)65045-8

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Yang H, Yang L. Targeting cAMP/PKA pathway for glycemic control and type 2 diabetes therapy. J Mol Endocrinol (2016) 57:R93–R108. doi: 10.1530/JME-15-0316

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Aamir K, Khan HU, Sethi G, Hossain MA, Arya A. Wnt signaling mediates TLR pathway and promote unrestrained adipogenesis and metaflammation: Therapeutic targets for obesity and type 2 diabetes. Pharmacol Res (2020) 152. doi: 10.1016/J.PHRS.2019.104602

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ardestani A, Lupse B, Maedler K. Hippo signaling: Key emerging pathway in cellular and whole-body metabolism. Trends Endocrinol Metab (2018) 29:492–509. doi: 10.1016/J.TEM.2018.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Knott TJ, Rall SC, Innerarity TL, Jacobson SF, Urdea MS, Levy-Wilson B, et al. Human apolipoprotein b: structure of carboxyl-terminal domains, sites of gene expression, and chromosomal localization. Science (1985) 230:37–43. doi: 10.1126/SCIENCE.2994225

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Duman BS, Öztürk M, Yilmazer S, Çağatay P, Hatemi H. Apolipoprotein b gene variants are involved in the determination of blood glucose and lipid levels in patients with non-insulin dependent diabetes mellitus. Cell Biochem Funct (2006) 24:261–7. doi: 10.1002/CBF.1218

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Hu J, Mu H, Gao L, Pan Y, Wu C, Zhang D, et al. Diagnostic value of candidate noncoding RNAs in leukocytes of patients with gestational diabetes mellitus. Exp Ther Med (2021) 21. doi: 10.3892/ETM.2020.9576

CrossRef Full Text | Google Scholar

47. Gupta HP, Jha RR, Ahmad H, Patel DK, Ravi Ram K. Xenobiotic mediated diabetogenesis: Developmental exposure to dichlorvos or atrazine leads to type 1 or type 2 diabetes in drosophila. Free Radic Biol Med (2019) 141:461–74. doi: 10.1016/J.FREERADBIOMED.2019.07.013

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Ding EL, Song Y, Manson JE, Rifai N, Buring JE, Liu S. Plasma sex steroid hormones and risk of developing type 2 diabetes in women: A prospective study. Diabetologia (2007) 50:2076–84. doi: 10.1007/S00125-007-0785-Y/TABLES/5

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Rhee EJ, Plutzky J. Retinoid metabolism and diabetes mellitus. Diabetes Metab J (2012) 36:167–80. doi: 10.4093/DMJ.2012.36.3.167

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Xu Y, Wu Y, Xiong Y, Tao J, Pan T, Tan S, et al. Ascorbate protects liver from metabolic disorder through inhibition of lipogenesis and suppressor of cytokine signaling 3 (SOCS3). Nutr Metab (Lond) (2020) 17:1–10. doi: 10.1186/S12986-020-0431-Y/FIGURES/5

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Zheng H, Wu J, Huang H, Meng C, Li W, Wei T, et al. Metabolomics analysis of the protective effect of rubusoside on palmitic acid-induced lipotoxicity in INS-1 cells using UPLC-Q/TOF MS. Mol Omics (2019) 15:222–32. doi: 10.1039/C9MO00029A

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Huang X, Liu G, Guo J, Su ZQ. The PI3K/AKT pathway in obesity and type 2 diabetes. Int J Biol Sci (2018) 14:1483. doi: 10.7150/IJBS.27173

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Gehart H, Kumpf S, Ittner A, Ricci R. MAPK signalling in cellular metabolism: stress or wellness? EMBO Rep (2010) 11:834–40. doi: 10.1038/EMBOR.2010.160

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Kelly P, Bailey CL, Fueger PT, Newgard CB, Casey PJ, Kimple ME. Rap1 promotes multiple pancreatic islet cell functions and signals through mammalian target of rapamycin complex 1 to enhance proliferation. J Biol Chem (2010) 285:15777. doi: 10.1074/JBC.M109.069112

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: DNA methylation, type 2 diabetes mellitus, Han, Tibetan, transcriptome

Citation: Wang X, Liu J, Wang Q and Chen Q (2023) The transcriptomic and epigenetic alterations in type 2 diabetes mellitus patients of Chinese Tibetan and Han populations. Front. Endocrinol. 14:1122047. doi: 10.3389/fendo.2023.1122047

Received: 12 December 2022; Accepted: 01 February 2023;
Published: 16 February 2023.

Edited by:

Sen Li, Beijing University of Chinese Medicine, China

Reviewed by:

Qian Zhang, Peking Union Medical College Hospital (CAMS), China
Wenshan Lv, The Affiliated Hospital of Qingdao University, China

Copyright © 2023 Wang, Liu, Wang and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Qiuhong Wang, qiuhongfortune@126.com; Qiu Chen, chenqiu1005@cdutcm.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.