Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Obesity (Silver Spring) ; 21(12): E571-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23512687

ABSTRACT

OBJECTIVE: Obesity is linked to both increased metabolic disturbances and increased adipose tissue macrophage infiltration. However, whether macrophage infiltration directly influences human metabolism is unclear. The aim of this study was to investigate if there are obesity-independent links between adipose tissue macrophages and metabolic disturbances. DESIGN AND METHODS: Expression of macrophage markers in adipose tissue was analyzed by DNA microarrays in the SOS Sib Pair study and in patients with type 2 diabetes and a BMI-matched healthy control group. RESULTS: The expression of macrophage markers in adipose tissue was increased in obesity and associated with several metabolic and anthropometric measurements. After adjustment for BMI, the expression remained associated with insulin sensitivity, serum levels of insulin, C-peptide, high density lipoprotein cholesterol (HDL-cholesterol) and triglycerides. In addition, the expression of most macrophage markers was significantly increased in patients with type 2 diabetes compared to the control group. CONCLUSION: Our study shows that infiltration of macrophages in human adipose tissue, estimated by the expression of macrophage markers, is increased in subjects with obesity and diabetes and associated with insulin sensitivity and serum lipid levels independent of BMI. This indicates that adipose tissue macrophages may contribute to the development of insulin resistance and dyslipidemia.


Subject(s)
Adipose Tissue/metabolism , Insulin Resistance/genetics , Macrophages/metabolism , Obesity/blood , Obesity/genetics , Body Mass Index , C-Peptide/blood , Case-Control Studies , Cholesterol, HDL/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Female , Gene Expression , Genetic Markers , Humans , Insulin/blood , Male , Oligonucleotide Array Sequence Analysis , Triglycerides/blood
2.
Diabet Med ; 29(11): 1399-406, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22413950

ABSTRACT

AIMS: Insulin sensitivity and acute insulin response measure key components of Type 2 diabetes aetiology that contribute independently to risk in the Insulin Resistance Atherosclerosis Study. As insulin sensitivity and acute insulin response are not routinely measured in a clinical setting, we evaluated three fasting biomarker models, homeostasis model assessment of insulin sensitivity (HOMA-%S), ß-cell function (HOMA-%B) and a Diabetes Risk Score, as potential surrogates for risk associated with insulin sensitivity, acute insulin response and the interaction of these two measures, the disposition index. METHODS: Models were calculated from baseline plasma biomarker concentrations for 664 participants who underwent a frequently sampled intravenous glucose tolerance test. To assess relationships among biomarker models and test measures, we calculated improvement in risk estimation gained by combining each fasting measure with each frequently sampled intravenous glucose tolerance test measure using logistic regression. RESULTS: The strongest correlates of acute insulin response, insulin sensitivity and disposition index were HOMA-%B (r(s)(2) = 0.23), HOMA-%S (r(s)(2) = 0.48) and Diabetes Risk Score (r(s)(2) = 0.34), respectively. Individual areas under the curves for prediction of diabetes were 0.549 (HOMA-%B), 0.694 (HOMA-%S), 0.700 (insulin sensitivity), 0.714 (acute insulin response), 0.756 (Diabetes Risk Score) and 0.817 (disposition index). Models combining acute insulin response with Diabetes Risk Score (area under the curve 0.798) or HOMA-%S (area under the curve 0.805) nearly equalled disposition index, outperforming other individual measures (P < 0.05). Insulin sensitivity plus Diabetes Risk Score (area under the curve 0.760) was better than insulin sensitivity (P = 0.03), but not better than Diabetes Risk Score alone. HOMA-%S plus insulin sensitivity (area under the curve 0.704) was not significantly better than either alone. CONCLUSIONS: The Diabetes Risk Score and HOMA-%S were excellent surrogates for insulin sensitivity, capturing the predictive power of insulin sensitivity. Diabetes Risk Score captured some of the additional predictive power of acute insulin response, but the HOMA models did not. No fasting model was as predictive as disposition index, but the Diabetes Risk Score was the best surrogate.


Subject(s)
Atherosclerosis/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Insulin Resistance , Insulin/blood , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Biomarkers/blood , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Fasting/blood , Female , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Secretion , Male , Middle Aged , Predictive Value of Tests , Risk Factors , Sensitivity and Specificity
3.
Diabetologia ; 53(4): 679-89, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20043145

ABSTRACT

AIMS/HYPOTHESIS: Exercise training improves glucose homeostasis, but large inter-individual differences are reported, suggesting a role of genetic factors. We investigated whether variants either confirmed or newly identified as diabetes susceptibility variants through genome-wide association studies (GWAS) modulate changes in phenotypes derived from an IVGTT in response to an endurance training programme. METHODS: We analysed eight polymorphisms in seven type 2 diabetes genes (CDKAL1 rs7756992; CDKN2A and CDKN2B rs10811661 and rs564398; HHEX rs7923837; IGF2BP2 rs4402960; KCNJ11 rs5215; PPARG rs1801282; and TCF7L2 rs7903146) in a maximum of 481 sedentary, non-diabetic white individuals, who participated in a 20-week endurance training programme. Associations were tested between the variants and changes in IVGTT-derived phenotypes. RESULTS: The only evidence of association with training response was found with PPARG rs1801282 (Pro12Ala). We observed that Ala carriers experienced greater increase in overall glucose tolerance (Deltaglucose disappearance index Ala/Ala 0.22 +/- 0.22, Pro/Ala 0.14 +/- 0.06, Pro/Pro 0.004 +/- 0.03; p = 0.0008), glucose effectiveness (Ala/Ala 0.28 +/- 0.41, Pro/Ala 0.44 +/- 0.14, Pro/Pro 0.09 +/- 0.06; p = 0.004), acute insulin response to glucose (Ala/Ala 64.21 +/- 37.73, Pro/Ala -11.92 +/- 40.30, Pro/Pro -46.30 +/- 14.70; p = 0.03) and disposition index (Ala/Ala 551.8 +/- 448.5, Pro/Ala 534.6 +/- 218.3, Pro/Pro -7.44 +/- 88.18; p = 0.003). CONCLUSIONS/INTERPRETATION: Compared with Pro/Pro individuals, PPARG Ala carriers experienced greater improvements in glucose and insulin metabolism in response to regular endurance training. However, we did not find evidence of association between type 2 diabetes susceptibility variants recently identified through GWAS and glucose homeostasis response to exercise. Our results extend those of previous studies showing that Ala carriers appear to be more responsive to beneficial health effects of lifestyle interventions.


Subject(s)
Blood Glucose/metabolism , Exercise/physiology , PPAR gamma/genetics , Amino Acid Substitution , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Diabetes Mellitus/psychology , Family , Gene Frequency , Genes, p16 , Genetic Variation , Genotype , Glucose Intolerance/blood , Glucose Intolerance/genetics , Glucose Tolerance Test , Homeostasis , Humans , Insulin-Like Growth Factor Binding Protein 2/genetics , Life Style , Polymorphism, Genetic , Risk Factors
4.
Diabetologia ; 53(2): 281-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19902172

ABSTRACT

AIMS/HYPOTHESIS: The majority of type 2 diabetes genome-wide association studies (GWAS) to date have been performed in European-derived populations and have identified few variants that mediate their effect through insulin resistance. The aim of this study was to evaluate two quantitative, directly assessed measures of insulin resistance, namely insulin sensitivity index (S(I)) and insulin disposition index (DI), in Hispanic-American participants using an agnostic, high-density single nucleotide polymorphism (SNP) scan, and to validate these findings in additional samples. METHODS: A two-stage GWAS was performed in Hispanic-American samples from the Insulin Resistance Atherosclerosis Family Study. In Stage 1, 317,000 SNPs were assessed using 229 DNA samples. SNPs with evidence of association with glucose homeostasis and adiposity traits were then genotyped on the entire set of Hispanic-American samples (n = 1,190). This report focuses on the glucose homeostasis traits: S(I) and DI. RESULTS: Although evidence of association did not reach genome-wide significance (p = 5 x 10(-7)), in the combined analysis SNPs had admixture-adjusted p values of p (ADD) = 0.00010-0.0020 with 8 to 41% differences in genotypic means for S(I) and DI. CONCLUSIONS/INTERPRETATION: Several candidate loci were identified that are nominally associated with S(I) and/or DI in Hispanic-American participants. Replication of these findings in independent cohorts and additional focused analysis of these loci is warranted.


Subject(s)
Atherosclerosis/genetics , Genome-Wide Association Study , Hispanic or Latino/genetics , Insulin Resistance/genetics , Polymorphism, Single Nucleotide , Adult , Chromosome Mapping/methods , DNA/genetics , Diabetes Mellitus, Type 2/genetics , Family , Fasting , Female , Genotype , Glucose/metabolism , Humans , Insulin/blood , Male , Middle Aged , Minority Groups/statistics & numerical data , Reproducibility of Results , United States , White People/genetics
5.
Diabetologia ; 52(7): 1326-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19430760

ABSTRACT

AIMS/HYPOTHESIS: This study sought to identify genes and regions in the human genome that are associated with the acute insulin response to glucose (AIRg), an important predictor of type 2 diabetes, in Hispanic-American participants from the Insulin Resistance Atherosclerosis Family Study (IRAS FS). METHODS: A two-stage genome-wide association scan (GWAS) was performed in IRAS FS Hispanic-American samples. In the first stage, 317K single nucleotide polymorphisms (SNPs) were assessed in 229 Hispanic-American DNA samples from 34 families from San Antonio, TX, USA. SNPs with the most significant associations with AIRg were genotyped in the entire set of IRAS FS Hispanic-American samples (n = 1,190). In chromosomal regions with evidence of association, additional SNPs were genotyped to capture variation in genes. RESULTS: No individual SNP achieved genome-wide levels of significance (p < 5 x 10(-7)); however, two regions (chromosomes 6p21 and 20p11) had multiple highly ranked SNPs that were associated with AIRg. Additional genotyping in these regions supported the initial evidence of variants contributing to variation in AIRg. One region resides in a gene desert between PXT1 and KCTD20 on 6p21, while the region on 20p11 has several viable candidate genes (ENTPD6, PYGB, GINS1 and RP4-691N24.1). CONCLUSIONS/INTERPRETATION: A GWAS in Hispanic-American samples identified several candidate genes and loci that may be associated with AIRg. These associations explain a small component of variation in AIRg. The genes identified are involved in phosphorylation and ion transport, and provide preliminary evidence that these processes are important in beta cell response.


Subject(s)
Atherosclerosis , Genome-Wide Association Study , Hispanic or Latino/genetics , Insulin Resistance/ethnology , Insulin Resistance/genetics , Adult , Atherosclerosis/ethnology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Blood Glucose/metabolism , Female , Genetic Predisposition to Disease/ethnology , Genotype , Humans , Insulin/blood , Insulin-Secreting Cells/physiology , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors , Texas/epidemiology
7.
Diabetologia ; 48(6): 1142-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15868134

ABSTRACT

AIMS/HYPOTHESIS: Impaired insulin secretion, insulin action, insulin-independent glucose effectiveness, glucose tolerance and the associated abnormalities in insulin and glucose metabolism phenotypes are precursors of type 2 diabetes. Genome-wide multipoint variance component linkage scans were carried out using 654 markers to identify quantitative trait loci for insulin sensitivity, acute insulin response to glucose, disposition index and glucose effectiveness training responses in whites and blacks in the HERITAGE Family Study. METHODS: These phenotypes were obtained from an IVGTT with the minimal model. The distributions of insulin sensitivity, acute insulin response to glucose and disposition index training responses (post-training minus baseline) were approximately normalised using a square-root transformation. All phenotypes were adjusted for the effects of age, BMI and their respective baseline values within sex and generation by race prior to linkage scans. RESULTS: In blacks, a promising linkage with a maximum lod score of 3.1 on 19q (54-62 Mb) for glucose effectiveness training response was found. Six interesting linkages with lod scores of at least 1.0 were found for disposition index training response in whites. They included 1p (30 Mb), 3q (152 Mb), 6p (23-42 Mb), 7q (95-96 Mb), 10p (15 Mb) and 12q (119-126 Mb). CONCLUSIONS/INTERPRETATION: Quantitative trait loci for 20 weeks of endurance exercise training responses in insulin action and glucose metabolism phenotypes were found on chromosome 19q as well as 6p and 7q, with nominal (6p, 7q) but consistent (6p) linkages across the races.


Subject(s)
Chromosome Mapping , Genome, Human , Physical Endurance/physiology , Physical Fitness , Prediabetic State/genetics , Adolescent , Adult , Black People , Body Mass Index , Family , Female , Genetic Markers , Humans , Lod Score , Male , Middle Aged , North America , Phenotype , Quantitative Trait Loci , Reference Values , White People
8.
Diabetologia ; 48(5): 967-75, 2005 May.
Article in English | MEDLINE | ID: mdl-15830188

ABSTRACT

AIMS: Glucagon-like peptide-1 (GLP-1) is an insulinotropic hormone released from the gut in response to nutrients. Besides its well-established direct effect on pancreatic beta cells, GLP-1 may also act by activating sensors in the hepatoportal area. We therefore studied the impact of putative GLP-1 sensors in the splanchnic circulation. METHODS: We infused GLP-1 into the portal vein of conscious dogs, while also infusing glucose intraportally or systemically. In the first experiment, we infused glucose intraportally, simulating portal glucose values obtained during a previous mixed-meal test, with or without co-infusion of intraportal GLP-1. In the second experiment, by infusing glucose systemically, with or without intraportal GLP-1, we investigated whether the effects of systemic glucose with or without portal GLP-1 infusion are similar to those observed in the first experiment. RESULTS: Intraportal infusion of GLP-1 and glucose significantly raised peripheral GLP-1 levels, but did not produce an insulin response different from intraportal glucose alone. However, the resulting peripheral glycaemia was significantly lower compared to glucose infusion alone, and there were elevations in glucagon, cortisol and lactate. In contrast to the portal glucose infusions, there were no significant differences in glucose, insulin, glucagon, cortisol or lactate levels between systemic glucose infusion with or without GLP-1. CONCLUSIONS/INTERPRETATION: Portal GLP-1 and portal glucose, but not systemic glucose, can produce decreased peripheral glucose levels independently of hyperinsulinaemia. This suggests that portal GLP-1 and glucose receptors mediate insulin-independent changes in peripheral glycaemia and determine a strong counter-regulatory response, as reflected by changes in glucagon and cortisol.


Subject(s)
Glucagon/pharmacology , Glucose/pharmacology , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Dogs , Drug Synergism , Glucagon/administration & dosage , Glucagon/blood , Glucagon/metabolism , Glucagon-Like Peptide 1 , Glucose/administration & dosage , Hydrocortisone/blood , Infusions, Intravenous , Male , Models, Animal , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Portal Vein , Protein Precursors/administration & dosage , Protein Precursors/metabolism
9.
Int J Obes (Lond) ; 29(1): 67-77, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15534617

ABSTRACT

OBJECTIVE: To conduct linkage analysis for body mass index (BMI, kg/m2), waist-to-hip ratio (WHR), visceral adipose tissue mass (VAT, cm2) and subcutaneous adipose tissue mass (SAT, cm2) using a whole genome scan. DESIGN: Cross-sectional family study. STUDY SUBJECTS: African-American families from Los Angeles (AA, n=21 extended pedigrees) and Hispanic-American families (HA) from San Antonio, TX (HA-SA, n=33 extended pedigrees) and San Luis Valley, CO (HA-SLV, n=12 extended pedigrees), totaling 1049 individuals in the Insulin Resistance and Atherosclerosis (IRAS) Family Study. MEASUREMENTS: VAT and SAT were measured using a computed tomography scan obtained at the fourth and fifth lumbar vertebrae. All phenotypes were adjusted for age, gender, and study center. VAT, SAT, and WHR were analyzed both unadjusted and adjusted for BMI. RESULTS: Significant linkage to BMI was found at D3S2387 (LOD=3.67) in African-Americans, and at D17S1290 in Hispanic-Americans (LOD=2.76). BMI-adjusted WHR was linked to 12q13-21 (D12S297 (LOD=2.67) and D12S1052 (LOD=2.60)) in Hispanic-Americans. The peak LOD score for BMI-adjusted VAT was found at D11S2006 (2.36) in Hispanic families from San Antonio. BMI-adjusted SAT was linked to D5S820 in Hispanic families (LOD=2.64). Evidence supporting linkage of WHR at D11S2006, VAT at D17S1290, and SAT at D1S1609, D3S2387, and D6S1056 was dependent on BMI, such that the LOD scores became nonsignificant after adjustment of these phenotypes for BMI. CONCLUSIONS: Our findings both replicate previous linkage regions and suggest novel regions in the genome that may harbor quantitative trait locis contributing to variation in measures of adiposity.


Subject(s)
Body Mass Index , Obesity/genetics , Quantitative Trait, Heritable , Adipose Tissue/diagnostic imaging , Adult , Black or African American , Cross-Sectional Studies , Female , Genetic Linkage , Genotype , Hispanic or Latino , Humans , Male , Obesity/diagnostic imaging , Phenotype , Tomography, X-Ray Computed , Waist-Hip Ratio
10.
Diabetologia ; 47(10): 1782-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15517149

ABSTRACT

AIMS/HYPOTHESIS: Resistin is a peptide hormone produced by adipocytes that is present at high levels in sera of obese mice and may be involved in glucose homeostasis through regulation of insulin sensitivity. Several studies in humans have found associations between polymorphisms in the resistin gene and obesity, insulin sensitivity and blood pressure. An association between variation in the resistin gene and type 2 diabetes has been reported in some, but not all studies. The aim of this study was to analyse variants of the resistin gene for association with type 2 diabetes and related traits in a Finnish sample. METHODS: In 781 cases with type 2 diabetes, 187 spouse controls and 222 elderly controls of Finnish origin, we genotyped four previously identified non-coding single-nucleotide polymorphisms (SNPs): -420C>G from the promoter region, +156C>T and +298G>A from intron 2, and +1084G>A from the 3' untranslated region. We then tested whether these SNPs were associated with type 2 diabetes and related traits. RESULTS: The SNPs were not significantly associated with type 2 diabetes. However, SNPs -420C>G, +156C>T and +298G>A and the common haplotype for these three markers were associated with increased values of weight-related traits and diastolic blood pressure in cases, lower weight in elderly control subjects, and lower insulin sensitivity and greater acute insulin response in spouses. Furthermore, the +1084G allele was associated with lower HDL cholesterol in both cases and controls, higher systolic blood pressure and waist circumference in cases, and greater acute insulin response in spouse controls. CONCLUSIONS/INTERPRETATION: Our results add to growing evidence that resistin is associated with variation in weight, fat distribution and insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Variation , Hormones, Ectopic/genetics , Insulin/genetics , Obesity/epidemiology , Polymorphism, Single Nucleotide , 3' Untranslated Regions/genetics , Age of Onset , Aged , Algorithms , Cohort Studies , Female , Finland/epidemiology , Genotype , Humans , Introns/genetics , Male , Middle Aged , Obesity/genetics , Phenotype , Promoter Regions, Genetic/genetics , Resistin
11.
Metabolism ; 53(9): 1202-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15334385

ABSTRACT

As small increments in insulin concentration profoundly affect lipolysis, our goal was to describe the free fatty acid (FFA) profile during the frequently sampled intravenous glucose tolerance test (FSIGT) and determine if both endogenous and exogenous insulin influenced the FFA profile. Thirteen subjects had both a glucose-only (GO-FSIGT) and insulin-modified FSIGT (IM-FSIGT). Both protocols were of 6 hours duration. At baseline an intravenous glucose bolus (0.3 g/kg) was given. In the IM-FSIGT, insulin was infused from 20 to 25 minutes (4 mU/kg. min). Six additional subjects had both an IM-FSIGT and a normal saline study (NS-Study). For the NS-Study, normal saline solution was infused instead of glucose and insulin. Fasting glucose, insulin, FFA and epinephrine concentrations were similar for all tests. Endogenous insulin peaked at 4 +/- 1 minute in both FSIGTs. The mean calculated peak time of exogenous insulin in the IM-FSIGT was 26 +/- 1 minute. Glucose concentrations were lower and epinephrine concentrations higher in the IM-FSIGT versus GO-FSIGT. During the FSIGTs, the FFA time course revealed four distinct phases, which did not differ between protocols. In phase I (0 to 11 minutes), FFA levels remained near basal (491 +/- 183 micromol/L); in phase II (11 to 79 minutes), FFA levels declined achieving a nadir of 139 +/- 63 micromol/L; in phase III (79 to 188 minutes), FFA levels rose linearly and reattained basal levels; and in phase IV (188 to 360 minutes), FFA levels rose above basal and plateaued at 732 +/- 214 micromol/L (P <.001). In the NS-Study, FFA levels remained near baseline (388 +/- 118 mEq/L) until 180 minutes and then trended upward to 618 +/- 258 micromol/L at 360 minutes. FFA concentrations from 180 to 360 minutes did not differ in the IM-FSIGT versus NS-Study. As the 4 FFA phases did not differ between protocols, the insulin effect on FFA levels in the FSIGT can be attributed to endogenous insulin. But the similarity in FFA levels from 180 to 360 minutes in the IM-FSIGT and NS-Study suggests diurnal variation and not a dynamic related to insulin or the FSIGT protocol is responsible for the final suprabasal FFA plateau.


Subject(s)
Fatty Acids, Nonesterified/blood , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Adult , Blood Glucose/metabolism , Body Mass Index , Epinephrine/blood , Female , Humans , Hypoglycemic Agents/blood , Immunochemistry , Insulin/blood , Luminescent Measurements , Male
12.
Eur J Clin Invest ; 32 Suppl 3: 35-45, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12028373

ABSTRACT

Insulin resistance and beta-cell dysfunction have important roles in the pathogenesis and evolution of type 2 diabetes. The development of precise methods to measure these factors has helped us to define the relationship between them and evidence is reviewed that changes in insulin sensitivity are compensated by inverse changes in beta-cell responsiveness such that the product of insulin sensitivity and insulin secretion (the disposition index) remains constant. While the disposition index promises to be a useful tool to predict individuals at high risk of developing type 2 diabetes, other factors that contribute to beta-cell dysfunction and mark disease onset and progression include impairments in proinsulin processing and insulin secretion, decreased beta-cell mass and islet amyloid deposition. Emerging data indicate that anti-diabetic agents, such as the thiazolidinediones that simultaneously target insulin resistance and beta-cell dysfunction, may have a beneficial impact on disease onset and progression. Several landmark clinical studies are underway to investigate if their initial promise is supported by data from large-scale trials.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance/physiology , Islets of Langerhans/physiopathology , Amyloid/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/therapy , Disease Progression , Humans , Insulin/metabolism , Risk Factors
13.
Clin Genet ; 60(1): 22-30, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11531966

ABSTRACT

Type 2 diabetes mellitus (T2DM), characterized by hyperglycemia, is a complex disease primarily caused by impairment in insulin sensitivity (SI) and insulin secretion. While a strong genetic component for T2DM is well established, there are few reports on racial differences in the magnitude of the genetic effects of T2DM and indices of glucose and insulin metabolism. We report here on the familial resemblance for traits related to glucose metabolism at pre-exercise training levels in 492 members from 99 sedentary White families and 259 members from 108 Black families participating in the multicenter HERITAGE Family Study. All these traits were obtained from the frequently sampled intravenous glucose tolerance test (IVGTT). They include glucose disappearance index (Kg), an overall index for glucose tolerance, acute insulin response to glucose (AIR(Glucose)) which is an index for insulin secretion, and those derived from the minimal model including SI and the disposition index (DI). DI, derived as the product of SI and AIR(Glucose), is a measure of the activity of the B-cells adjusted for insulin resistance. After adjustment for age, sex, and body mass index, the maximal heritability estimates in Blacks (Whites) are 48+/-14% (25+/-8%) for Kg, 44+/-14% (46+/-8%) for AIR(Glucose), 38+/-12% (44+/-8%) for SI and 32+/-14% (24+/-8%) for DI. Interestingly, Blacks have higher heritability for overall glucose tolerance than Whites but there is no race difference in heritability estimates for insulin sensitivity or insulin secretion.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Adult , Analysis of Variance , Black People/genetics , Blood Glucose/metabolism , Body Mass Index , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Exercise/physiology , Family Health , Glucose/administration & dosage , Glucose Tolerance Test , Humans , Infant , Male , Middle Aged , Physical Endurance/genetics , Physical Endurance/physiology , White People/genetics
14.
Am J Physiol Endocrinol Metab ; 281(4): E757-65, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11551852

ABSTRACT

In a longitudinal study of the effects of moderate (70%) dietary restriction (DR) on aging, plasma glucose and insulin concentrations were measured from semiannual, frequently sampled intravenous glucose tolerance tests (FSIGTT) in 30 adult male rhesus monkeys. FSIGTT data were analyzed with Bergman's minimal model, and analysis of covariance revealed that restricted (R) monkeys exhibited increased insulin sensitivity (S(I), P < 0.001) and plasma glucose disappearance rate (K(G), P = 0.015), and reduced fasting plasma insulin (I(b), P < 0.001) and insulin response to glucose (AIR(G), P = 0.023) compared with control (C; ad libitum-fed) monkeys. DR reduced the baseline fasting hyperinsulinemia of two R monkeys, whereas four C monkeys have maintained from baseline, or subsequently developed, fasting hyperinsulinemia; one has progressed to diabetes. Compared with only the normoinsulinemic C monkeys, R monkeys exhibited similarly improved FSIGTT and minimal-model parameters. Thus chronic DR not only has protected against the development of insulin resistance in aging rhesus monkeys, but has also improved glucoregulatory parameters compared with those of otherwise normoinsulinemic monkeys.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, Reducing , Energy Intake , Hyperinsulinism/physiopathology , Insulin Resistance/physiology , Insulin/blood , Macaca mulatta/growth & development , Aging , Analysis of Variance , Animals , Body Mass Index , Body Weight , Disease Progression , Fasting , Glucose Tolerance Test , Homeostasis , Insulin/metabolism , Insulin Secretion , Male , Models, Biological , Time Factors
15.
Obes Res ; 9(8): 423-31, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11500522

ABSTRACT

OBJECTIVE: To examine whether total body fat (FAT) in general or visceral fat (VFAT) in particular is associated with greater metabolic risk in white and African American children. RESEARCH METHODS AND PROCEDURES: A total of 68 white and 51 African American children had measures of insulin sensitivity (Si) and acute insulin response (AIR) by a frequently sampled intravenous glucose tolerance test, total body fat by DXA and abdominal fat distribution (visceral vs. subcutaneous) by computed tomography. The influence of FAT and VFAT on insulin parameters were examined by comparing subgroups of children with high or low FAT vs. high or low VFAT and by multiple regression analysis. RESULTS: In whites, fasting insulin, Si, and AIR were significantly influenced by FAT, but not VFAT (e.g., for Si, 9.8 +/- 0.8 in low FAT vs. 4.6 +/- 0.7 x 10(-4)/min/[microIU/mL[ in high FAT, p < 0.05; 6.8 +/- 0.7 in low VFAT vs. 7.5 +/- 0.8 x 10(-4)/min/[microIU/mL] in high VFAT, p > 0.1). In African Americans, fasting insulin and Si were also primarily influenced by FAT (e.g., for Si, 4.9 +/- 0.4 in low FAT vs. 2.8 +/- 0.5 x 10(-4)/min/[microIU/mL] in high FAT, p < 0.05) but not by VFAT, and there were no significant effects of either fat compartment on AIR. In multiple regression analysis, Si was significantly influenced by FAT (negative effect), ethnicity (lower in African Americans), and gender (lower in females), whereas fasting insulin was significantly influenced by VFAT (positive effect), ethnicity (higher in African Americans), and fat free mass (positive effect). DISCUSSION: Body fat in general is the predominant factor influencing Si, but VFAT may have additional effects on fasting insulin. The lack of major effects of VFAT on Si in children may be explained by lower levels of VFAT or because VFAT affects aspects of whole body insulin action that are not measured by the minimal model.


Subject(s)
Adipose Tissue/metabolism , Black People , Insulin/metabolism , Obesity/physiopathology , White People , Adipose Tissue/anatomy & histology , Body Composition , Child , Female , Glucose Tolerance Test , Humans , Insulin/blood , Insulin Resistance , Insulin Secretion , Male , Obesity/genetics , Obesity/metabolism , Regression Analysis , Viscera
16.
Am J Physiol Endocrinol Metab ; 281(2): E375-83, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11440915

ABSTRACT

It has been proposed that the glycogenolytic and gluconeogenic pathways contributing to endogenous glucose production are interrelated. Thus a change in one source of glucose 6-phosphate might be compensated for by an inverse change in the other pathway. We therefore investigated the effects of 1,4-dideoxy-1,4-imino-D-arabinitol (DAB), a potent glycogen phosphorylase inhibitor, on glucose production in fasted conscious dogs. When dogs were treated acutely with high glucagon, glucose production rose from 1.93 +/- 0.14 to 3.07 +/- 0.37 mg x kg(-1) x min(-1) (P < 0.01). When dogs were treated acutely with DAB in addition to high glucagon infusion, the stimulation of the glycogenolytic rate was completely suppressed. Glucose production rose from 1.85 +/- 0.20 to 2.41 +/- 0.17 mg x kg(-1) x min(-1) (P < 0.05), which was due to the increase in gluconeogenesis from 0.93 +/- 0.09 to 1.54 +/- 0.08 mg x kg(-1) x min(-1) (P < 0.001). In conclusion, infusion of DAB inhibited glycogenolysis; however, the absolute contribution of gluconeogenesis to glucose production was not affected. These results suggest that inhibition of glycogenolysis could be an effective antidiabetic treatment.


Subject(s)
Glycogen/metabolism , Liver/metabolism , Animals , Arabinose , Blood Glucose/drug effects , Dogs , Enzyme Inhibitors/pharmacology , Glucagon/administration & dosage , Gluconeogenesis/drug effects , Glucose Clamp Technique , Hydrolysis/drug effects , Imino Furanoses , Infusions, Intravenous , Liver/drug effects , Male , Phosphorylases/antagonists & inhibitors , Portal Vein , Sugar Alcohols/administration & dosage
17.
Genome Res ; 11(7): 1221-6, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435404

ABSTRACT

Linkage disequilibrium (LD) is a proven tool for evaluating population structure and localizing genes for monogenic disorders. LD-based methods may also help localize genes for complex traits. We evaluated marker-marker LD using 43 microsatellite markers spanning chromosome 20 with an average density of 2.3 cM. We studied 837 individuals affected with type 2 diabetes and 386 mostly unaffected spouse controls. A test of homogeneity between the affected individuals and their spouses showed no difference, allowing the 1223 individuals to be analyzed together. Significant (P < 0.01) LD was observed using a likelihood ratio test in all (11/11) marker pairs within 1 cM, 78% (25/32) of pairs 1-3 cM apart, and 39% (7/18) of pairs 3-4 cM apart, but for only 12 of 842 pairs more than 4 cM apart. We used the human genome project working draft sequence to estimate kilobase (kb) intermarker distances, and observed highly significant LD (P < 10(-10)) for all six marker pairs up to 350 kb apart, although the correlation of LD with cM is slightly better than the correlation with megabases. These data suggest that microsatellites present at 1-cM density are sufficient to observe marker-marker LD in the Finnish population.


Subject(s)
Chromosomes, Human, Pair 20/genetics , Linkage Disequilibrium/genetics , Microsatellite Repeats/genetics , Alleles , Chromosome Mapping , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Finland/epidemiology , Genotype , Haplotypes , Humans
18.
Diabetes Metab Res Rev ; 17(2): 137-45, 2001.
Article in English | MEDLINE | ID: mdl-11307179

ABSTRACT

BACKGROUND: This work was conducted to evaluate associations of insulin secretion with overall and central obesity, dietary fats, physical activity, and alcohol. METHODS: A frequently sampled intravenous glucose tolerance test (FSIGT) was used to assess acute insulin response to glucose (AIR) and insulin sensitivity (S(I)) among adult participants (n=675 with normal, NGT; n=332 with impaired glucose tolerance, IGT) in the Insulin Resistance Atherosclerosis Study (IRAS). Disposition index (DI) was calculated as the sum of the log-transformed AIR and S(I) to reflect pancreatic compensation for insulin resistance. Obesity was measured as body mass index (kg/m(2), BMI) and central fat distribution by waist circumference (cm). Dietary fat intake (total, saturated, polyunsaturated, oleic acid), physical activity, and alcohol intake were assessed by standardized interview. RESULTS: In unadjusted analyses, BMI and waist were each positively correlated with AIR among NGTs (r=0.26 and 0.23, respectively; p<0.0001) but correlations were weaker among the IGTs (r=0.10, NS; r=0.13, p<0.05 for BMI and waist, respectively). BMI and waist were inversely correlated with DI among NGTs (r=-0.13 and -0.20, respectively; p<0.0001) and among IGTs (r=-0.20 and -0.19, respectively, p<0.0001). Dietary fat variables were positively related, and alcohol was inversely related, to AIR among NGTs (p<0.01) but not among IGTs. With all factors considered simultaneously in a pooled analysis of IGTs and NGTs, waist, but not BMI, was positively associated with AIR (p<0.001) and inversely associated with DI (p<0.01). None of the behavioral variables were independently related to either outcome. CONCLUSION: Among non-diabetic patients, central obesity appears to be related to higher insulin secretion, but to lower capacity of the pancreas to respond to the ambient insulin resistance.


Subject(s)
Arteriosclerosis/epidemiology , Blood Glucose/metabolism , Glucose Intolerance/physiopathology , Insulin Resistance , Insulin/metabolism , Life Style , Obesity/physiopathology , Adult , Black or African American , Alcohol Drinking , Body Mass Index , Dietary Fats , Energy Metabolism , Exercise , Female , Glucose Intolerance/blood , Glucose Tolerance Test , Hispanic or Latino , Humans , Insulin/blood , Insulin Secretion , Leisure Activities , Male , Middle Aged , Obesity/blood , Risk Factors , Smoking , United States , White People
19.
Diabetes ; 50(4): 886-90, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11289057

ABSTRACT

Recent studies have identified a common proline-to-alanine substitution (Pro12Ala) in the peroxisome proliferator-activated receptor-gamma2 (PPAR-gamma2), a nuclear receptor that regulates adipocyte differentiation and possibly insulin sensitivity. The Pro12Ala variant has been associated in some studies with diabetes-related traits and/or protection against type 2 diabetes. We examined this variant in 935 Finnish subjects, including 522 subjects with type 2 diabetes, 193 nondiabetic spouses, and 220 elderly nondiabetic control subjects. The frequency of the Pro12Ala variant was significantly lower in diabetic subjects than in nondiabetic subjects (0.15 vs. 0.21; P = 0.001). We also compared diabetes-related traits between subjects with and without the Pro12Ala variant within subgroups. Among diabetic subjects, the variant was associated with greater weight gain after age 20 years (P = 0.023) and lower triglyceride levels (P = 0.033). Diastolic blood pressure was higher in grossly obese (BMI >40 kg/m2) diabetic subjects with the variant. In nondiabetic spouses, the variant was associated with higher fasting insulin (P = 0.033), systolic blood pressure (P = 0.021), and diastolic blood pressure (P = 0.045). These findings support a role for the PPAR-gamma2 Pro12Ala variant in the etiology of type 2 diabetes and the insulin resistance syndrome.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Variation , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Aged , Blood Pressure , Diabetes Mellitus/genetics , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Fasting/blood , Female , Gene Frequency , Humans , Insulin/blood , Male , Middle Aged , Obesity , Reference Values , Triglycerides/blood , Weight Gain
20.
Diabetes ; 50(2): 399-403, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272153

ABSTRACT

Our laboratory has previously demonstrated that hypoglycemic detection occurs in the portal vein, not the liver. To ascertain whether hypoglycemic detection may also occur in the hepatic artery, normoglycemia was established across the liver via a localized hepatic artery glucose infusion. Male mongrel dogs (n = 7) were infused with insulin (5.0 mU x kg(-1) x min(-1)) via the jugular vein to induce systemic hypoglycemia. Animals participated in two hyperinsulinemic-hypoglycemic clamp experiments distinguished by the site of glucose infusion. During the liver irrigation protocol, glucose was infused via the hepatic artery (HA protocol) to maintain liver normoglycemia as systemic glucose concentrations were systematically lowered over 260 min (nadir = 2.2 +/- 0.01 mmol/l). During control experiments, glucose was infused peripherally (PER protocol) to control reductions in blood glucose. Arterial glucose concentrations were not significantly different at any time between the two protocols (P = 0.73). Hepatic artery and liver glucose concentrations were significantly elevated in the HA versus PER protocol throughout the duration of the progressive hyperinsulinemic-hypoglycemic clamp. During the PER protocol, epinephrine and norepinephrine concentrations increased significantly above basal values (0.53 +/- 0.06 and 0.85 +/- 0.2 nmol/l, respectively) to plateaus of 4.4 +/- 0.86 (P = 0.0001) and 3.6 +/- 0.69 nmol/l (P = 0.001), respectively. There were no significant differences between the two protocols in the epinephrine (P = 0.81) and the norepinephrine (P = 0.68) response to hypoglycemia. The current findings indicate that glucosensors important to hypoglycemic detection do not reside in the hepatic artery. Furthermore, these data confirm our previous findings that glucosensors important to hypoglycemic detection are not present in the liver, but are in fact localized to the portal vein.


Subject(s)
Blood Glucose/metabolism , Chemoreceptor Cells/physiology , Glucose/metabolism , Hypoglycemia/metabolism , Liver/metabolism , Animals , Dogs , Epinephrine/blood , Glucose/administration & dosage , Glucose/pharmacology , Hepatic Artery , Injections, Intra-Arterial , Injections, Intravenous , Male , Norepinephrine/blood , Portal Vein
SELECTION OF CITATIONS
SEARCH DETAIL
...