Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Curr Res Physiol ; 4: 47-59, 2021.
Article in English | MEDLINE | ID: mdl-34746826

ABSTRACT

µ-Crystallin, encoded by the CRYM gene, binds the thyroid hormones, T3 and T4. Because T3 and T4 are potent regulators of metabolism and gene expression, and CRYM levels in human skeletal muscle can vary widely, we investigated the effects of overexpression of Crym. We generated transgenic mice, Crym tg, that expressed Crym protein specifically in skeletal muscle at levels 2.6-147.5 fold higher than in controls. Muscular functions, Ca2+ transients, contractile force, fatigue, running on treadmills or wheels, were not significantly altered, although T3 levels in tibialis anterior (TA) muscle were elevated ~190-fold and serum T4 was decreased 1.2-fold. Serum T3 and thyroid stimulating hormone (TSH) levels were unaffected. Crym transgenic mice studied in metabolic chambers showed a significant decrease in the respiratory exchange ratio (RER) corresponding to a 13.7% increase in fat utilization as an energy source compared to controls. Female but not male Crym tg mice gained weight more rapidly than controls when fed high fat or high simple carbohydrate diets. Although labeling for myosin heavy chains showed no fiber type differences in TA or soleus muscles, application of machine learning algorithms revealed small but significant morphological differences between Crym tg and control soleus fibers. RNA-seq and gene ontology enrichment analysis showed a significant shift towards genes associated with slower muscle function and its metabolic correlate, ß-oxidation. Protein expression showed a similar shift, though with little overlap. Our study shows that µ-crystallin plays an important role in determining substrate utilization in mammalian muscle and that high levels of µ-crystallin are associated with a shift toward greater fat metabolism.

2.
Am J Physiol Endocrinol Metab ; 308(9): E792-804, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25738781

ABSTRACT

Thromboxane A2, an arachidonic acid-derived eicosanoid generated by thromboxane synthase (TBXAS), plays critical roles in hemostasis and inflammation. However, the contribution of thromboxane A2 to obesity-linked metabolic dysfunction remains incompletely understood. Here, we used in vitro and mouse models to better define the role of TBXAS in metabolic homeostasis. We found that adipose expression of Tbxas and thromboxane A2 receptor (Tbxa2r) was significantly upregulated in genetic and dietary mouse models of obesity and diabetes. Expression of Tbxas and Tbxa2r was detected in adipose stromal cells, including macrophages. Furthermore, stimulation of macrophages with interferon-γ or resistin factors known to be upregulated in obesity induced Tbxas and Tbxa2r expression. Mice lacking Tbxas had similar weight gain, food intake, and energy expenditure. However, loss of Tbxas markedly enhanced insulin sensitivity in mice fed a low-fat diet. Improvement in glucose homeostasis was correlated with the upregulated expression of multiple secreted metabolic regulators (Ctrp3, Ctrp9, and Ctrp12) in the visceral fat depot. Following a challenge with a high-fat diet, Tbxas deficiency led to attenuated adipose tissue fibrosis and reduced circulating IL-6 levels without adipose tissue macrophages being affected; however, these changes were not sufficient to improve whole body insulin action. Together, our results highlight a novel, diet-dependent role for thromboxane A2 in modulating peripheral tissue insulin sensitivity and adipose tissue fibrosis.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Insulin/metabolism , Thromboxane-A Synthase/genetics , 3T3-L1 Cells , Animals , Cells, Cultured , Cytokines/pharmacology , Female , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Regulation/drug effects , Inflammation Mediators/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Thromboxane-A Synthase/deficiency
3.
Microvasc Res ; 97: 55-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25283348

ABSTRACT

Angiogenesis is regulated by hyperglycemic conditions, which can induce cellular stress responses, reactive oxygen species (ROS), and anti-oxidant defenses that modulate intracellular signaling to prevent oxidative damage. The RUNX2 DNA-binding transcription factor is activated by a glucose-mediated intracellular pathway, plays an important role in endothelial cell (EC) function and angiogenesis, and is a target of oxidative stress. RUNX2 DNA-binding and EC differentiation in response to glucose were conserved in ECs from different tissues and inhibited by hyperglycemia, which stimulated ROS production through the aldose reductase glucose-utilization pathway. Furthermore, the redox status of cysteine and methionine residues regulated RUNX2 DNA-binding and reversal of oxidative inhibition was consistent with an endogenous Methionine sulfoxide reductase-A (MsrA) activity. Low molecular weight MsrA substrates and sulfoxide scavengers were potent inhibitors of RUNX2 DNA binding in the absence of oxidative stress, but acted as antioxidants to increase DNA binding in the presence of oxidants. MsrA was associated with RUNX2:DNA complexes, as measured by a sensitive, quantitative DNA-binding ELISA. The related RUNX2 protein family member, RUNX1, which contains an identical DNA-binding domain, was a catalytic substrate of recombinant MsrA. These findings define novel redox pathways involving aldose reductase and MsrA that regulate RUNX2 transcription factor activity and biological function in ECs. Targeting of these pathways could result in more effective strategies to alleviate the vascular dysfunction associated with diabetes or cancer.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , DNA/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Hyperglycemia/metabolism , Neovascularization, Pathologic , Aldehyde Reductase/metabolism , Angiogenesis Inhibitors/pharmacology , Antioxidants/pharmacology , Binding Sites , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hyperglycemia/pathology , Hyperglycemia/physiopathology , Methionine Sulfoxide Reductases/metabolism , Oxidants/pharmacology , Oxidation-Reduction , Oxidative Stress , Phenotype , Signal Transduction , Substrate Specificity , Time Factors
4.
N Engl J Med ; 370(24): 2307-2315, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24848981

ABSTRACT

BACKGROUND: Lipolysis regulates energy homeostasis through the hydrolysis of intracellular triglycerides and the release of fatty acids for use as energy substrates or lipid mediators in cellular processes. Genes encoding proteins that regulate energy homeostasis through lipolysis are thus likely to play an important role in determining susceptibility to metabolic disorders. METHODS: We sequenced 12 lipolytic-pathway genes in Old Order Amish participants whose fasting serum triglyceride levels were at the extremes of the distribution and identified a novel 19-bp frameshift deletion in exon 9 of LIPE, encoding hormone-sensitive lipase (HSL), a key enzyme for lipolysis. We genotyped the deletion in DNA from 2738 Amish participants and performed association analyses to determine the effects of the deletion on metabolic traits. We also obtained biopsy specimens of abdominal subcutaneous adipose tissue from 2 study participants who were homozygous for the deletion (DD genotype), 10 who were heterozygous (ID genotype), and 7 who were noncarriers (II genotype) for assessment of adipose histologic characteristics, lipolysis, enzyme activity, cytokine release, and messenger RNA (mRNA) and protein levels. RESULTS: Carriers of the mutation had dyslipidemia, hepatic steatosis, systemic insulin resistance, and diabetes. In adipose tissue from study participants with the DD genotype, the mutation resulted in the absence of HSL protein, small adipocytes, impaired lipolysis, insulin resistance, and inflammation. Transcription factors responsive to peroxisome-proliferator-activated receptor γ (PPAR-γ) and downstream target genes were down-regulated in adipose tissue from participants with the DD genotype, altering the regulation of pathways influencing adipogenesis, insulin sensitivity, and lipid metabolism. CONCLUSIONS: These findings indicate the physiological significance of HSL in adipocyte function and the regulation of systemic lipid and glucose homeostasis and underscore the severe metabolic consequences of impaired lipolysis. (Funded by the National Institutes of Health and others).


Subject(s)
Diabetes Mellitus, Type 2/genetics , Frameshift Mutation , Genetic Predisposition to Disease , Lipolysis/genetics , Sterol Esterase/genetics , Adult , Aged , Amish/genetics , Diabetes Mellitus, Type 2/metabolism , Dyslipidemias/genetics , Dyslipidemias/metabolism , Female , Heterozygote , Humans , Insulin Resistance/genetics , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Pedigree
5.
J Gerontol A Biol Sci Med Sci ; 69(7): 790-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24357038

ABSTRACT

The purpose of this study was to determine the effects of 6-month aerobic exercise training + weight loss (AEX + WL) on basal and insulin activation of glycogen synthase, basal citrate synthase activity, and Akt and AS160 phosphorylation in older, overweight/obese insulin-resistant men (n = 14; 63 ± 2 years; body mass index, 32 ± kg/m(2)). Muscle samples of the vastus lateralis were collected before and during a 3-hour 80 mU/m(2)/min hyperinsulinemic-euglycemic clamp. AEX + WL increased VO2max by 11% (p < .05) and decreased body weight (-9%, p < .001). AEX + WL increased basal citrate synthase activity by 46% (p < .01) and insulin activation of independent (2.9-fold) and fractional (2.3-fold) activities (both p < .001) of glycogen synthase. AEX + WL had no effect on phosphorylation of Akt or AS160. Glucose utilization (M) improved 25% (p < .01), and the change tended to be related to the increase in insulin activation of glycogen synthase fractional activity (r = .50, p = .08) following AEX + WL. In summary, AEX + WL has a robust effect on insulin activation of skeletal muscle glycogen synthase activity that likely contributes to improved glucose utilization in older insulin-resistant men.


Subject(s)
Exercise/physiology , Glycogen Synthase/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Weight Loss/physiology , Aged , Aging/physiology , Body Composition , Diet, Reducing , GTPase-Activating Proteins/metabolism , Glucose/metabolism , Glucose Clamp Technique , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-akt/metabolism
6.
Endocr Connect ; 2(2): 79-86, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23781323

ABSTRACT

The purpose of this study is to compare central obesity, insulin sensitivity, and cardiovascular disease risk factors between premenopausal and postmenopausal women with a history of gestational diabetes mellitus (GDM), controls, and women with type 2 diabetes (T2DM). Subjects were 73 overweight/obese and sedentary women who had a history of GDM (n=31) and were either premenopausal (n=11, 44±1 years, X±s.e.m.), postmenopausal (n=20, 58±1 years), or without a history of GDM as healthy postmenopausal controls (n=27, 57±1 years) or postmenopausal with T2DM (n=16, 59±1 years). The premenopausal GDM women had higher maximal oxygen uptake and lower visceral fat than the other three groups (P<0.05). BMI, %body fat, subcutaneous abdominal fat, and intramuscular fat did not differ significantly among the four groups. Glucose utilization (M, 3 h 40 mU/m(2) per min hyperinsulinemic-euglycemic clamps) was 27% higher (P=0.05) in pre- than postmenopausal GDM and was not different between premenopausal GDM and postmenopausal controls. M was 28% lower (P=0.06) in postmenopausal GDM than controls and was not significantly different between postmenopausal GDM and T2DM groups. Thus, despite being younger and more physically fit, premenopausal women with prior GDM display similar central obesity, glucose, and metabolic profiles as postmenopausal controls. Postmenopausal women with prior GDM are more insulin resistant than controls of similar age, adiposity, and fitness levels and display comparable glucose utilization rates as similar as women with T2DM suggesting that a prior history of GDM may be an early manifestation of increased risk of later T2DM.

7.
J Bone Miner Res ; 27(2): 331-41, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22028037

ABSTRACT

EIF2AK3 is a type I transmembrane protein that functions as an endoplasmic reticulum (ER) stress sensor to regulate global protein synthesis. Rare mutations in EIF2AK3 cause Wolcott-Rallison syndrome (OMIM 226980), an autosomal recessive disorder characterized by diabetes, epiphyseal dysplasia, osteoporosis, and growth retardation. To investigate the role of common genetic variation in EIF2AK3 as a determinant of bone mineral density (BMD) and osteoporosis, we sequenced all exons and flanking regions, then genotyped six potentially functional single nucleotide polymorphisms (SNPs) in this gene in 997 Amish subjects for association analysis, and attempted replication in 887 Mexican Americans. We found that the minor allele of a nonsynonymous SNP rs13045 had borderline associations with decreased forearm BMD in both discovery and replication cohorts (unadjusted p = 0.036 and ß = -0.007 for the Amish; unadjusted p = 0.031 and ß = -0.008 for Mexican Americans). A meta-analysis indicated this association achieved statistical significance in the combined sample (unadjusted p = 0.003; Bonferroni corrected p = 0.009). Rs13045 and three other potentially functional SNPs, a promoter SNP (rs6547787) and two nonsynonymous SNPs (rs867529 and rs1805165), formed two haplotypes: a low-BMD associated haplotype, denoted haplotype B [minor allele frequency (MAF) = 0.311] and a common haplotype A (MAF = 0.676). There were no differences in mRNA expression in lymphoblastoid cell lines between the two haplotypes. However, after treating lymphoblastoid cell lines with thapsigargin to induce ER stress, cell lines with haplotype B showed increased sensitivity to ER stress (p = 0.014) compared with cell lines with haplotype A. Taken together, our results suggest that common nonsynonymous sequence variants in EIF2AK3 have a modest effect on ER stress response and may contribute to the risk for low BMD through this mechanism.


Subject(s)
Bone Density/genetics , Endoplasmic Reticulum Stress/genetics , Genetic Association Studies , Haplotypes/genetics , eIF-2 Kinase/genetics , Adult , Alleles , Amino Acid Sequence , Amish/genetics , Cell Line , Female , Heterozygote , Humans , Linkage Disequilibrium/genetics , Male , Mexican Americans/genetics , Middle Aged , Molecular Sequence Data , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA , Signal Transduction/genetics , eIF-2 Kinase/chemistry
8.
Bone ; 47(2): 272-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20450993

ABSTRACT

We previously detected strong evidence for linkage of forearm bone mineral density (BMD) to chromosome 4p (lod=4.3) in a set of 29 large Mexican American families. Fibroblast growth factor binding protein 1 (FGFBP1) is a strong candidate gene for bone homeostasis in this region. We sequenced the coding region of FGFBP1 in a subset of our Mexican American study population and performed association studies with BMD on SNPs genotyped in the entire cohort. We then attempted to replicate these findings in an independent study cohort and performed in vitro functional studies on replicated, potentially functional polymorphisms using a luciferase reporter construct to evaluate influence on gene expression. Several SNPs spanning the gene, all in one large block of linkage disequilibrium, were significantly associated with BMD at various skeletal sites (n=872, p=0.001-0.04). The associations were then replicated in an independent population of European ancestry (n=972; p=0.02-0.04). Sex-stratified association analyses in both study populations suggest this association is much stronger in men. Subsequent luciferase reporter gene assays revealed marked differences in FGFBP1 expression among the three common haplotypes. Further experiments revealed that a promoter polymorphism, rs12503796, results in decreased expression of FGFBP1 and inhibits upregulation of the gene by testosterone in vitro. Collectively, these findings suggest that sequence variation in FGFBP1 may contribute to variation in BMD, possibly influencing osteoporosis risk.


Subject(s)
Bone Density/genetics , Carrier Proteins/genetics , Gene Expression Regulation , Polymorphism, Single Nucleotide/genetics , Adult , Binding Sites , Female , Haplotypes/genetics , Humans , Intercellular Signaling Peptides and Proteins , Linkage Disequilibrium/genetics , Male , Mexican Americans/genetics , Middle Aged , Reproducibility of Results , Transcription Factors/metabolism , White People/genetics
9.
Hum Genet ; 126(4): 567-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19526250

ABSTRACT

Postprandial triglyceridemia is an emerging risk factor for cardiovascular disease. However, most of the genes that influence postprandial triglyceridemia are not known. We evaluated whether a common nonsynonymous SNP rs1260326/P446L in the glucokinase regulatory protein (GCKR) gene influenced variation in the postprandial lipid response after a high-fat challenge in seven hundred and seventy participants in the Amish HAPI Heart Study who underwent an oral high-fat challenge and had blood samples taken in the fasting state and during the postprandial phase at 1, 2, 3, 4, and 6 h. We found that the minor T allele at rs1260326 was associated with significantly higher fasting TG levels after adjusting for age, sex, and family structure (P (a) = 0.06 for additive model, and P (r) = 0.0003 for recessive model). During the fat challenge, the T allele was associated with significantly higher maximum TG level (P (a) = 0.006), incremental maximum TG level (P (a) = 0.006), TG area under the curve (P (a) = 0.02) and incremental TG area under the curve (P (a) = 0.03). Our data indicate that the rs1260326 T allele of GCKR is associated with both higher fasting levels of TG as well as the postprandial TG response, which may result in higher atherogenic risk.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Dietary Fats/administration & dosage , Glucokinase/genetics , Lipids/analysis , Polymorphism, Single Nucleotide/genetics , Postprandial Period/genetics , Adult , Fasting , Female , Genotype , Humans , Male , Middle Aged , Phenotype , Risk Factors , Triglycerides/metabolism
10.
Science ; 322(5908): 1702-5, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19074352

ABSTRACT

Apolipoprotein C-III (apoC-III) inhibits triglyceride hydrolysis and has been implicated in coronary artery disease. Through a genome-wide association study, we have found that about 5% of the Lancaster Amish are heterozygous carriers of a null mutation (R19X) in the gene encoding apoC-III (APOC3) and, as a result, express half the amount of apoC-III present in noncarriers. Mutation carriers compared with noncarriers had lower fasting and postprandial serum triglycerides, higher levels of HDL-cholesterol and lower levels of LDL-cholesterol. Subclinical atherosclerosis, as measured by coronary artery calcification, was less common in carriers than noncarriers, which suggests that lifelong deficiency of apoC-III has a cardioprotective effect.


Subject(s)
Apolipoprotein C-III/genetics , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Coronary Artery Disease/prevention & control , Lipids/blood , Mutation , Triglycerides/blood , Adult , Apolipoprotein C-III/blood , Cholesterol/blood , Christianity , Coronary Artery Disease/genetics , Dietary Fats/administration & dosage , Fasting , Female , Genome-Wide Association Study , Haplotypes , Heterozygote , Humans , Linkage Disequilibrium , Male , Middle Aged , Pedigree , Pennsylvania , Polymorphism, Single Nucleotide , Risk Factors
11.
J Rehabil Res Dev ; 45(2): 273-81, 2008.
Article in English | MEDLINE | ID: mdl-18566945

ABSTRACT

Hemiparetic stroke leads to major skeletal muscle abnormalities, as illustrated by paretic leg atrophy, weakness, and spasticity. Furthermore, the hemiparetic limb muscle shifts to a fast-twitch muscle fiber phenotype with anaerobic metabolism. This study investigated whether skeletal muscle genes were altered in chronic hemiparetic stroke. The nonparetic leg muscle served as an internal control. We used Affymetrix microarray analysis to survey gene expression differences between paretic and nonparetic vastus lateralis muscle punch biopsies from 10 subjects with chronic hemiparetic stroke. Stroke latency was greater than 6 months. We found that 116 genes were significantly altered between the paretic and non paretic vastus lateralis muscles. These gene differences were consistent with reported differences after stroke in areas such as injury and inflammation markers, the myosin heavy chain profile, and high prevalence of impaired glucose tolerance and type 2 diabetes. Furthermore, while many other families of genes were altered, the gene families with the most genes altered included inflammation, cell cycle regulation, signal transduction, metabolism, and muscle contractile protein genes. This study is an early step toward identification of specific gene regulatory pathways that might lead to these differences, propagate disability, and increase vascular disease risk.


Subject(s)
Lower Extremity/physiopathology , Muscle, Skeletal/metabolism , Paresis/genetics , Stroke/genetics , Aged , Aged, 80 and over , Cell Cycle , Chronic Disease , Female , Gene Expression , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/physiopathology , Male , Microarray Analysis , Middle Aged , Muscle Contraction/genetics , Muscle, Skeletal/physiopathology , Paresis/metabolism , Paresis/physiopathology , Paresis/rehabilitation , RNA/metabolism , Signal Transduction , Stroke/metabolism , Stroke/physiopathology , Stroke Rehabilitation , Transcription, Genetic
12.
Diabetes ; 57(8): 2037-45, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18487449

ABSTRACT

OBJECTIVE: Accumulation of intracellular lipid droplets (LDs) in non-adipose tissues is recognized as a strong prognostic factor for the development of insulin resistance in obesity. LDs are coated with perilipin, adipose differentiation-related protein, tail interacting protein of 47 kd (PAT) proteins that are thought to regulate LD turnover by modulating lipolysis. Our hypothesis is that PAT proteins modulate LD metabolism and therefore insulin resistance. RESEARCH DESIGN AND METHODS: We used a cell culture model (murine AML12 loaded with oleic acid) and small interfering RNA to directly assess the impact of PAT proteins on LD accumulation, lipid metabolism, and insulin action. PAT proteins associated with excess fat deposited in livers of diet-induced obese (DIO) mice were also measured. RESULTS: Cells lacking PAT proteins exhibited a dramatic increase in LD size and a decrease in LD number. Further, the lipolytic rate increased by approximately 2- to 2.5-fold in association with increased adipose triglyceride lipase (ATGL) at the LD surface. Downregulation of PAT proteins also produced insulin resistance, as indicated by decreased insulin stimulation of Akt phosphorylation (P < 0.001). Phosphoinositide-dependent kinase-1 and phosphoinositide 3-kinase decreased, and insulin receptor substrate-1 307 phosphorylation increased. Increased lipids in DIO mice livers were accompanied by changes in PAT composition but also increased ATGL, suggesting a relative PAT deficiency. CONCLUSIONS: These data establish an important role for PAT proteins as surfactant at the LD surface, packaging lipids in smaller units and restricting access of lipases and thus preventing insulin resistance. We suggest that a deficiency of PAT proteins relative to the quantity of ectopic fat could contribute to cellular dysfunction in obesity and type 2 diabetes.


Subject(s)
Carrier Proteins/physiology , Hepatocytes/metabolism , Membrane Proteins/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Chromatography, Thin Layer , Down-Regulation , Fatty Acids, Nonesterified/metabolism , Hepatocytes/cytology , Immunoblotting , Immunohistochemistry , Insulin Resistance/physiology , Lipid Metabolism/physiology , Lipolysis/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Perilipin-2 , Perilipin-3 , RNA, Small Interfering/genetics
13.
Diabetes ; 56(6): 1655-61, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17329619

ABSTRACT

Central obesity and the accumulation of visceral fat are risk factors for the development of type 2 diabetes and cardiovascular disease. Omentin is a protein expressed and secreted from visceral but not subcutaneous adipose tissue that increases insulin sensitivity in human adipocytes. To determine the impact of obesity-dependent insulin resistance on the regulation of two omentin isoforms, gene expression and plasma levels were measured in lean, overweight, and obese subjects. Omentin 1 was shown to be the major circulating isoform in human plasma. Lean subjects had significantly higher plasma omentin 1 levels than obese and overweight subjects. In addition, higher plasma omentin 1 levels were detected in women compared with men. Plasma omentin 1 levels were inversely correlated with BMI, waist circumference, leptin levels, and insulin resistance as measured by homeostasis model assessment and positively correlated with adiponectin and HDL levels. Both omentin 1 and omentin 2 gene expression were decreased with obesity and were highly correlated with each other in visceral adipose tissue. In summary, decreased omentin levels are associated with increasing obesity and insulin resistance. Therefore, omentin levels may be predictive of the metabolic consequences or co-morbidities associated with obesity.


Subject(s)
Cytokines/blood , Cytokines/genetics , Gene Expression Regulation , Lectins/blood , Lectins/genetics , Obesity/blood , Obesity/genetics , Adiponectin/blood , Adipose Tissue/metabolism , Adult , Cytokines/isolation & purification , Female , GPI-Linked Proteins , Humans , Lectins/isolation & purification , Male , Middle Aged , Patient Selection , Polymerase Chain Reaction , RNA, Messenger/genetics
14.
Am J Physiol Endocrinol Metab ; 290(6): E1253-61, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16531507

ABSTRACT

Central (visceral) obesity is more closely associated with insulin resistance, type 2 diabetes, and cardiovascular disease than is peripheral [subcutaneous (sc)] obesity, but the underlying mechanism for this pathophysiological difference is largely unknown. To understand the molecular basis of this difference, we sequenced 10,437 expressed sequence tags (ESTs) from a human omental fat cDNA library and discovered a novel visceral fat depot-specific secretory protein, which we have named omentin. Omentin ESTs were more abundant than many known adipose genes, such as perilipin, adiponectin, and leptin in the cDNA library. Protein sequence analysis indicated that omentin mRNA encodes a peptide of 313 amino acids, containing a secretory signal sequence and a fibrinogen-related domain. Northern analysis demonstrated that omentin mRNA was predominantly expressed in visceral adipose tissue and was barely detectable in sc fat depots in humans and rhesus monkeys. Quantative real-time PCR showed that omentin mRNA was expressed in stromal vascular cells, but not fat cells, isolated from omental adipose tissue, with >150-fold less in sc cell fractions. Accordingly, omentin protein was secreted into the culture medium of omental, but not sc, fat explants. Omentin was detectable in human serum by Western blot analysis. Addition of recombinant omentin in vitro did not affect basal but enhanced insulin-stimulated glucose uptake in both sc (47%, n = 9, P = 0.003) and omental (approximately 30%, n = 3, P < 0.05) human adipocytes. Omentin increased Akt phosphorylation in the absence and presence of insulin. In conclusion, omentin is a new adipokine that is expressed in omental adipose tissue in humans and may regulate insulin action.


Subject(s)
Adipose Tissue/metabolism , Cytokines/genetics , Insulin/pharmacology , Lectins/genetics , Adipose Tissue/blood supply , Adipose Tissue/cytology , Animals , Cytokines/metabolism , Expressed Sequence Tags , GPI-Linked Proteins , Glucose Transport Proteins, Facilitative , Humans , Lectins/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt , Stromal Cells/metabolism
15.
Am J Physiol Heart Circ Physiol ; 290(5): H1848-55, 2006 May.
Article in English | MEDLINE | ID: mdl-16339827

ABSTRACT

In its role as an endothelial cell proliferation and migration factor, vascular endothelial growth factor (VEGF) can affect peripheral circulation and therefore impact maximal oxygen consumption (Vo2 max). Because of the role of VEGF, and because variation in the VEGF gene has the ability to alter VEGF gene expression and VEGF protein level, we hypothesized that VEGF gene polymorphisms are related to VEGF gene expression in human myoblasts and Vo2 max before and after aerobic exercise training. We analyzed the effects of the VEGF -2578/-1154/-634 promoter region haplotype on VEGF gene expression by using a luciferase reporter assay in cultured human myoblasts and found that the AAG and CGC haplotypes resulted in significantly higher hypoxia-stimulated VEGF gene expression than the AGG and CGG haplotypes. Consistent with these results, we found that individuals with at least one copy of the AAG or CGC haplotype had higher Vo2 max before and after aerobic exercise training than did subjects with only the AGG and/or CGG haplotype. In conclusion, we found that VEGF -2578/-1154/-634 haplotype impacts VEGF gene expression in human myoblasts and is associated with Vo2 max. These results have potential implications for aerobic exercise training and may prove relevant in the study of pathological conditions that can be affected by angiogenesis, such as coronary artery disease and peripheral artery disease.


Subject(s)
Exercise/physiology , Gene Expression Regulation/physiology , Muscle Fibers, Skeletal/physiology , Oxygen Consumption/genetics , Oxygen/metabolism , Physical Endurance/genetics , Vascular Endothelial Growth Factor A/physiology , Aged , DNA/genetics , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics
16.
Diabetes ; 54(1): 268-74, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15616038

ABSTRACT

We performed a genome-wide linkage scan of plasma adiponectin levels in 569 nondiabetic participants in the Amish Family Diabetes Study. The highest logarithm of odds (LOD) score (2.13; P = 0.0009) occurred on chromosome 3q27 between markers D3S1602 and D3S1580, which flank APM1/ACDC, the adiponectin gene. The APM1 +2019 A/- insertion/deletion polymorphism in the 3' untranslated region (single nucleotide polymorphism [SNP] +2019; deletion allele frequency 0.30 in Amish) showed strong association with adiponectin levels in a dosage-dependent manner in a direction consistent with that reported in previous studies, with deletion heterozygosity increasing adiponectin levels by 1.3 +/- 0.5 microg/ml and deletion homozygosity increasing levels by 3.0 +/- 0.8 microg/ml (P < 0.0001). Two other SNPs, rs2241766 and rs1501299, showed moderate association. In a subset of 523 subjects genotyped for both SNP +2019 and rs2241766, including the APM1 SNP +2019 genotype as a covariate reduced the linkage signal at 3q27 by 1.26 LOD units (from 2.22 to 0.96) and including both SNPs reduced the signal by 1.51 LOD units (to 0.71). These findings, combined with a two-point LOD score of 2.35 for SNP +2019, provide evidence that variation in APM1 is responsible for linkage of adiponectin levels to 3q27 in the Old Order Amish.


Subject(s)
Chromosomes, Human, Pair 3 , Genetic Variation , Intercellular Signaling Peptides and Proteins/genetics , Adiponectin , Chromosome Mapping , Genetic Markers , Genotype , Humans , Intercellular Signaling Peptides and Proteins/blood , Polymorphism, Single Nucleotide , Repetitive Sequences, Nucleic Acid
17.
J Clin Invest ; 114(10): 1414-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15545992

ABSTRACT

The Randle cycle, which has been invoked to explain the reciprocal relationship between fatty acid oxidation and glucose oxidation, has long been implicated as a potential mechanism for hyperglycemia and type 2 diabetes mellitus (T2DM). Now genetic, functional genomic, and transgenic approaches have identified PPARgamma coactivators (PGC-1alpha and PGC-1beta) as key regulators of mitochondrial number and function. They regulate adaptive thermogenesis as well as glucose and fat oxidation in muscle and fat tissue, gluconeogenesis in liver, and even glucose-regulated insulin secretion in beta cells. PGC-1alpha and PGC-1beta mRNA levels and the mitochondrial genes they regulate are decreased in muscle of people with prediabetes and T2DM. A new report indicates that PGC-1alpha and PGC-1beta mRNA levels decrease with age in individuals with a genetic variant in PGC-1alpha, and these decreases correlate with alterations in whole-body glucose and fatty acid oxidation. These findings provide insights into how aging modifies genetic susceptibility to alterations in oxidative phosphorylation and T2DM.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Genes , Transcription Factors/genetics , Transcription Factors/metabolism , Age Factors , Blood Glucose/metabolism , Diet , Fats/metabolism , Fatty Acids/metabolism , Genetic Variation , Gluconeogenesis , Humans , Hyperglycemia/etiology , Hyperglycemia/physiopathology , Insulin/metabolism , Models, Biological , Muscle, Skeletal/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , RNA, Messenger/analysis , RNA, Messenger/metabolism , Thermogenesis
18.
Biochem Biophys Res Commun ; 310(3): 927-35, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-14550293

ABSTRACT

Resistin is a newly identified adipocytokine that has been proposed to be a link between obesity and type 2 diabetes based on animal studies. However, the role of resistin in the pathogenesis of insulin resistance associated with obesity in humans remains unclear. We comparatively and quantitatively studied the tissue distributions of resistin mRNA between human and mouse. The expression level of resistin mRNA in human adipose tissue is extremely low but detectable by real-time PCR and is about 1/250 of that in the mouse. Remarkably, resistin mRNA is abundant in human primary acute leukemia cells and myeloid cell lines U937 and HL60, but not in the Raw264 mouse myeloid cell line. Resistin expression in U937 cells was not affected by lipopolysaccharide (LPS) or by ciglitazone, a PPARgamma ligand. Phylogenomics revealed that the human resistin gene is the ortholog of its murine counterpart and is located in a region of chromosome 19p13.3, which is syntenic to mouse chromosome 8A1. In addition to the resistin-like molecule (RELM) sequences already reported, bioinformatics analysis disclosed another RELM sequence in the vicinity of RELMbeta on human chromosome 3q13.1, but this sequence is unlikely to encode an expressed gene. Therefore, only two RELMs, resistin and RELMbeta, exist in humans, instead of the three RELMs, resistin, RELMalpha, and RELMbeta, that exist in mice. This finding provides a possible answer to the question of why only two RELMs have been cloned in humans and suggests that the RELM family is not well conserved in evolution and may function differently between species. Therefore, caution should be exercised in interpreting resistin as a link between obesity and insulin resistance in humans. The high expression of resistin in human leukemia cells suggests a hitherto unidentified biological function of resistin in leukocytes.


Subject(s)
Hormones, Ectopic/biosynthesis , Hormones, Ectopic/genetics , Intercellular Signaling Peptides and Proteins , Proteins , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , Cell Line , Chromosome Mapping , DNA, Complementary/metabolism , Genome , HL-60 Cells , Humans , Leukocytes/metabolism , Ligands , Lipopolysaccharides/metabolism , Mice , Molecular Sequence Data , Nerve Growth Factor , Oligonucleotides/metabolism , Phylogeny , RNA, Messenger/metabolism , Resistin , Reverse Transcriptase Polymerase Chain Reaction , Thiazolidinediones/pharmacology , Tissue Distribution , U937 Cells
19.
Curr Diab Rep ; 2(1): 83-95, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12643127

ABSTRACT

Insulin resistance, defined as the decreased ability of insulin to perform its biological functions, is likely to represent the primary physiologic defect underlying the insulin resistance syndrome (IRS), which includes insulin resistance/hyperinsulinemia, glucose intolerance and/or type 2 diabetes mellitus, visceral obesity, hypertension, and dyslipidemia. This constellation of traits is a leading cause of cardiovascular mortality and morbidity. Insulin sensitivity varies widely among individuals. Although environmental provocations including physical inactivity and caloric excess play an important role in the development of obesity and thus insulin resistance, epidemiologic and family studies show that there are also moderate genetic influences on the development of insulin resistance. Extreme forms of insulin resistance may be caused rarely by mutations in the genes for the insulin receptor and peroxisome proliferator-activated receptor gamma. However, the genetic basis for common more moderate forms of insulin resistance is likely to be polygenic and heterogeneous. Evidence further suggests that gene variants may have phenotypic influences on more than one IRS trait (so-called pleiotrophy), which may explain, in part, the clustering of these traits. This article reviews the evidence that insulin resistance has a genetic basis. Progress to date toward identifying specific gene variants are reviewed. Ultimately, the identification of specific gene variants that influence insulin resistance and other IRS traits will have profound influences on our understanding of the molecular and pathophysiologic basis of these disorders, from which new and more effective preventive and therapeutic interventions will be possible.


Subject(s)
Insulin Resistance/genetics , Chromosomes, Human, Pair 19/genetics , Humans , Insulin/physiology , Insulin Receptor Substrate Proteins , Mutation , Phosphoproteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...