Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Front Aging ; 3: 1005322, 2022.
Article in English | MEDLINE | ID: mdl-36313181

ABSTRACT

Despite efficient repair, DNA damage inevitably accumulates with time affecting proper cell function and viability, thereby driving systemic aging. Interventions that either prevent DNA damage or enhance DNA repair are thus likely to extend health- and lifespan across species. However, effective genome-protecting compounds are largely lacking. Here, we use Ercc1 Δ/- and Xpg -/- DNA repair-deficient mutants as two bona fide accelerated aging mouse models to test propitious anti-aging pharmaceutical interventions. Ercc1 Δ/- and Xpg -/- mice show shortened lifespan with accelerated aging across numerous organs and tissues. Previously, we demonstrated that a well-established anti-aging intervention, dietary restriction, reduced DNA damage, and dramatically improved healthspan, strongly extended lifespan, and delayed all aging pathology investigated. Here, we further utilize the short lifespan and early onset of signs of neurological degeneration in Ercc1 Δ/- and Xpg -/- mice to test compounds that influence nutrient sensing (metformin, acarbose, resveratrol), inflammation (aspirin, ibuprofen), mitochondrial processes (idebenone, sodium nitrate, dichloroacetate), glucose homeostasis (trehalose, GlcNAc) and nicotinamide adenine dinucleotide (NAD+) metabolism. While some of the compounds have shown anti-aging features in WT animals, most of them failed to significantly alter lifespan or features of neurodegeneration of our mice. The two NAD+ precursors; nicotinamide riboside (NR) and nicotinic acid (NA), did however induce benefits, consistent with the role of NAD+ in facilitating DNA damage repair. Together, our results illustrate the applicability of short-lived repair mutants for systematic screening of anti-aging interventions capable of reducing DNA damage accumulation.

3.
Clin Sci (Lond) ; 131(15): 1941-1953, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28620011

ABSTRACT

DNA damage is an important contributor to endothelial dysfunction and age-related vascular disease. Recently, we demonstrated in a DNA repair-deficient, prematurely aging mouse model (Ercc1Δ/- mice) that dietary restriction (DR) strongly increases life- and health span, including ameliorating endothelial dysfunction, by preserving genomic integrity. In this mouse mutant displaying prominent accelerated, age-dependent endothelial dysfunction we investigated the signaling pathways involved in improved endothelium-mediated vasodilation by DR, and explore the potential role of the renin-angiotensin system (RAS). Ercc1Δ/- mice showed increased blood pressure and decreased aortic relaxations to acetylcholine (ACh) in organ bath experiments. Nitric oxide (NO) signaling and phospho-Ser1177-eNOS were compromised in Ercc1Δ/- DR improved relaxations by increasing prostaglandin-mediated responses. Increase of cyclo-oxygenase 2 and decrease of phosphodiesterase 4B were identified as potential mechanisms. DR also prevented loss of NO signaling in vascular smooth muscle cells and normalized angiotensin II (Ang II) vasoconstrictions, which were increased in Ercc1Δ/- mice. Ercc1Δ/- mutants showed a loss of Ang II type 2 receptor-mediated counter-regulation of Ang II type 1 receptor-induced vasoconstrictions. Chronic losartan treatment effectively decreased blood pressure, but did not improve endothelium-dependent relaxations. This result might relate to the aging-associated loss of treatment efficacy of RAS blockade with respect to endothelial function improvement. In summary, DR effectively prevents endothelium-dependent vasodilator dysfunction by augmenting prostaglandin-mediated responses, whereas chronic Ang II type 1 receptor blockade is ineffective.


Subject(s)
Aging/metabolism , DNA Damage , Receptor, Angiotensin, Type 1/metabolism , Vascular Diseases/diet therapy , Aging/genetics , Angiotensin II/metabolism , Animals , Blood Pressure , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet , Endonucleases/genetics , Endonucleases/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Receptor, Angiotensin, Type 1/genetics , Vascular Diseases/genetics , Vascular Diseases/metabolism , Vascular Diseases/physiopathology , Vasodilation
4.
PLoS One ; 9(4): e93568, 2014.
Article in English | MEDLINE | ID: mdl-24740260

ABSTRACT

Ku80 and DNA-PKCS are both involved in the repair of double strand DNA breaks via the nonhomologous end joining (NHEJ) pathway. While ku80-/- mice exhibit a severely reduced lifespan and size, this phenotype is less pronounced in dna-pkcs-/- mice. However, these observations are based on independent studies with varying genetic backgrounds. Here, we generated ku80-/-, dna-pkcs-/- and double knock out mice in a C57Bl6/J*FVB F1 hybrid background and compared their lifespan, end of life pathology and mutation frequency in liver and spleen using a lacZ reporter. Our data confirm that inactivation of Ku80 and DNA-PKCS causes reduced lifespan and bodyweights, which is most severe in ku80-/- mice. All mutant mice exhibited a strong increase in lymphoma incidence as well as other aging-related pathology (skin epidermal and adnexal atrophy, trabacular bone reduction, kidney tubular anisokaryosis, and cortical and medullar atrophy) and severe lymphoid depletion. LacZ mutation frequency analysis did not show strong differences in mutation frequencies between knock out and wild type mice. The ku80-/- mice had the most severe phenotype and the Ku80-mutation was dominant over the DNA-PKCS-mutation. Presumably, the more severe degenerative effect of Ku80 inactivation on lifespan compared to DNA-PKCS inactivation is caused by additional functions of Ku80 or activity of free Ku70 since both Ku80 and DNA-PKCS are essential for NHEJ.


Subject(s)
Aging/genetics , Antigens, Nuclear/genetics , DNA End-Joining Repair/genetics , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Animals , Antigens, Nuclear/physiology , Body Weight/genetics , DNA Damage , DNA-Activated Protein Kinase/physiology , DNA-Binding Proteins/physiology , Female , Gene Knockout Techniques , Ku Autoantigen , Longevity/genetics , Male , Mice, Inbred C57BL , Nuclear Proteins/physiology , Phenotype
5.
Sci Rep ; 4: 4091, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24522546

ABSTRACT

Variation in TP53 has been associated with cancer. The pro-allele of a TP53 polymorphism in codon 72 (rs1042522) has been associated with longevity. Recently, we showed that the same allele might be involved in preservation of glucose metabolism, body composition and blood pressure during ageing. Here, we assessed glucose tolerance and body composition in mice carrying the human polymorphism. Our data do not support the previous findings in humans, suggesting that this polymorphism does not play a major role in development of glucose metabolism and body composition during ageing. Alternatively, the mouse model may not be suitable to validate these rs1042522-associated traits up to the age tested.


Subject(s)
Glucose/metabolism , Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Aging , Alleles , Animals , Body Composition , Body Weight , Glucose Tolerance Test , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal
6.
PLoS One ; 9(1): e86358, 2014.
Article in English | MEDLINE | ID: mdl-24466051

ABSTRACT

Ku70 and Ku80 form a heterodimer called Ku that forms a holoenzyme with DNA dependent-protein kinase catalytic subunit (DNA-PKCS) to repair DNA double strand breaks (DSBs) through the nonhomologous end joining (NHEJ) pathway. As expected mutating these genes in mice caused a similar DSB repair-defective phenotype. However, ku70(-/-) cells and ku80(-/-) cells also appeared to have a defect in base excision repair (BER). BER corrects base lesions, apurinic/apyrimidinic (AP) sites and single stand breaks (SSBs) utilizing a variety of proteins including glycosylases, AP endonuclease 1 (APE1) and DNA Polymerase ß (Pol ß). In addition, deleting Ku70 was not equivalent to deleting Ku80 in cells and mice. Therefore, we hypothesized that free Ku70 (not bound to Ku80) and/or free Ku80 (not bound to Ku70) possessed activity that influenced BER. To further test this hypothesis we performed two general sets of experiments. The first set showed that deleting either Ku70 or Ku80 caused an NHEJ-independent defect. We found ku80(-/-) mice had a shorter life span than dna-pkcs(-/-) mice demonstrating a phenotype that was greater than deleting the holoenzyme. We also found Ku70-deletion induced a p53 response that reduced the level of small mutations in the brain suggesting defective BER. We further confirmed that Ku80-deletion impaired BER via a mechanism that was not epistatic to Pol ß. The second set of experiments showed that free Ku70 and free Ku80 could influence BER. We observed that deletion of either Ku70 or Ku80, but not both, increased sensitivity of cells to CRT0044876 (CRT), an agent that interferes with APE1. In addition, free Ku70 and free Ku80 bound to AP sites and in the case of Ku70 inhibited APE1 activity. These observations support a novel role for free Ku70 and free Ku80 in altering BER.


Subject(s)
Antigens, Nuclear/genetics , DNA End-Joining Repair , DNA-Binding Proteins/genetics , Animals , Apurinic Acid/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-Activated Protein Kinase/deficiency , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/deficiency , Epistasis, Genetic , Female , Gene Deletion , Indoles/pharmacology , Ku Autoantigen , Longevity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , Point Mutation , Protein Subunits/deficiency , Protein Subunits/genetics , Radiation Tolerance , Tumor Suppressor Protein p53/metabolism
7.
Aging Cell ; 12(3): 467-77, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23496256

ABSTRACT

Steady-state levels of spontaneous DNA damage, the by-product of normal metabolism and environmental exposure, are controlled by DNA repair pathways. Incomplete repair or an age-related increase in damage production and/or decline in repair could lead to an accumulation of DNA damage, increasing mutation rate, affecting transcription, and/or activating programmed cell death or senescence. These consequences of DNA damage metabolism are highly conserved, and the accumulation of lesions in the DNA of the genome could therefore provide a universal cause of aging. An important corollary of this hypothesis is that defects in DNA repair cause both premature aging and accelerated DNA damage accumulation. While the former has been well-documented, the reliable quantification of the various lesions thought to accumulate in DNA during aging has been a challenge. Here, we quantified inhibition of long-distance PCR as a measure of DNA damage in liver and brain of both normal and prematurely aging, DNA repair defective mice. The results indicate a marginal, but statistically significant, increase in spontaneous DNA damage with age in normal mouse liver but not in brain. Increased levels of DNA damage were not observed in the DNA repair defective mice. We also show that oxidative lesions do not increase with age. These results indicate that neither normal nor premature aging is accompanied by a dramatic increase in DNA damage. This suggests that factors other than DNA damage per se, for example, cellular responses to DNA damage, are responsible for the aging phenotype in mice.


Subject(s)
Aging, Premature/genetics , Aging/genetics , DNA Damage , DNA Repair , Aging/metabolism , Aging, Premature/metabolism , Animals , Brain/metabolism , Cells, Cultured , DNA/genetics , Liver/metabolism , Mice , Oxidation-Reduction
8.
PLoS One ; 7(3): e32148, 2012.
Article in English | MEDLINE | ID: mdl-22403629

ABSTRACT

BACKGROUND: Genome-wide association studies in Japanese populations recently identified common variants in the KCNQ1 gene to be associated with type 2 diabetes. We examined the association of these variants within KCNQ1 with type 2 diabetes in a Dutch population, investigated their effects on insulin secretion and metabolic traits and on the risk of developing complications in type 2 diabetes patients. METHODOLOGY: The KCNQ1 variants rs151290, rs2237892, and rs2237895 were genotyped in a total of 4620 type 2 diabetes patients and 5285 healthy controls from the Netherlands. Data on macrovascular complications, nephropathy and retinopathy were available in a subset of diabetic patients. Association between genotype and insulin secretion/action was assessed in the additional sample of 335 individuals who underwent a hyperglycaemic clamp. PRINCIPAL FINDINGS: We found that all the genotyped KCNQ1 variants were significantly associated with type 2 diabetes in our Dutch population, and the association of rs151290 was the strongest (OR 1.20, 95% CI 1.07-1.35, p = 0.002). The risk C-allele of rs151290 was nominally associated with reduced first-phase glucose-stimulated insulin secretion, while the non-risk T-allele of rs2237892 was significantly correlated with increased second-phase glucose-stimulated insulin secretion (p = 0.025 and 0.0016, respectively). In addition, the risk C-allele of rs2237892 was associated with higher LDL and total cholesterol levels (p = 0.015 and 0.003, respectively). We found no evidence for an association of KCNQ1 with diabetic complications. CONCLUSIONS: Common variants in the KCNQ1 gene are associated with type 2 diabetes in a Dutch population, which can be explained at least in part by an effect on insulin secretion. Furthermore, our data suggest that KCNQ1 is also associated with lipid metabolism.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose Clamp Technique , Hyperglycemia/metabolism , Insulin/metabolism , KCNQ1 Potassium Channel/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Diabetes Complications/genetics , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/physiopathology , Female , Genetic Predisposition to Disease/genetics , Humans , Insulin Secretion , Male , Middle Aged
9.
Eur J Hum Genet ; 20(6): 696-700, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22189267

ABSTRACT

p53 is involved in stress response, metabolism and cardiovascular functioning. The C-allele of rs1042522 in the gene encoding for p53 is associated with longevity and cancer. In this study, we aimed to investigate the association of rs1042522 with changes in blood pressure, BMI and waist circumference using a longitudinal approach. Rs1042522 was analyzed in two longitudinal studies; the Doetinchem Cohort Study (DCS) and the Botnia Prospective Study (BPS). Changes in quantitative traits over time were investigated according to rs1042522 genotypes. An association between rs1042522 and changes in diastolic blood pressure (DBP) in the DCS over time was observed (P=0.004). Furthermore, a borderline significant association was detected with changes in waist circumference over time (P=0.03). These findings were also observed in the BPS (P=0.02 and P=0.05). The C/C-genotype (Pro/Pro) showed the most moderate time-related increase for the studied endpoints. Furthermore, data from the BPS suggested that the C/C-genotype protects against increases in glucose levels over time at 30 and 60 min during oral glucose tolerance test (P=0.01 and P=0.02). In conclusion, we found an association between the C/C-genotype of rs1042522 and changes in DBP and waist circumference over time. This might contribute to the longevity phenotype observed for the same genotype by others.


Subject(s)
Blood Pressure/genetics , Codon , Polymorphism, Genetic , Tumor Suppressor Protein p53/genetics , Adult , Aged , Cohort Studies , Female , Genotype , Humans , Middle Aged , Phenotype
10.
Cell Metab ; 13(1): 80-91, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21195351

ABSTRACT

Type 2 diabetes (T2D) evolves when insulin secretion fails. Insulin release from the pancreatic ß cell is controlled by mitochondrial metabolism, which translates fluctuations in blood glucose into metabolic coupling signals. We identified a common variant (rs950994) in the human transcription factor B1 mitochondrial (TFB1M) gene associated with reduced insulin secretion, elevated postprandial glucose levels, and future risk of T2D. Because islet TFB1M mRNA levels were lower in carriers of the risk allele and correlated with insulin secretion, we examined mice heterozygous for Tfb1m deficiency. These mice displayed lower expression of TFB1M in islets and impaired mitochondrial function and released less insulin in response to glucose in vivo and in vitro. Reducing TFB1M mRNA and protein in clonal ß cells by RNA interference impaired complexes of the mitochondrial oxidative phosphorylation system. Consequently, nutrient-stimulated ATP generation was reduced, leading to perturbed insulin secretion. We conclude that a deficiency in TFB1M and impaired mitochondrial function contribute to the pathogenesis of T2D.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Mitochondrial Proteins/genetics , Transcription Factors/genetics , Animals , Blood Glucose , Cell Line , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression , Gene Silencing , Genetic Loci , Genetic Variation , Humans , Insulin/blood , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Quantitative Trait Loci , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism
11.
J Clin Endocrinol Metab ; 95(4): 1909-15, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20150578

ABSTRACT

CONTEXT: It has been shown that mitochondrial DNA (mtDNA) content is associated with type 2 diabetes (T2D) and related traits. However, empirical data, often based on small samples, did not confirm this observation in all studies. Therefore, the role of mtDNA content in T2D remains elusive. OBJECTIVE: In this study, we assessed the heritability of mtDNA content in buccal cells and analyzed the association of mtDNA content in blood with prevalent and incident T2D. DESIGN AND SETTING: mtDNA content from cells from buccal and blood samples was assessed using a real-time PCR-based assay. Heritability of mtDNA content was estimated in 391 twins from the Netherlands Twin Register. The association with prevalent T2D was tested in a case control study from The Netherlands (n = 329). Incident T2D was analyzed using prospective samples from Finland (n = 444) and The Netherlands (n = 238). MAIN OUTCOME MEASURES: We measured the heritability of mtDNA content and the association of mtDNA content in blood with prevalent and incident T2D. RESULTS: A heritability of mtDNA content of 35% (19-48%) was estimated in the twin families. We did not observe evidence of an association between mtDNA content and prevalent or incident T2D and related traits. Furthermore, we observed a decline in mtDNA content with increasing age that was male specific (P = 0.001). CONCLUSION: In this study, we show that mtDNA content has a heritability of 35% in Dutch twins. There is no association between mtDNA content in blood and prevalent or incident T2D and related traits in our study samples.


Subject(s)
DNA, Mitochondrial/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Mitochondria/metabolism , Case-Control Studies , Diabetes Mellitus, Type 2/epidemiology , Epithelial Cells/metabolism , Female , Finland/epidemiology , Humans , Male , Mouth Mucosa/cytology , Mouth Mucosa/metabolism , Netherlands/epidemiology , Prospective Studies , Registries , Reverse Transcriptase Polymerase Chain Reaction
12.
Diabetes ; 59(1): 287-92, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19808892

ABSTRACT

OBJECTIVE: At least 20 type 2 diabetes loci have now been identified, and several of these are associated with altered beta-cell function. In this study, we have investigated the combined effects of eight known beta-cell loci on insulin secretion stimulated by three different secretagogues during hyperglycemic clamps. RESEARCH DESIGN AND METHODS: A total of 447 subjects originating from four independent studies in the Netherlands and Germany (256 with normal glucose tolerance [NGT]/191 with impaired glucose tolerance [IGT]) underwent a hyperglycemic clamp. A subset had an extended clamp with additional glucagon-like peptide (GLP)-1 and arginine (n = 224). We next genotyped single nucleotide polymorphisms in TCF7L2, KCNJ11, CDKAL1, IGF2BP2, HHEX/IDE, CDKN2A/B, SLC30A8, and MTNR1B and calculated a risk allele score by risk allele counting. RESULTS: The risk allele score was associated with lower first-phase glucose-stimulated insulin secretion (GSIS) (P = 7.1 x 10(-6)). The effect size was equal in subjects with NGT and IGT. We also noted an inverse correlation with the disposition index (P = 1.6 x 10(-3)). When we stratified the study population according to the number of risk alleles into three groups, those with a medium- or high-risk allele score had 9 and 23% lower first-phase GSIS. Second-phase GSIS, insulin sensitivity index and GLP-1, or arginine-stimulated insulin release were not significantly different. CONCLUSIONS: A combined risk allele score for eight known beta-cell genes is associated with the rapid first-phase GSIS and the disposition index. The slower second-phase GSIS, GLP-1, and arginine-stimulated insulin secretion are not associated, suggesting that especially processes involved in rapid granule recruitment and exocytosis are affected in the majority of risk loci.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose/pharmacology , Insulin/metabolism , Polymorphism, Single Nucleotide , Adult , Aged , Alleles , Body Mass Index , Diabetes Mellitus, Type 2/epidemiology , Female , Genotype , Germany/epidemiology , Glucose Clamp Technique , Glucose Intolerance/epidemiology , Glucose Intolerance/genetics , Humans , Hyperglycemia/blood , Hyperglycemia/chemically induced , Insulin Secretion , Male , Middle Aged , Netherlands/epidemiology , Reference Values , Risk Assessment , Risk Factors
13.
Diabetes ; 59(1): 293-301, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19833888

ABSTRACT

OBJECTIVE: Recently, results from a meta-analysis of genome-wide association studies have yielded a number of novel type 2 diabetes loci. However, conflicting results have been published regarding their effects on insulin secretion and insulin sensitivity. In this study we used hyperglycemic clamps with three different stimuli to test associations between these novel loci and various measures of beta-cell function. RESEARCH DESIGN AND METHODS: For this study, 336 participants, 180 normal glucose tolerant and 156 impaired glucose tolerant, underwent a 2-h hyperglycemic clamp. In a subset we also assessed the response to glucagon-like peptide (GLP)-1 and arginine during an extended clamp (n = 123). All subjects were genotyped for gene variants in JAZF1, CDC123/CAMK1D, TSPAN8/LGR5, THADA, ADAMTS9, NOTCH2/ADAMS30, DCD, VEGFA, BCL11A, HNF1B, WFS1, and MTNR1B. RESULTS: Gene variants in CDC123/CAMK1D, ADAMTS9, BCL11A, and MTNR1B affected various aspects of the insulin response to glucose (all P < 6.9 x 10(-3)). The THADA gene variant was associated with lower beta-cell response to GLP-1 and arginine (both P < 1.6 x 10(-3)), suggesting lower beta-cell mass as a possible pathogenic mechanism. Remarkably, we also noted a trend toward an increased insulin response to GLP-1 in carriers of MTNR1B (P = 0.03), which may offer new therapeutic possibilities. The other seven loci were not detectably associated with beta-cell function. CONCLUSIONS: Diabetes risk alleles in CDC123/CAMK1D, THADA, ADAMTS9, BCL11A, and MTNR1B are associated with various specific aspects of beta-cell function. These findings point to a clear diversity in the impact that these various gene variants may have on (dys)function of pancreatic beta-cells.


Subject(s)
ADAM Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Chromosome Mapping , Diabetes Mellitus, Type 2/genetics , Genetic Variation , Insulin-Secreting Cells/physiology , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , ADAMTS9 Protein , Adult , Aged , Carrier State , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Female , Glucose Clamp Technique , Humans , Male , Middle Aged , Repressor Proteins , Risk Assessment
14.
Eur J Hum Genet ; 17(8): 1056-62, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19209188

ABSTRACT

Mitochondria play an important role in many processes, like glucose metabolism, fatty acid oxidation and ATP synthesis. In this study, we aimed to identify association of common polymorphisms in nuclear-encoded genes involved in mitochondrial protein synthesis and biogenesis with type II diabetes mellitus (T2DM) using a two-stage design. In the first stage, we analyzed 62 tagging single nucleotide polymorphisms (SNPs) in the Hoorn study (n=999 participants) covering all common variation in 13 biological candidate genes. These 13 candidate genes were selected from four clusters regarded essential for correct mitochondrial protein synthesis and biogenesis: aminoacyl tRNA synthetases, translation initiation factors, tRNA modifying enzymes and mitochondrial DNA transcription and replication. SNPs showing evidence for association with T2DM were measured in second stage genotyping (n=10164 participants). After a meta-analysis, only one SNP in SIRT4 (rs2522138) remained significant (P=0.01). Extending the second stage with samples from the Danish Steno Study (n=1220 participants) resulted in a common odds ratio (OR) of 0.92 (0.85-1.00), P=0.06. Moreover, in a large meta-analysis of three genome-wide association studies, this SNP was also not associated with T2DM (P=0.72). In conclusion, we did not find evidence for association of common variants in 13 nuclear-encoded mitochondrial proteins with T2DM.


Subject(s)
Cell Nucleus/genetics , Diabetes Mellitus, Type 2/genetics , Genome-Wide Association Study , Mitochondria/metabolism , Protein Biosynthesis/genetics , Aged , Aged, 80 and over , Case-Control Studies , Cohort Studies , Denmark , Diabetes Mellitus, Type 2/metabolism , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
15.
Diabetes ; 55(11): 3193-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065362

ABSTRACT

The short-chain l-3-hydroxyacyl-CoA dehydrogenase (SCHAD) protein is involved in the penultimate step of mitochondrial fatty acid oxidation. Previously, it has been shown that mutations in the corresponding gene (HADHSC) are associated with hyperinsulinism in infancy. The presumed function of the SCHAD enzyme in glucose-stimulated insulin secretion led us to the hypothesis that common variants in HADHSC on chromosome 4q22-26 might be associated with development of type 2 diabetes. In this study, we have performed a large-scale association study in four different cohorts from the Netherlands and Denmark (n = 7,365). Direct sequencing of HADHSC cDNA and databank analysis identified four tagging single nucleotide polymorphisms (SNPs) including one missense variant (P86L). Neither the SNPs nor haplotypes investigated were associated with the disease, enzyme function, or any relevant quantitative measure (all P > 0.1). The present study provides no evidence that the specific HADHSC variants or haplotypes examined do influence susceptibility to develop type 2 diabetes. We conclude that it is unlikely that variation in HADHSC plays a major role in the pathogenesis of type 2 diabetes in the examined cohorts.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , Diabetes Mellitus, Type 2/genetics , Body Mass Index , Case-Control Studies , Databases, Nucleic Acid , Diabetes Mellitus, Type 2/blood , Female , Genetic Predisposition to Disease , Glucose Tolerance Test , Glycated Hemoglobin , Humans , Hyperinsulinism/genetics , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...