Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Clin Endocrinol Metab ; 107(3): 668-684, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34718610

ABSTRACT

CONTEXT: Genes causing familial forms of diabetes mellitus are only partially known. OBJECTIVE: We set out to identify the genetic cause of hyperglycemia in multigenerational families with an apparent autosomal dominant form of adult-onset diabetes not due to mutations in known monogenic diabetes genes. METHODS: Existing whole-exome sequencing (WES) data were used to identify exonic variants segregating with diabetes in 60 families from the United States and Italy. Functional studies were carried out in vitro (transduced MIN6-K8 cells) and in vivo (Caenorhabditis elegans) to assess the diabetogenic potential of 2 variants in the malate dehydrogenase 2 (MDH2) gene linked with hyperglycemia in 2 of the families. RESULTS: A very rare mutation (p.Arg52Cys) in MDH2 strongly segregated with hyperglycemia in 1 family from the United States. An infrequent MDH2 missense variant (p.Val160Met) also showed disease cosegregation in a family from Italy, although with reduced penetrance. In silico, both Arg52Cys and Val160Met were shown to affect MDH2 protein structure and function. In transfected HepG2 cells, both variants significantly increased MDH2 enzymatic activity, thereby decreasing the NAD+/NADH ratio-a change known to affect insulin signaling and secretion. Stable expression of human wild-type MDH2 in MIN6-K8 cell lines enhanced glucose- and GLP-1-stimulated insulin secretion. This effect was blunted by the Cys52 or Met160 substitutions. Nematodes carrying equivalent changes at the orthologous positions of the mdh-2 gene showed impaired glucose-stimulated insulin secretion. CONCLUSION: Our findings suggest a central role of MDH2 in human glucose homeostasis and indicate that gain of function variants in this gene may be involved in the etiology of familial forms of diabetes.


Subject(s)
Blood Glucose/metabolism , Hyperglycemia/genetics , Malate Dehydrogenase/genetics , Adult , Aged , Aged, 80 and over , Animals , Animals, Genetically Modified , Blood Glucose/analysis , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Case-Control Studies , Cell Line, Tumor , DNA Mutational Analysis , Female , Gain of Function Mutation , Humans , Hyperglycemia/blood , Insulin/analysis , Insulin/metabolism , Insulin Secretion/genetics , Islets of Langerhans , Malate Dehydrogenase/metabolism , Male , Mice , Middle Aged , Models, Animal , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Exome Sequencing
2.
Diabetes ; 68(5): 1084-1093, 2019 05.
Article in English | MEDLINE | ID: mdl-30833467

ABSTRACT

Solute Carrier Family 19 Member 2 (SLC19A2) encodes thiamine transporter 1 (THTR1), which facilitates thiamine transport across the cell membrane. SLC19A2 homozygous mutations have been described as a cause of thiamine-responsive megaloblastic anemia (TRMA), an autosomal recessive syndrome characterized by megaloblastic anemia, diabetes, and sensorineural deafness. Here we describe a loss-of-function SLC19A2 mutation (c.A1063C: p.Lys355Gln) in a family with early-onset diabetes and mild TRMA traits transmitted in an autosomal dominant fashion. We show that SLC19A2-deficient ß-cells are characterized by impaired thiamine uptake, which is not rescued by overexpression of the p.Lys355Gln mutant protein. We further demonstrate that SLC19A2 deficit causes impaired insulin secretion in conjunction with mitochondrial dysfunction, loss of protection against oxidative stress, and cell cycle arrest. These findings link SLC19A2 mutations to autosomal dominant diabetes and suggest a role of SLC19A2 in ß-cell function and survival.


Subject(s)
Anemia, Megaloblastic/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Anemia, Megaloblastic/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Humans , Insulin/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mutation/genetics , Oxidative Stress/genetics , Oxidative Stress/physiology , Thiamine/metabolism
3.
Diabetes Care ; 41(11): 2404-2413, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30262460

ABSTRACT

OBJECTIVE: We evaluated whether the increasing number of genetic loci for coronary artery disease (CAD) identified in the general population could be used to predict the risk of major CAD events (MCE) among participants with type 2 diabetes at high cardiovascular risk. RESEARCH DESIGN AND METHODS: A weighted genetic risk score (GRS) derived from 204 variants representative of all the 160 CAD loci identified in the general population as of December 2017 was calculated in 5,360 and 1,931 white participants in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) and Outcome Reduction With Initial Glargine Intervention (ORIGIN) studies, respectively. The association between GRS and MCE (combining fatal CAD events, nonfatal myocardial infarction, and unstable angina) was assessed by Cox proportional hazards regression. RESULTS: The GRS was associated with MCE risk in both ACCORD and ORIGIN (hazard ratio [HR] per SD 1.27, 95% CI 1.18-1.37, P = 4 × 10-10, and HR per SD 1.35, 95% CI 1.16-1.58, P = 2 × 10-4, respectively). This association was independent from interventions tested in the trials and persisted, though attenuated, after adjustment for classic cardiovascular risk predictors. Adding the GRS to clinical predictors improved incident MCE risk classification (relative integrated discrimination improvement +8%, P = 7 × 10-4). The performance of this GRS was superior to that of GRS based on the smaller number of CAD loci available in previous years. CONCLUSIONS: When combined into a GRS, CAD loci identified in the general population are associated with CAD also in type 2 diabetes. This GRS provides a significant improvement in the ability to correctly predict future MCE, which may increase further with the discovery of new CAD loci.


Subject(s)
Coronary Artery Disease/diagnosis , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/diagnosis , Genetic Association Studies , Genetic Markers , Aged , Cohort Studies , Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/genetics , Diabetic Angiopathies/prevention & control , Female , Fenofibrate/administration & dosage , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Molecular Diagnostic Techniques , Polymorphism, Single Nucleotide , Risk Assessment , Risk Factors , Simvastatin/administration & dosage
4.
Diabetes Care ; 39(11): 1915-1924, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27527847

ABSTRACT

OBJECTIVE: To identify genetic determinants of increased cardiovascular mortality among subjects with type 2 diabetes who underwent intensive glycemic therapy in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. RESEARCH DESIGN AND METHODS: A total of 6.8 million common variants were analyzed for genome-wide association with cardiovascular mortality among 2,667 self-reported white subjects in the ACCORD intensive treatment arm. Significant loci were examined in the entire ACCORD white genetic dataset (n = 5,360) for their modulation of cardiovascular responses to glycemic treatment assignment and in a Joslin Clinic cohort (n = 422) for their interaction with long-term glycemic control on cardiovascular mortality. RESULTS: Two loci, at 10q26 and 5q13, attained genome-wide significance as determinants of cardiovascular mortality in the ACCORD intensive arm (P = 9.8 × 10-9 and P = 2 × 10-8, respectively). A genetic risk score (GRS) defined by the two variants was a significant modulator of cardiovascular mortality response to treatment assignment in the entire ACCORD white genetic dataset. Participants with GRS = 0 experienced a fourfold reduction in cardiovascular mortality in response to intensive treatment (hazard ratio [HR] 0.24 [95% CI 0.07-0.86]), those with GRS = 1 experienced no difference (HR 0.92 [95% CI 0.54-1.56]), and those with GRS ≥2 experienced a threefold increase (HR 3.08 [95% CI 1.82-5.21]). The modulatory effect of the GRS on the association between glycemic control and cardiovascular mortality was confirmed in the Joslin cohort (P = 0.029). CONCLUSIONS: Two genetic variants predict the cardiovascular effects of intensive glycemic control in ACCORD. Further studies are warranted to determine whether these findings can be translated into new strategies to prevent cardiovascular complications of diabetes.


Subject(s)
Cardiovascular Diseases/genetics , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Aged , Blood Glucose/metabolism , Cardiovascular Diseases/mortality , Cohort Studies , Diabetes Mellitus, Type 2/metabolism , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Proportional Hazards Models , Randomized Controlled Trials as Topic , Risk Factors
5.
Atherosclerosis ; 242(1): 334-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26253791

ABSTRACT

OBJECTIVE: Genes that modulate insulin sensitivity may also be involved in shaping the risk of coronary artery disease (CAD). The relatively common TRIB3 Q84R polymorphism (rs2295490) has been associated with abnormal insulin signaling, endothelial dysfunction, insulin resistance, and pro-atherogenic phenotypes. The aim of our study was to investigate the association between low-frequency TRIB3 coding variants and CAD in patients with type 2 diabetes (T2D). METHODS: Three case-control studies for CAD from Italy and US were analyzed, for a total of 1565 individuals, all with type 2 diabetes. Infrequent variants were identified by re-sequencing TRIB3 exons in 140 "extreme cases" and 140 "super-controls" and then genotyped in all study subjects. RESULTS: TRIB3 infrequent variants (n = 8), considered according to a collapsing rare variants framework, were significantly associated with CAD in diabetic patients from Italy (n = 700, OR = 0.43, 95% CI 0.20-0.91; p = 0.027), but not from the US (n = 865, OR = 1.22, 95% CI 0.69-2.18; p = 0.49). In the Italian sets, the association was especially strong among individuals who also carried the common R84 variant. CONCLUSION: Although preliminary, our finding suggests a role of TRIB3 low-frequency variants on CAD among Italian patients with T2D. Further studies are needed to address the role of TRIB3 infrequent variants in other populations of both European and non-European ancestries.


Subject(s)
Cell Cycle Proteins/genetics , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/antagonists & inhibitors , Repressor Proteins/genetics , Aged , Case-Control Studies , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/epidemiology , Exons , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Italy , Logistic Models , Male , Middle Aged , Odds Ratio , Phenotype , Protein Serine-Threonine Kinases/genetics , Risk Factors , United States/epidemiology
6.
Am J Hum Genet ; 97(1): 177-85, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26073777

ABSTRACT

Diabetes mellitus is a highly heterogeneous disorder encompassing several distinct forms with different clinical manifestations including a wide spectrum of age at onset. Despite many advances, the causal genetic defect remains unknown for many subtypes of the disease, including some of those forms with an apparent Mendelian mode of inheritance. Here we report two loss-of-function mutations (c.1655T>A [p.Leu552(∗)] and c.280G>A [p.Asp94Asn]) in the gene for the Adaptor Protein, Phosphotyrosine Interaction, PH domain, and leucine zipper containing 1 (APPL1) that were identified by means of whole-exome sequencing in two large families with a high prevalence of diabetes not due to mutations in known genes involved in maturity onset diabetes of the young (MODY). APPL1 binds to AKT2, a key molecule in the insulin signaling pathway, thereby enhancing insulin-induced AKT2 activation and downstream signaling leading to insulin action and secretion. Both mutations cause APPL1 loss of function. The p.Leu552(∗) alteration totally abolishes APPL1 protein expression in HepG2 transfected cells and the p.Asp94Asn alteration causes significant reduction in the enhancement of the insulin-stimulated AKT2 and GSK3ß phosphorylation that is observed after wild-type APPL1 transfection. These findings-linking APPL1 mutations to familial forms of diabetes-reaffirm the critical role of APPL1 in glucose homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Diabetes Mellitus/genetics , Models, Molecular , Mutation, Missense/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adult , Aged , Female , Hep G2 Cells , Humans , Immunoblotting , Insulin/metabolism , Italy , Male , Middle Aged , Pedigree , Proto-Oncogene Proteins c-akt/metabolism , United States
7.
Atherosclerosis ; 237(2): 639-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25463099

ABSTRACT

OBJECTIVE: We have previously reported the combined effect of SNPs perturbing insulin signaling (ENPP1 K121Q, rs1044498; IRS1 G972R, rs1801278; TRIB3 Q84R, rs2295490) on insulin resistance (IR), type 2 diabetes (T2D) and cardiovascular events. We here investigated whether such a combined effect affects also all-cause mortality in a sample of 1851 Whites of European ancestry. METHODS: We investigated a first sample of 721 patients, 232 deaths, 3389 person-years (py). Replication was assessed in two samples of patients with T2D: the Gargano Mortality Study (GMS) of 714 patients, 127 deaths, 5426 py and the Joslin Kidney Study (JKS) comprising 416 patients, 214 deaths, 5325 py. RESULTS: In the first sample, individuals carrying 1 or ≥ 2 risk alleles had 33% (p = 0.06) and 51% (p = 0.02) increased risk of mortality, as compared with individuals with no risk alleles. A similar, though not significant, trend was obtained in the two replication samples only for subject carrying ≥ 2 risk alleles. In a pooled analysis, individuals carrying ≥ 2 risk alleles had higher mortality rate as compared to those carrying 0 risk alleles (HR = 1.34, 95%CI = 1.08-1.67; p = 0.008), and as compared to those carrying only one risk allele (HR = 1.41, 95%CI = 1.13-1.75; p = 0.002). This association was independent from several possible confounders including sex, age, BMI, hypertension and diabetes status. CONCLUSION: Our data suggest that variants affecting insulin signaling exert a joint effect on all-cause mortality and is consistent with a role of abnormal insulin signaling on mortality risk.


Subject(s)
Insulin/metabolism , Mortality , Aged , Alleles , Atherosclerosis/blood , Atherosclerosis/diagnosis , Atherosclerosis/mortality , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/mortality , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/mortality , Female , Genetic Predisposition to Disease , Genetic Variation , Genotype , Humans , Insulin Resistance , Italy/epidemiology , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Myocardial Infarction/mortality , Polymorphism, Single Nucleotide , Proportional Hazards Models , Prospective Studies , Signal Transduction , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...