Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


No abstract provided.

Free full text 


Logo of diabetesLink to Publisher's site
Diabetes. 2009 Dec; 58(12): 2718–2725.
PMCID: PMC2780862
PMID: 19940235

Epigenetics: A Molecular Link Between Environmental Factors and Type 2 Diabetes

Although obesity, reduced physical activity, and aging increase susceptibility to type 2 diabetes, many people exposed to these risk factors do not develop the disease. Recent genome-wide association studies have identified a number of genetic variants that explain some of the interindividual variation in diabetes susceptibility (15). There is also a growing body of literature suggesting a role for epigenetic factors in the complex interplay between genes and the environment. Nevertheless, our knowledge about the molecular mechanisms linking environmental factors and type 2 diabetes still remains limited. This perspective will provide some insights into epigenetic mechanisms associated with type 2 diabetes.

An overview of epigenetic regulation.

Although there is no uniform definition of epigenetics, it has been described as heritable changes in gene function that occur without a change in the nucleotide sequence (6). Epigenetic modifications can be passed from one cell generation to the next (mitotic inheritance) and between generations of a species (meiotic inheritance). In plants, it is well established that epigenetic modifications can be inherited from one generation to the next (7). However, there is only limited information about the inheritance of epigenetic traits between generations in animals (8,9). Notably, epigenetic effects may also be affected by the environment, making them potentially important pathogenic mechanisms in complex multifactorial diseases such as type 2 diabetes (Fig. 1). Epigenetic factors include DNA methylations, histone modifications, and microRNAs, and they can help to explain how cells with identical DNA can differentiate into different cell types with different phenotypes. This perspective will focus on the roles of DNA methylation and histone modification in the pathogenesis of type 2 diabetes.

An external file that holds a picture, illustration, etc.
Object name is zdb0120959580001.jpg

Model proposing a role for epigenetic mechanisms in the pathogenesis of type 2 diabetes.

Cytosine residues occurring in CG dinucleotides are targets for DNA methylation in vertebrates, and DNA methylation is associated with transcriptional silencing (e.g., on the inactive X chromosome). This silencing can be achieved by either repressing the binding of transcription factors (Fig. 2A) or by recruiting proteins that specifically bind to methylated CGs (methyl-CG–binding proteins, e.g., MeCP2), which can further recruit histone deacetyltransferases (HDACs) and corepressors (Fig. 2B) (10).

An external file that holds a picture, illustration, etc.
Object name is zdb0120959580002.jpg

Effects of DNA methylation on gene expression. A: Whereas low levels of DNA methylation at gene promoters have been proposed to generate active genes through increased binding of transcription factors, elevated DNA methylation at promoters may inhibit binding of transcription factors resulting in inactive genes. B: DNA methylation at gene promoters may also repress gene transcription via specific proteins that bind to methylated CpGs (methyl-CpG binding proteins, e.g., MeCP2), and these proteins may then recruit HDACs and transcriptional corepressors (e.g., NCoR), resulting in an altered chromatin structure and inactive genes.

DNA methylation requires the activity of methyltransferases. There are two groups of DNA methyltransferases: DNMT1, which copies the DNA methylation pattern between cell generations during replication (maintenance methylation), and DNMT3a and DNMT3b, which are responsible for de novo methylation of DNA (10). The process leading to demethylation of DNA is still poorly understood; for a recent review see Patra et al. (11).

Genomic DNA in eukaryotic cells is packed together with special proteins, termed histones, to form chromatin. The basic building block of chromatin is the nucleosome, which consists of ~147 base pairs of DNA wrapped around an octamer of histone proteins that is composed of an H3-H4 tetramer flanked on either side with an H2A-H2B dimer. Although the core histones are densely packed, their NH2-terminal tails can be modified by histone- modifying enzymes, resulting in acetylation, methylation, phosphorylation, sumoylation, or ubiquitination (12). These modifications are important for determining the accessibility of the DNA to the transcription machinery as well as for replication, recombination, and chromosomal organization.

HDACs remove and histone acetyl transferases (HATs) add acetyl groups to lysine residues on histone tails (1214). Although, it is well established that HAT activity and increased histone acetylation correlate with increased gene transcription, the exact mechanisms promoting transcription are less clear (15). Native lysine residues on histone tails contain a positive charge that can bind negatively charged DNA to form a condensed structure with low transcriptional activity. An early suggestion was that histone acetylation removes these positive charges, thereby relaxing chromatin structure and facilitating access to the DNA for the transcriptional machinery to initiate transcription (13,15). However, different models have recently been proposed, including the histone code hypothesis, where multiple histone modifications act in combination to regulate transcription (15,16). Histone acetylation may also recruit bromodomain proteins that can act as transcriptional activators (13). Histone methylation can result in either transcriptional activation or inactivation, depending on the degree of methylation and the specific lysine and/or arginine residues modified (17,18). Histone methyltransferases and histone demethylases mediate these processes (18).

New techniques have made it easier to analyze DNA methylation and histone modifications on a genome-wide scale (19,20). These techniques may be useful when studying the impact of epigenetics on the pathogenesis of type 2 diabetes.

Epigenetic changes induced by aging.

Aging is associated with an increased risk of type 2 diabetes. Correspondingly, oxidative capacity and mitochondrial function decline with age as well as in patients with type 2 diabetes (2126). The mechanisms behind these defects may be both genetic and environmental (2731). Recent data further suggest that the epigenetic pattern may change during the course of life, affecting key genes in the respiratory chain (3234). COX7A1, which is part of complex 4 of the respiratory chain and which shows decreased expression in diabetic muscle, is a target of age-related DNA methylation (23,34). Whereas DNA methylation of the COX7A1 promoter is increased in skeletal muscle of elderly compared with young twins, the opposite pattern is found for COX7A1 gene expression (34). Additionally, the transcript level of COX7A1 in skeletal muscle is associated with increased in vivo glucose uptake and Vo2max (34). These data demonstrate how age can influence DNA methylation, gene expression, and subsequently in vivo metabolism. The interaction between nongenetic and epigenetic mechanisms may further be affected by genetic factors. Indeed, a polymorphism introducing a possible DNA methylation site, a CG dinucleotide, and a putative transcription factor binding site in the NDUFB6 promoter is associated with increased DNA methylation, decreased gene expression, and decreased in vivo metabolism with increasing age (33). This study provides an example of interactions between genetic (polymorphism), epigenetic (DNA methylation), and nongenetic (age) factors in the determination of human metabolism.

Hepatic insulin resistance is another important characteristic of both aging and type 2 diabetes. Glucokinase is a key enzyme in hepatic glucose utilization, and its activity is decreased in the liver of diabetic patients (35). Mutations in the glucokinase gene can cause a monogenic form of diabetes (maturity-onset diabetes of the young [MODY] 2) (36). Moreover, in aged compared with young rats, the liver displays reduced levels of glucokinase expression and enzyme activity in parallel with increased DNA methylation of the glucokinase promoter (37). When hepatocytes of aged rats were cultured in vitro and the DNA was chemically demethylated, there was a substantial increase in glucokinase expression, suggesting an important role for DNA methylation in the age-related regulation of this gene. Similar studies in humans with diabetes are still lacking.

Although aging is associated with gene-specific hypermethylation, many mammalian tissues demonstrate global hypomethylation of DNA and decreased methyltransferase (DNMT1 and DNMT3a) expression with increased age (33,34,3745). Global hypomethylation of DNA is seen in repetitive sequences and may promote genomic instability during aging. Increased age is also associated with hypomethylation of specific genes, e.g., proto-oncogenes, thereby increasing susceptibility to cancer, especially if combined with hypermethylation of tumor suppressor genes. Further studies examining the effects of aging on genome-wide epigenetic patterns in target tissues may help to improve our understanding of the molecular mechanisms influencing the pathogenesis of type 2 diabetes.

Links between obesity, energy metabolism, nutrients, and epigenetic modifications.

The prevalence of type 2 diabetes is increasing rapidly worldwide, partly due to the epidemic in obesity seen among most ethnic groups. The fact that loss of function of the histone demethylase, Jhdm2a, is associated with obesity, decreased expression of metabolically active genes (e.g., peroxisome proliferator–activated receptor-α and medium-chain acyl-CoA dehydrogenase) in skeletal muscle, and an impaired cold-induced uncoupling protein 1 expression in brown adipose tissue in rodents suggests a relationship between epigenetic mechanisms and obesity (46). Another class of enzymes involved in epigenetic control of metabolism is the nicotinamide adenine dinucleotide (NAD+)-dependent sirtuins (class III HDACs), which target both histone and nonhistone proteins (47). The most well-characterized member, SIRT1, regulates several metabolic pathways including adipogenesis, mitochondrial biogenesis, glucose utilization, fat oxidation, and insulin secretion. Moreover, ATP-citrate lyase is an enzyme that regulates the conversion of citrate to acetyl CoA, which is a metabolite required for acetylation of histones by HATs. It has recently been suggested that glucose availability can affect histone acetylation in an ATP-citrate lyase-dependent manner, further linking energy metabolism to epigenetic regulation (48).

Leptin is a hormone secreted from adipocytes that regulates appetite and energy homeostasis. It is predominantly expressed in mature adipocytes, and leptin expression can therefore be used as a marker of differentiating preadipocytes. The leptin promoter is embedded within a CG-rich region, called a CpG island. Although there is a high degree of DNA methylation of the leptin promoter and no leptin expression in preadipocytes, the promoter is demethylated in parallel with induction of leptin expression in differentiated cells (49,50). Although a high-fat diet increases DNA methylation of one CpG site in the leptin promoter of rat adipocytes (51), it remains to be examined whether food intake and obesity are associated with epigenetic regulation of leptin expression in human adipocytes.

Environmental exposures to nutrients may change gene expression and alter disease susceptibility through epigenetic modifications. Similar mechanisms are operative in the agouti mouse; the agouti gene encodes a paracrine-signaling molecule that promotes melanocytes to produce yellow rather than black coat pigment and makes mice prone to develop obesity, diabetes, and tumors (5254). The degree to which the agouti gene is methylated regulates agouti expression and thereby coat color and risk for disease. Moreover, supplementation of the diets of pregnant mice with methyl donors such as folic acid, vitamin B12, choline, or betaine increases DNA methylation of the gene in the offspring, resulting in low agouti expression and a brown coat color (55). The effect of maternal methyl-donor supplementation on coat color is also inherited in the F2 generation through germline epigenetic modifications (56).

Pdx1/insulin promoter factor (IPF)-1 is a transcription factor regulating pancreas development and β-cell differentiation, and mutations in this gene can cause a monogenic form of diabetes (MODY 4) (36). Intrauterine growth retardation due to uteroplacental insufficiency has recently been associated with progressive epigenetic silencing of Pdx1, impaired β-cell function, and type 2 diabetes in adult offspring (57). Whether PDX1 is a target for similar epigenetic mechanisms in humans born to mothers with uteroplacental insufficiency remains unknown. However, the prenatal environment has been associated with insulin resistance and a risk for type 2 diabetes in humans (5860), and the prenatal nutrient supply may induce epigenetic changes in humans. Indeed, individuals from the The Dutch Hunger Winter Families Study who were prenatally exposed to famine in 1944–1945 showed less DNA methylation of the imprinted IGF2 and INSIGF genes and increased DNA methylation of the GNASAS, MEG3, IL10, ABCA1, and LEP genes in parallel with impaired glucose tolerance compared with their unexposed same-sex siblings (6063). Moreover, a high-fat diet during pregnancy in rats is associated with impaired glucose homeostasis and mitochondrial and cardiovascular dysfunctions in adult rats, possibly due to epigenetic modifications (6466). In future studies it will be interesting to study the effects of short- and long-term weight gain and weight loss on epigenetic changes in relevant tissues.

Histone modifications induced by exercise.

Poor physical fitness and a low Vo2max predict risk of developing type 2 diabetes (67). Mitochondrial dysfunction, changes in muscle fiber–type composition, and insulin resistance are potential mechanisms linking poor physical fitness with an increased risk for disease. Exercise induces the expression of a number of genes that regulate glucose uptake in skeletal muscle, including GLUT isoform 4 (GLUT4), (68). GLUT4 expression is further regulated by the transcription factor myocyte enhancer factor 2 (MEF2).

At rest, it has been proposed that MEF2 interacts with HDAC5 in the nucleus (69). Histone tails at the GLUT4 gene are thereby deacetylated by HDAC5, resulting in a condense chromatin structure and subsequently reduced GLUT4 expression (69). After exercise, HDAC5 is phosphorylated by AMP-activated protein kinase, dissociated from MEF2, and exported from the nucleus to the cytosol (6971). MEF2 may then interact with the coactivator protein PPARγ coactivator-1α (PPARGC1A) and HATs in the nucleus, resulting in acetylated histones at the GLUT4 gene, enhanced transcriptional activity, and increased GLUT4 expression (69,72,73). It is possible that other histone modifications also influence the regulation of GLUT4 expression in skeletal muscle. Ca+/calmodulin-dependent protein kinase (CaMK) also seems to modulate MEF2 activity via histone acetylation in response to acute exercise (74). Moreover, gene expression of MYST4, a HAT, correlates positively with the percentage of type 1 fibers and Vo2max in human skeletal muscle (75). Together, these data suggest that some of the biological changes induced by exercise could be due to histone modifications, a research area that deserves further exploration.

Epigenetic changes in patients with type 2 diabetes.

Although data mining analysis has suggested a role for epigenetic factors in the pathogenesis of type 2 diabetes (76), there are only a limited number of studies that have examined epigenetic changes in target tissues from patients with type 2 diabetes. The transcriptional coactivator PPARGC1A coordinates gene expression that stimulates mitochondrial oxidative metabolism in multiple tissues (77). Whereas DNA methylation of the PPARGC1A promoter is elevated in pancreatic islets from patients with type 2 diabetes compared with that of healthy control subjects, PPARGC1A expression is reduced in diabetic islets and correlates inversely with the degree of DNA methylation (78). Importantly, PPARGC1A expression correlates positively with glucose-stimulated insulin secretion in human pancreatic islets (78), suggesting that epigenetic mechanisms may regulate gene expression and, subsequently, insulin secretion in human islets.

Moreover, there have been some efforts to understand the epigenetic regulation of insulin gene expression in pancreatic β-cells. In β-cells, the insulin gene displays hyperacetylation of H4 and hypermethylation of H3 at lysine 4, typical of active genes; however, these epigenetic marks are not seen at the insulin gene in other cell types, e.g., HeLa cells (79,80). Furthermore, in a β-cell line, the HAT p300 and the histone methyltransferase SET7/9 are recruited to the insulin promoter to activate the gene (80). Interestingly, it has been suggested that HDACs influence pancreatic development in rodents because treatment with HDAC inhibitors during embryonic development enhances the pool of β-cells (81). However, it remains to be established whether any of these epigenetic marks in the insulin gene are affected in β-cells from patients with type 2 diabetes.

Although pancreatic islet β-cell proliferation declines after birth, β-cell proliferation may play a role in the islets adaption to increased insulin demands imposed by insulin resistance. In support of this, an increased expression of Ink4a/Arf (Cdkn2a locus) was associated with reduced β-cell regeneration in aging mice (82). The elevated Ink4a/Arf expression in elderly mice further coincided with reduced levels of histone H3 lysine 27 trimethylation at Ink4a/Arf and the histone methyltransferase, Ezh2, together with decreased Bmi-1 binding and a loss of H2A ubiquitylation at Ink4a/Arf (83,84). Interestingly, a common variant at the CDKN2A locus has been associated with an increased risk for type 2 diabetes (14). However, whether this variant is associated with decreased β-cell proliferation in human islets remains unknown.

Monogenic diabetes and epigenetic factors.

Most forms of MODY are caused by mutations in genes encoding transcription factors, including HNF1A, -4A, and -1B as well as IPF1/PDX1 and NEUROD1, some of which regulate transcription of their target genes through associations with HATs and HDACs.

HNF1α activates transcription through two different mechanisms: 1) recruitment of the general transcription machinery and 2) chromatin remodeling of promoter regions (85). The chromatin remodeling involves recruitment of HATs (e.g., p300/CBP), resulting in hyperacetylation of histones at specific promoters, including GLUT2 and pyruvate kinase, in β-cells (8688). Interestingly, a missense mutation (R263L) in the HNF1A gene that is associated with a MODY phenotype results in reduced affinity for p300 (89). Moreover, MODY mutations in the HNF1B gene influence the capacity of HNF1β to bind proteins with HAT activity and may thereby affect the chromatin structure (90).

Whereas Pdx1 influences glucose-induced expression of insulin in β-cells, this regulation requires an interaction between Pdx1 and p300 and thereby hyperacetylation of histone H4 at the insulin gene promoter (9193). It has been suggested that a low glucose level decreases insulin expression due to recruitment of HDAC1 and HDAC2 by Pdx1 (94). Insulin transcription also involves methylation of histone H3 at the insulin promoter, possibly by Pdx1 recruiting methyltransferase SET9 (95). Several PDX1 mutations associated with diabetes in humans modulate the affinity of PDX1 for both p300 and DNA. If operative in humans in vivo, this suggests that HAT activity and, therefore the chromatin structure of target genes may influence the risk for diabetes (92). NEUROD plays an important role in the development of the pancreas and regulates the transcription of insulin (36). One mutation in NEUROD, which results in a truncated protein and diabetes, also prevents it from binding to p300/CBP (96). Collectively, the data described above suggest mechanisms by which chromatin modifications can influence the risk of diabetes, which thereby opens new possible avenues for therapeutically preserving β-cell function.

DNA methylation and transient neonatal diabetes.

Transient neonatal diabetes (TND) is a rare form of diabetes that begins in the first 6 weeks of life in growth-retarded neonates (97). Although insulin therapy is only required for an average of 3 months, the majority of these patients develop type 2 diabetes later in life. Three different chromosome 6 anomalies have been described in TND: hypomethylation at chromosome 6q24, paternally inherited duplication of 6q24, and paternal uniparental isodisomy of chromosome 6 (97,98). Interestingly, it has recently been shown that mutations in a zinc-finger transcription factor, ZFP57, are associated with TND and hypomethylation of regions on 6q24, including the imprinted genes PLAGL1 and HYMAI (98).

Epigenetic changes associated with diabetic complications.

One major event in the progression of diabetic complications is vascular inflammation with increased expression of inflammatory genes. Enhanced oxidative stress, dyslipidemia, and hyperglycemia have also been suggested to influence the development of diabetic complications. Recent studies have proposed that hyperglycemia may induce epigenetic modifications of genes involved in vascular inflammation. Nuclear factor-κB (NF-κB) is a transcription factor regulating expression of genes involved in inflammatory diseases, including atherosclerosis and diabetic complications (99). Poor glycemic control increases NF-κB activity in monocytes and thereby gene expression of inflammatory cytokines (100,101). This regulation involves an interaction between NF-κB and HATs (e.g., CBP/p300), resulting in hyperacetylation of target genes including the tumor necrosis factor (TNF)-α and cyclooxygenase-2 promoters (99). The histone H3 lysine 4 methyltransferase SET7/9 can also influence the recruitment of NF-κB p65 to gene promoters and thereby its regulation of proinflammatory genes (102). Moreover, vascular smooth muscle cells from diabetic db/db mice show decreased levels of histone H3 lysine 9 trimethylation (H3K9me3) and elevated levels of histone H3 lysine 4 dimethylation (H3K4me2) at the promoters of inflammatory genes, e.g., IL-6 and MCP-1, in parallel with decreased levels of the H3K9me3 methyltransferase Suv39h1 and a histone demethylase, the lysine-specific demethylase 1 (LSD1) (103,104). Interestingly, whereas overexpression of Suv39h1 in vascular smooth muscle cells from diabetic db/db mice reversed the diabetic phenotype, gene silencing of SUV39H1 in normal human vascular smooth muscle cells increased the expression of inflammatory genes (104). NFB and IL-6 also represent genes with altered histone H3 lysine 9 dimethylation in lymphocytes from patients with type 1 diabetes (105). Together, these studies suggest that hyperglycemia may induce epigenetic changes of proinflammatory genes, which subsequently regulate gene expression and thereby the development of vascular inflammation. However, improved glycemic control for 3–5 years in diabetic patients did not reduce the risk of macrovascular complications (106,107). One reason could be that the effects of hyperglycemia may be long-term and that epigenetic modifications induced by hyperglycemia may persist for more than 5 years. Moreover, because the time-averaged mean levels of glycemia, measured as A1C, only explain part of the variation in risk of developing diabetic complications, it was recently hypothesized that transient exposures to hyperglycemia may induce sustained epigenetic changes and thereby NF-κB–regulated gene expression and increased risk for vascular complications over a longer period of time (108,109). Indeed, a transient exposure to hyperglycemia (16 h) induces epigenetic changes in the promoter of the NFB subunit p65 and subsequently p65 expression and NF-κB activity in aortic endothelial cells. These changes persist for 6 days during culture at normal glucose levels. Interestingly, when genes that reduce mitochondrial superoxide production (e.g., uncoupling protein-1) are overexpressed, the changes induced by the transient hyperglycemia are prevented (109). It was further shown that both a histone methylase (SET7) and a histone demethylase (LSD1) may regulate the epigenetic changes in the NFB p65 promoter induced by transient hyperglycemia (110). In fact, epigenetic modifications induced by transient hyperglycemia may explain the hyperglycemic memory that has been proposed in epidemiological studies. In the future it may be possible that drugs using and/or affecting epigenetic mechanisms, e.g., HDAC inhibitors, can be used in the treatment of diabetic complications (13,111,112). In support of this idea, a recent study showed that myocardial infarction and ischemia induce HDAC activity in parallel with decreased histone acetylation of histone H3 and 4 in the heart (113). The use of chemical HDAC inhibitors during myocardial infarction reduced the infarct area as well as cell death (113).

Conclusions.

The use of genome-wide technologies to study gene expression and genetic variation in patients with type 2 diabetes has increased rapidly over the recent years, generating long lists of new type 2 diabetes candidate genes. However, the use of global techniques to study epigenetic modifications in these same patients has been limited. Epigenetic changes associated with type 2 diabetes are therefore still poorly understood. Nevertheless, epigenetics may play an important role in the growing incidence of type 2 diabetes, and over the next few years, it will be a great challenge to dissect the role of histone modifications and DNA methylation in the pathogenesis of the disease and its complications. Two additional important questions are whether the epigenetic changes induced by today's sedentary lifestyle can be inherited by coming generations and whether these changes are reversible. Currently, several epigenetic drugs are being tested in clinical trials or are already being used (e.g., anticancer or antiepileptic drugs); it may thus be possible to test epigenetic drugs as putative novel drugs for the treatment of diabetes and its complications.

Acknowledgments

Research from the authors included in this review has been supported by grants from the Swedish Research Council, Region Skåne, Malmö University Hospital, the Diabetes Programme at Lund University, and Linne′ Grant (B31 5631/2006). The authors also want to acknowledge grants from the following foundations: Knut and Alice Wallenberg, Novo Nordisk, Söderberg, Bergvall, and Påhlsson.

L.G. has been a consultant for and served on advisory boards for sanfo-aventis, GSK, Novartis, Merck, Tethys Bioscience, and Xoma and received lecture fees from Eli Lilly and Novartis. No other potential conflicts of interest relevant to this article were reported.

REFERENCES

1. Diabetes Genetics Initiative of Broad Institute of Harvard and MIT, Lund University, and Novartis Institutes of BioMedical Research. Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ, Kathiresan S, Hirschhorn JN, Daly MJ, Hughes TE, Groop L, Altshuler D, Almgren P, Florez JC, Meyer J, Ardlie K, Bengtsson Boström K, Isomaa B, Lettre G, Lindblad U, Lyon HN, Melander O, Newton-Cheh C, Nilsson P, Orho-Melander M, Råstam L, Speliotes EK, Taskinen MR, Tuomi T, Guiducci C, Berglund A, Carlson J, Gianniny L, Hackett R, Hall L, Holmkvist J, Laurila E, Sjögren M, Sterner M, Surti A, Svensson M, Svensson M, Tewhey R, Blumenstiel B, Parkin M, Defelice M, Barry R, Brodeur W, Camarata J, Chia N, Fava M, Gibbons J, Handsaker B, Healy C, Nguyen K, Gates C, Sougnez C, Gage D, Nizzari M, Gabriel SB, Chirn GW, Ma Q, Parikh H, Richardson D, Ricke D, Purcell S: Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 2007; 316: 1331– 1336 [Abstract] [Google Scholar]
2. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M: A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 2007; 316: 1341– 1345 [Abstract] [Google Scholar]
3. Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, Boutin P, Vincent D, Belisle A, Hadjadj S, Balkau B, Heude B, Charpentier G, Hudson TJ, Montpetit A, Pshezhetsky AV, Prentki M, Posner BI, Balding DJ, Meyre D, Polychronakos C, Froguel P: A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 2007; 445: 881– 885 [Abstract] [Google Scholar]
4. Zeggini E, Weedon MN, Lindgren CM, Frayling TM, Elliott KS, Lango H, Timpson NJ, Perry JR, Rayner NW, Freathy RM, Barrett JC, Shields B, Morris AP, Ellard S, Groves CJ, Harries LW, Marchini JL, Owen KR, Knight B, Cardon LR, Walker M, Hitman GA, Morris AD, Doney AS, Wellcome Trust Case Control Consortium (WTCCC) McCarthy MI, Hattersley AT: Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science 2007; 316: 1336– 1341 [Abstract] [Google Scholar]
5. Zeggini E, Scott LJ, Saxena R, Voight BF, Marchini JL, Hu T, de Bakker PI, Abecasis GR, Almgren P, Andersen G, Ardlie K, Boström KB, Bergman RN, Bonnycastle LL, Borch-Johnsen K, Burtt NP, Chen H, Chines PS, Daly MJ, Deodhar P, Ding CJ, Doney AS, Duren WL, Elliott KS, Erdos MR, Frayling TM, Freathy RM, Gianniny L, Grallert H, Grarup N, Groves CJ, Guiducci C, Hansen T, Herder C, Hitman GA, Hughes TE, Isomaa B, Jackson AU, Jørgensen T, Kong A, Kubalanza K, Kuruvilla FG, Kuusisto J, Langenberg C, Lango H, Lauritzen T, Li Y, Lindgren CM, Lyssenko V, Marvelle AF, Meisinger C, Midthjell K, Mohlke KL, Morken MA, Morris AD, Narisu N, Nilsson P, Owen KR, Palmer CN, Payne F, Perry JR, Pettersen E, Platou C, Prokopenko I, Qi L, Qin L, Rayner NW, Rees M, Roix JJ, Sandbaek A, Shields B, Sjögren M, Steinthorsdottir V, Stringham HM, Swift AJ, Thorleifsson G, Thorsteinsdottir U, Timpson NJ, Tuomi T, Tuomilehto J, Walker M, Watanabe RM, Weedon MN, Willer CJ, Wellcome Trust Case Control Consortium. Illig T, Hveem K, Hu FB, Laakso M, Stefansson K, Pedersen O, Wareham NJ, Barroso I, Hattersley AT, Collins FS, Groop L, McCarthy MI, Boehnke M, Altshuler D: Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet 2008; 40: 638– 645 [Europe PMC free article] [Abstract] [Google Scholar]
6. Bird A: Perceptions of epigenetics. Nature 2007; 447: 396– 398 [Abstract] [Google Scholar]
7. Cubas P, Vincent C, Coen E: An epigenetic mutation responsible for natural variation in floral symmetry. Nature 1999; 401: 157– 161 [Abstract] [Google Scholar]
8. Chong S, Whitelaw E: Epigenetic germline inheritance. Curr Opin Genet Dev 2004; 14: 692– 696 [Abstract] [Google Scholar]
9. Anway MD, Cupp AS, Uzumcu M, Skinner MK: Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 2005; 308: 1466– 1469 [Abstract] [Google Scholar]
10. Clouaire T, Stancheva I: Methyl-CpG binding proteins: specialized transcriptional repressors or structural components of chromatin? Cell Mol Life Sci 2008; 65: 1509– 1522 [Europe PMC free article] [Abstract] [Google Scholar]
11. Patra SK, Patra A, Rizzi F, Ghosh TC, Bettuzzi S: Demethylation of (cytosine-5-C-methyl) DNA and regulation of transcription in the epigenetic pathways of cancer development. Cancer Metastasis Rev 2008; 27: 315– 334 [Abstract] [Google Scholar]
12. Kouzarides T: Chromatin modifications and their function. Cell 2007; 128: 693– 705 [Abstract] [Google Scholar]
13. Haberland M, Montgomery RL, Olson EN: The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 2009; 10: 32– 42 [Abstract] [Google Scholar]
14. Avvakumov N, Côté J: The MYST family of histone acetyltransferases and their intimate links to cancer. Oncogene 2007; 26: 5395– 5407 [Abstract] [Google Scholar]
15. Shahbazian MD, Grunstein M: Functions of site-specific histone acetylation and deacetylation. Annu Rev Biochem 2007; 76: 75– 100 [Abstract] [Google Scholar]
16. Wang Z, Zang C, Rosenfeld JA, Schones DE, Barski A, Cuddapah S, Cui K, Roh TY, Peng W, Zhang MQ, Zhao K: Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet 2008; 40: 897– 903 [Europe PMC free article] [Abstract] [Google Scholar]
17. Bártová E, Krejcí J, Harnicarová A, Galiová G, Kozubek S: Histone modifications and nuclear architecture: a review. J Histochem Cytochem 2008; 56: 711– 721 [Europe PMC free article] [Abstract] [Google Scholar]
18. Marmorstein R, Trievel RC: Histone modifying enzymes: structures, mechanisms, and specificities. Biochim Biophys Acta 2009; 1789: 58– 68 [Abstract] [Google Scholar]
19. Suzuki MM, Bird A: DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet 2008; 9: 465– 476 [Abstract] [Google Scholar]
20. Lennartsson A, Ekwall K: Histone modification patterns and epigenetic codes. Biochim Biophys Acta 2009; 1790: 863– 868 [Abstract] [Google Scholar]
21. Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Rothman DL, DiPietro L, Cline GW, Shulman GI: Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science 2003; 300: 1140– 1142 [Europe PMC free article] [Abstract] [Google Scholar]
22. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI: Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. N Engl J Med 2004; 350: 664– 671 [Europe PMC free article] [Abstract] [Google Scholar]
23. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn JN, Altshuler D, Groop LC: PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003; 34: 267– 273 [Abstract] [Google Scholar]
24. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, Kashyap S, Miyazaki Y, Kohane I, Costello M, Saccone R, Landaker EJ, Goldfine AB, Mun E, DeFronzo R, Finlayson J, Kahn CR, Mandarino LJ: Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1. Proc Natl Acad Sci U S A 2003; 100: 8466– 8471 [Europe PMC free article] [Abstract] [Google Scholar]
25. Kelley DE, He J, Menshikova EV, Ritov VB: Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 2002; 51: 2944– 2950 [Abstract] [Google Scholar]
26. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster BH, Kelley DE: Deficiency of subsarcolemmal mitochondria in obesity and type 2 diabetes. Diabetes 2005; 54: 8– 14 [Abstract] [Google Scholar]
27. Ling C, Poulsen P, Carlsson E, Ridderstråle M, Almgren P, Wojtaszewski J, Beck-Nielsen H, Groop L, Vaag A: Multiple environmental and genetic factors influence skeletal muscle PGC-1alpha and PGC-1beta gene expression in twins. J Clin Invest 2004; 114: 1518– 1526 [Europe PMC free article] [Abstract] [Google Scholar]
28. Ling C, Wegner L, Andersen G, Almgren P, Hansen T, Pedersen O, Groop L, Vaag A, Poulsen P: Impact of the peroxisome proliferator activated receptor-gamma coactivator-1beta (PGC-1beta) Ala203Pro polymorphism on in vivo metabolism, PGC-1beta expression and fibre type composition in human skeletal muscle. Diabetologia 2007; 50: 1615– 1620 [Abstract] [Google Scholar]
29. Ronn T, Poulsen P, Tuomi T, Isomaa B, Groop L, Vaag A, Ling C: Genetic variation in ATP5O is associated with skeletal muscle ATP50 mRNA expression and glucose uptake in young twins. PLoS ONE 2009; 4: e4793. [Europe PMC free article] [Abstract] [Google Scholar]
30. Kujoth GC, Hiona A, Pugh TD, Someya S, Panzer K, Wohlgemuth SE, Hofer T, Seo AY, Sullivan R, Jobling WA, Morrow JD, Van Remmen H, Sedivy JM, Yamasoba T, Tanokura M, Weindruch R, Leeuwenburgh C, Prolla TA: Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 2005; 309: 481– 484 [Abstract] [Google Scholar]
31. Bua E, Johnson J, Herbst A, Delong B, McKenzie D, Salamat S, Aiken JM: Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. Am J Hum Genet 2006; 79: 469– 480 [Europe PMC free article] [Abstract] [Google Scholar]
32. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M: Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A 2005; 102: 10604– 10609 [Europe PMC free article] [Abstract] [Google Scholar]
33. Ling C, Poulsen P, Simonsson S, Rönn T, Holmkvist J, Almgren P, Hagert P, Nilsson E, Mabey AG, Nilsson P, Vaag A, Groop L: Genetic and epigenetic factors are associated with expression of respiratory chain component NDUFB6 in human skeletal muscle. J Clin Invest 2007; 117: 3427– 3435 [Europe PMC free article] [Abstract] [Google Scholar]
34. Rönn T, Poulsen P, Hansson O, Holmkvist J, Almgren P, Nilsson P, Tuomi T, Isomaa B, Groop L, Vaag A, Ling C: Age influences DNA methylation and gene expression of COX7A1 in human skeletal muscle. Diabetologia 2008; 51: 1159– 1168 [Abstract] [Google Scholar]
35. Caro JF, Triester S, Patel VK, Tapscott EB, Frazier NL, Dohm GL: Liver glucokinase: decreased activity in patients with type II diabetes. Horm Metab Res 1995; 27: 19– 22 [Abstract] [Google Scholar]
36. Vaxillaire M, Froguel P: Monogenic diabetes in the young, pharmacogenetics and relevance to multifactorial forms of type 2 diabetes. Endocr Rev 2008; 29: 254– 264 [Abstract] [Google Scholar]
37. Jiang MH, Fei J, Lan MS, Lu ZP, Liu M, Fan WW, Gao X, Lu DR: Hypermethylation of hepatic Gck promoter in ageing rats contributes to diabetogenic potential. Diabetologia 2008; 51: 1525– 1533 [Abstract] [Google Scholar]
38. Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP: CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A 1999; 96: 8681– 8686 [Europe PMC free article] [Abstract] [Google Scholar]
39. Issa JP: The epigenetics of colorectal cancer. Ann N Y Acad Sci 2000; 910: 140– 153; discussion 153–155 [Abstract] [Google Scholar]
40. Issa JP: Age-related epigenetic changes and the immune system. Clin Immunol 2003; 109: 103– 108 [Abstract] [Google Scholar]
41. Youssef EM, Estecio MR, Issa JP: Methylation and regulation of expression of different retinoic acid receptor beta isoforms in human colon cancer. Cancer Biol Ther 2004; 3: 82– 86 [Abstract] [Google Scholar]
42. Burzynski SR: Aging: gene silencing or gene activation? Med Hypotheses 2005; 64: 201– 208 [Abstract] [Google Scholar]
43. Zhang Z, Deng C, Lu Q, Richardson B: Age-dependent DNA methylation changes in the ITGAL (CD11a) promoter. Mech Ageing Dev 2002; 123: 1257– 1268 [Abstract] [Google Scholar]
44. Wilson VL, Jones PA: DNA methylation decreases in aging but not in immortal cells. Science 1983; 220: 1055– 1057 [Abstract] [Google Scholar]
45. Wilson VL, Smith RA, Ma S, Cutler RG: Genomic 5-methyldeoxycytidine decreases with age. J Biol Chem 1987; 262: 9948– 9951 [Abstract] [Google Scholar]
46. Tateishi K, Okada Y, Kallin EM, Zhang Y: Role of Jhdm2a in regulating metabolic gene expression and obesity resistance. Nature 2009; 458: 757– 761 [Abstract] [Google Scholar]
47. Schwer B, Verdin E: Conserved metabolic regulatory functions of sirtuins. Cell Metab 2008; 7: 104– 112 [Abstract] [Google Scholar]
48. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB: ATP-citrate lyase links cellular metabolism to histone acetylation. Science 2009; 324: 1076– 1080 [Europe PMC free article] [Abstract] [Google Scholar]
49. Melzner I, Scott V, Dorsch K, Fischer P, Wabitsch M, Brüderlein S, Hasel C, Möller P: Leptin gene expression in human preadipocytes is switched on by maturation-induced demethylation of distinct CpGs in its proximal promoter. J Biol Chem 2002; 277: 45420– 45427 [Abstract] [Google Scholar]
50. Yokomori N, Tawata M, Onaya T: DNA demethylation modulates mouse leptin promoter activity during the differentiation of 3T3–L1 cells. Diabetologia 2002; 45: 140– 148 [Abstract] [Google Scholar]
51. Milagro FI, Campión J, García-Díaz DF, Goyenechea E, Paternain L, Martínez JA: High fat diet-induced obesity modifies the methylation pattern of leptin promoter in rats. J Physiol Biochem 2009; 65: 1– 9 [Abstract] [Google Scholar]
52. Duhl DM, Vrieling H, Miller KA, Wolff GL, Barsh GS: Neomorphic agouti mutations in obese yellow mice. Nat Genet 1994; 8: 59– 65 [Abstract] [Google Scholar]
53. Morgan HD, Sutherland HG, Martin DI, Whitelaw E: Epigenetic inheritance at the agouti locus in the mouse. Nat Genet 1999; 23: 314– 318 [Abstract] [Google Scholar]
54. Wolff GL, Kodell RL, Moore SR, Cooney CA: Maternal epigenetics and methyl supplements affect agouti gene expression in Avy/a mice. Faseb J 1998; 12: 949– 957 [Abstract] [Google Scholar]
55. Jirtle RL, Skinner MK: Environmental epigenomics and disease susceptibility. Nat Rev Genet 2007; 8: 253– 262 [Europe PMC free article] [Abstract] [Google Scholar]
56. Cropley JE, Suter CM, Beckman KB, Martin DI: Germ-line epigenetic modification of the murine A vy allele by nutritional supplementation. Proc Natl Acad Sci U S A 2006; 103: 17308– 17312 [Europe PMC free article] [Abstract] [Google Scholar]
57. Park JH, Stoffers DA, Nicholls RD, Simmons RA: Development of type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1. J Clin Invest 2008; 118: 2316– 2324 [Europe PMC free article] [Abstract] [Google Scholar]
58. Jensen CB, Storgaard H, Dela F, Holst JJ, Madsbad S, Vaag AA: Early differential defects of insulin secretion and action in 19-year-old caucasian men who had low birth weight. Diabetes 2002; 51: 1271– 1280 [Abstract] [Google Scholar]
59. Jensen CB, Storgaard H, Madsbad S, Richter EA, 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 2007; 92: 1530– 1534 [Abstract] [Google Scholar]
60. Ravelli AC, van der Meulen JH, Michels RP, Osmond C, Barker DJ, Hales CN, Bleker OP: Glucose tolerance in adults after prenatal exposure to famine. Lancet 1998; 351: 173– 177 [Abstract] [Google Scholar]
61. Lumey LH, Stein AD, Kahn HS, van der Pal-de Bruin KM, Blauw GJ, Zybert PA, Susser ES: Cohort profile: the Dutch Hunger Winter families study. Int J Epidemiol 2007; 36: 1196– 1204 [Abstract] [Google Scholar]
62. Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, Slagboom PE, Lumey LH: Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A 2008; 105: 17046– 17049 [Europe PMC free article] [Abstract] [Google Scholar]
63. Tobi EW, Lumey LH, Talens RP, Kremer D, Putter H, Stein AD, Slagboom PE, Heijmans BT: DNA methylation differences after exposure to prenatal famine are common and timing- and sex-specific. Hum Mol Genet 2009; 18: 4046– 4053 [Europe PMC free article] [Abstract] [Google Scholar]
64. Armitage JA, Pearce AD, Sinclair AJ, Vingrys AJ, Weisinger RS, Weisinger HS: Increased blood pressure later in life may be associated with perinatal n-3 fatty acid deficiency. Lipids 2003; 38: 459– 464 [Abstract] [Google Scholar]
65. Khan IY, Dekou V, Douglas G, Jensen R, Hanson MA, Poston L, Taylor PD: A high-fat diet during rat pregnancy or suckling induces cardiovascular dysfunction in adult offspring. Am J Physiol Regul Integr Comp Physiol 2005; 288: R127– R133 [Abstract] [Google Scholar]
66. Taylor PD, McConnell J, Khan IY, Holemans K, Lawrence KM, Asare-Anane H, Persaud SJ, Jones PM, Petrie L, Hanson MA, Poston L: Impaired glucose homeostasis and mitochondrial abnormalities in offspring of rats fed a fat-rich diet in pregnancy. Am J Physiol Regul Integr Comp Physiol 2005; 288: R134– R139 [Abstract] [Google Scholar]
67. Eriksson KF, Lindgärde F: Poor physical fitness, and impaired early insulin response but late hyperinsulinaemia, as predictors of NIDDM in middle-aged Swedish men. Diabetologia 1996; 39: 573– 579 [Abstract] [Google Scholar]
68. Neufer PD, Dohm GL: Exercise induces a transient increase in transcription of the GLUT-4 gene in skeletal muscle. Am J Physiol 1993; 265: C1597– C1603 [Abstract] [Google Scholar]
69. McGee SL, Hargreaves M: Exercise and skeletal muscle glucose transporter 4 expression: molecular mechanisms. Clin Exp Pharmacol Physiol 2006; 33: 395– 399 [Abstract] [Google Scholar]
70. McGee SL, van Denderen BJ, Howlett KF, Mollica J, Schertzer JD, Kemp BE, Hargreaves M: AMP-activated protein kinase regulates GLUT4 transcription by phosphorylating histone deacetylase 5. Diabetes 2008; 57: 860– 867 [Abstract] [Google Scholar]
71. McGee SL, Hargreaves M: Exercise and myocyte enhancer factor 2 regulation in human skeletal muscle. Diabetes 2004; 53: 1208– 1214 [Abstract] [Google Scholar]
72. McGee SL, Sparling D, Olson AL, Hargreaves M: Exercise increases MEF2- and GEF DNA-binding activity in human skeletal muscle. Faseb J 2006; 20: 348– 349 [Abstract] [Google Scholar]
73. Vissing K, McGee SL, Roepstorff C, Schjerling P, Hargreaves M, Kiens B: Effect of sex differences on human MEF2 regulation during endurance exercise. Am J Physiol Endocrinol Metab 2008; 294: E408– E415 [Abstract] [Google Scholar]
74. Smith JA, Kohn TA, Chetty AK, Ojuka EO: CaMK activation during exercise is required for histone hyperacetylation and MEF2A binding at the MEF2 site on the Glut4 gene. Am J Physiol Endocrinol Metab 2008; 295: E698– E704 [Abstract] [Google Scholar]
75. Parikh H, Nilsson E, Ling C, Poulsen P, Almgren P, Nittby H, Eriksson KF, Vaag A, Groop LC: Molecular correlates for maximal oxygen uptake and type 1 fibers. Am J Physiol Endocrinol Metab 2008; 294: E1152– E1159 [Abstract] [Google Scholar]
76. Wren JD, Garner HR: Data-mining analysis suggests an epigenetic pathogenesis for type 2 diabetes. J Biomed Biotechnol 2005; 2005: 104– 112 [Europe PMC free article] [Abstract] [Google Scholar]
77. Puigserver P, Spiegelman BM: Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator. Endocr Rev 2003; 24: 78– 90 [Abstract] [Google Scholar]
78. Ling C, Del Guerra S, Lupi R, Rönn T, Granhall C, Luthman H, Masiello P, Marchetti P, Groop L, Del Prato S: Epigenetic regulation of PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. Diabetologia 2008; 51: 615– 622 [Europe PMC free article] [Abstract] [Google Scholar]
79. Mutskov V, Raaka BM, Felsenfeld G, Gershengorn MC: The human insulin gene displays transcriptionally active epigenetic marks in islet-derived mesenchymal precursor cells in the absence of insulin expression. Stem Cells 2007; 25: 3223– 3233 [Abstract] [Google Scholar]
80. Chakrabarti SK, Francis J, Ziesmann SM, Garmey JC, Mirmira RG: Covalent histone modifications underlie the developmental regulation of insulin gene transcription in pancreatic beta cells. J Biol Chem 2003; 278: 23617– 23623 [Abstract] [Google Scholar]
81. Haumaitre C, Lenoir O, Scharfmann R: Histone deacetylase inhibitors modify pancreatic cell fate determination and amplify endocrine progenitors. Mol Cell Biol 2008; 28: 6373– 6383 [Europe PMC free article] [Abstract] [Google Scholar]
82. Krishnamurthy J, Ramsey MR, Ligon KL, Torrice C, Koh A, Bonner-Weir S, Sharpless NE: p16INK4a induces an age-dependent decline in islet regenerative potential. Nature 2006; 443: 453– 457 [Abstract] [Google Scholar]
83. Chen H, Gu X, Su IH, Bottino R, Contreras JL, Tarakhovsky A, Kim SK: Polycomb protein Ezh2 regulates pancreatic beta-cell Ink4a/Arf expression and regeneration in diabetes mellitus. Genes Dev 2009; 23: 975– 985 [Europe PMC free article] [Abstract] [Google Scholar]
84. Dhawan S, Tschen SI, Bhushan A: Bmi-1 regulates the Ink4a/Arf locus to control pancreatic beta-cell proliferation. Genes Dev 2009; 23: 906– 911 [Europe PMC free article] [Abstract] [Google Scholar]
85. Pontoglio M, Faust DM, Doyen A, Yaniv M, Weiss MC: Hepatocyte nuclear factor 1alpha gene inactivation impairs chromatin remodeling and demethylation of the phenylalanine hydroxylase gene. Mol Cell Biol 1997; 17: 4948– 4956 [Europe PMC free article] [Abstract] [Google Scholar]
86. Soutoglou E, Papafotiou G, Katrakili N, Talianidis I: Transcriptional activation by hepatocyte nuclear factor-1 requires synergism between multiple coactivator proteins. J Biol Chem 2000; 275: 12515– 12520 [Abstract] [Google Scholar]
87. Párrizas M, Maestro MA, Boj SF, Paniagua A, Casamitjana R, Gomis R, Rivera F, Ferrer J: Hepatic nuclear factor 1-alpha directs nucleosomal hyperacetylation to its tissue-specific transcriptional targets. Mol Cell Biol 2001; 21: 3234– 3243 [Europe PMC free article] [Abstract] [Google Scholar]
88. Ban N, Yamada Y, Someya Y, Miyawaki K, Ihara Y, Hosokawa M, Toyokuni S, Tsuda K, Seino Y: Hepatocyte nuclear factor-1alpha recruits the transcriptional co-activator p300 on the GLUT2 gene promoter. Diabetes 2002; 51: 1409– 1418 [Abstract] [Google Scholar]
89. Kim KA, Kang K, Chi YI, Chang I, Lee MK, Kim KW, Shoelson SE, Lee MS: Identification and functional characterization of a novel mutation of hepatocyte nuclear factor-1alpha gene in a Korean family with MODY3. Diabetologia 2003; 46: 721– 727 [Abstract] [Google Scholar]
90. Barbacci E, Chalkiadaki A, Masdeu C, Haumaitre C, Lokmane L, Loirat C, Cloarec S, Talianidis I, Bellanne-Chantelot C, Cereghini S: HNF1beta/TCF2 mutations impair transactivation potential through altered co-regulator recruitment. Hum Mol Genet 2004; 13: 3139– 3149 [Abstract] [Google Scholar]
91. Qiu Y, Guo M, Huang S, Stein R: Insulin gene transcription is mediated by interactions between the p300 coactivator and PDX-1, BETA2, and E47. Mol Cell Biol 2002; 22: 412– 420 [Europe PMC free article] [Abstract] [Google Scholar]
92. Stanojevic V, Habener JF, Thomas MK: Pancreas duodenum homeobox-1 transcriptional activation requires interactions with p300. Endocrinology 2004; 145: 2918– 2928 [Abstract] [Google Scholar]
93. Mosley AL, Ozcan S: Glucose regulates insulin gene transcription by hyperacetylation of histone h4. J Biol Chem 2003; 278: 19660– 19666 [Abstract] [Google Scholar]
94. Mosley AL, Ozcan S: The pancreatic duodenal homeobox-1 protein (Pdx-1) interacts with histone deacetylases Hdac-1 and Hdac-2 on low levels of glucose. J Biol Chem 2004; 279: 54241– 54247 [Abstract] [Google Scholar]
95. Francis J, Chakrabarti SK, Garmey JC, Mirmira RG: Pdx-1 links histone H3-Lys-4 methylation to RNA polymerase II elongation during activation of insulin transcription. J Biol Chem 2005; 280: 36244– 36253 [Europe PMC free article] [Abstract] [Google Scholar]
96. Malecki MT, Jhala US, Antonellis A, Fields L, Doria A, Orban T, Saad M, Warram JH, Montminy M, Krolewski AS: Mutations in NEUROD1 are associated with the development of type 2 diabetes mellitus. Nat Genet 1999; 23: 323– 328 [Abstract] [Google Scholar]
97. Temple IK, Shield JP: Transient neonatal diabetes, a disorder of imprinting. J Med Genet 2002; 39: 872– 875 [Europe PMC free article] [Abstract] [Google Scholar]
98. Mackay DJ, Callaway JL, Marks SM, White HE, Acerini CL, Boonen SE, Dayanikli P, Firth HV, Goodship JA, Haemers AP, Hahnemann JM, Kordonouri O, Masoud AF, Oestergaard E, Storr J, Ellard S, Hattersley AT, Robinson DO, Temple IK: Hypomethylation of multiple imprinted loci in individuals with transient neonatal diabetes is associated with mutations in ZFP57. Nat Genet 2008; 40: 949– 951 [Abstract] [Google Scholar]
99. Miao F, Gonzalo IG, Lanting L, Natarajan R: In vivo chromatin remodeling events leading to inflammatory gene transcription under diabetic conditions. J Biol Chem 2004; 279: 18091– 18097 [Abstract] [Google Scholar]
100. Hofmann MA, Schiekofer S, Kanitz M, Klevesath MS, Joswig M, Lee V, Morcos M, Tritschler H, Ziegler R, Wahl P, Bierhaus A, Nawroth PP: Insufficient glycemic control increases nuclear factor-kappa B binding activity in peripheral blood mononuclear cells isolated from patients with type 1 diabetes. Diabetes Care 1998; 21: 1310– 1316 [Abstract] [Google Scholar]
101. Shanmugam N, Reddy MA, Guha M, Natarajan R: High glucose-induced expression of proinflammatory cytokine and chemokine genes in monocytic cells. Diabetes 2003; 52: 1256– 1264 [Abstract] [Google Scholar]
102. Li Y, Reddy MA, Miao F, Shanmugam N, Yee JK, Hawkins D, Ren B, Natarajan R: Role of the histone H3 lysine 4 methyltransferase, SET7/9, in the regulation of NF-kappaB-dependent inflammatory genes. Relevance to diabetes and inflammation. J Biol Chem 2008; 283: 26771– 26781 [Europe PMC free article] [Abstract] [Google Scholar]
103. Reddy MA, Villeneuve LM, Wang M, Lanting L, Natarajan R: Role of the lysine-specific demethylase 1 in the proinflammatory phenotype of vascular smooth muscle cells of diabetic mice. Circ Res 2008; 103: 615– 623 [Europe PMC free article] [Abstract] [Google Scholar] Retracted
104. Villeneuve LM, Reddy MA, Lanting LL, Wang M, Meng L, Natarajan R: Epigenetic histone H3 lysine 9 methylation in metabolic memory and inflammatory phenotype of vascular smooth muscle cells in diabetes. Proc Natl Acad Sci U S A 2008; 105: 9047– 9052 [Europe PMC free article] [Abstract] [Google Scholar]
105. Miao F, Smith DD, Zhang L, Min A, Feng W, Natarajan R: Lymphocytes from patients with type 1 diabetes display a distinct profile of chromatin histone H3 lysine 9 dimethylation: an epigenetic study in diabetes. Diabetes 2008; 57: 3189– 3198 [Europe PMC free article] [Abstract] [Google Scholar]
106. Action to Control Cardiovascular Risk in Diabetes Study Group. Gerstein HC, Miller ME, Byington RP, Goff DC, Jr, Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr, Probstfield JL, Simons-Morton DG, Friedewald WT: Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008; 358: 2545– 2559 [Abstract] [Google Scholar]
107. ADVANCE Collaborative Group. Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F: Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 2008; 358: 2560– 2572 [Abstract] [Google Scholar]
108. The relationship of glycemic exposure (HbA1c) to the risk of development and progression of retinopathy in the Diabetes Control and Complications Trial. Diabetes 1995; 44: 968– 983 [Abstract] [Google Scholar]
109. El-Osta A, Brasacchio D, Yao D, Pocai A, Jones PL, Roeder RG, Cooper ME, Brownlee M: Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med 2008; 205: 2409– 2417 [Europe PMC free article] [Abstract] [Google Scholar]
110. Brasacchio D, Okabe J, Tikellis C, Balcerczyk A, George P, Baker EK, Calkin AC, Brownlee M, Cooper ME, El-Osta A: Hyperglycemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes 2009; 58: 1229– 1236 [Europe PMC free article] [Abstract] [Google Scholar]
111. Szyf M: Epigenetics, DNA methylation, and chromatin modifying drugs. Annu Rev Pharmacol Toxicol 2009; 49: 243– 263 [Abstract] [Google Scholar]
112. Bieliauskas AV, Pflum MK: Isoform-selective histone deacetylase inhibitors. Chem Soc Rev 2008; 37: 1402– 1413 [Europe PMC free article] [Abstract] [Google Scholar]
113. Granger A, Abdullah I, Huebner F, Stout A, Wang T, Huebner T, Epstein JA, Gruber PJ: Histone deacetylase inhibition reduces myocardial ischemia-reperfusion injury in mice. Faseb J 2008; 22: 3549– 3560 [Europe PMC free article] [Abstract] [Google Scholar]

Articles from Diabetes are provided here courtesy of American Diabetes Association

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/101798221
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/101798221

Article citations


Go to all (350) article citations

Other citations

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.