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










Publication year range
1.
J Prev Alzheimers Dis ; 9(1): 22-29, 2022.
Article in English | MEDLINE | ID: mdl-35098970

ABSTRACT

Preclinical studies indicate an age-associated accumulation of senescent cells across multiple organ systems. Emerging evidence suggests that tau protein accumulation, which closely correlates with cognitive decline in Alzheimer's disease and other tauopathies, drives cellular senescence in the brain. Pharmacologically clearing senescent cells in mouse models of tauopathy reduced brain pathogenesis. Compared to vehicle treated mice, intermittent senolytic administration reduced tau accumulation and neuroinflammation, preserved neuronal and synaptic density, restored aberrant cerebral blood flow, and reduced ventricular enlargement. Intermittent dosing of the senolytics, dasatinib plus quercetin, has shown an acceptable safety profile in clinical studies for other senescence-associated conditions. With these data, we proposed and herein describe the objectives and methods for a clinical vanguard study. This initial open-label clinical trial pilots an intermittent senolytic combination therapy of dasatinib plus quercetin in five older adults with early-stage Alzheimer's disease. The primary objective is to evaluate the central nervous system penetration of dasatinib and quercetin through analysis of cerebrospinal fluid collected at baseline and after 12 weeks of treatment. Further, through a series of secondary outcome measures to assess target engagement of the senolytic compounds and Alzheimer's disease-relevant cognitive, functional, and physical outcomes, we will collect preliminary data on safety, feasibility, and efficacy. The results of this study will be used to inform the development of a randomized, double-blind, placebo-controlled multicenter phase II trial to further explore of the safety, feasibility, and efficacy of senolytics for modulating the progression of Alzheimer's disease. Clinicaltrials.gov registration number and date: NCT04063124 (08/21/2019).


Subject(s)
Alzheimer Disease , Tauopathies , Aged , Animals , Cellular Senescence , Dasatinib/pharmacology , Dasatinib/therapeutic use , Humans , Mice , Senotherapeutics
2.
Curr Mol Biol Rep ; 7(3): 31-40, 2021.
Article in English | MEDLINE | ID: mdl-34401216

ABSTRACT

Cellular senescence (CS) is increasingly implicated in the etiology of age-related diseases. While CS can facilitate physiological processes such as tissue repair and wound healing, senescent cells also contribute to pathophysiological processes involving macromolecular damage and metabolic dysregulation that characterize multiple morbid and prevalent diseases, including Alzheimer's disease, osteoarthritis, atherosclerotic vascular disease, diabetes mellitus, and idiopathic pulmonary fibrosis (IPF). Preclinical studies targeting senescent cells and the senescence-associated secretory phenotype (SASP) with "senotherapeutics" have demonstrated improvement in age-related morbidity associated with these disease states. Despite promising results from these preclinical trials, few human clinical trials have been conducted. A first-in-human, open-label, pilot study of the senolytic combination of dasatinib and quercetin (DQ) in patients with IPF showed improved physical function and mobility. In this review, we will discuss our current understanding of cellular senescence, its role in age-associated diseases, with a specific focus on IPF, and potential for senotherapeutics in the treatment of fibrotic lung diseases.

3.
Diabet Med ; 32(5): 657-64, 2015 May.
Article in English | MEDLINE | ID: mdl-25484175

ABSTRACT

AIMS: The molecular mechanisms by which muraglitazar (peroxisome proliferator-activated receptor γ/α agonist) improves insulin sensitivity in Type 2 diabetes mellitus are not fully understood. We hypothesized that muraglitazar would increase expression of 5'-monophosphate-activated protein kinase and genes involved in adiponectin signalling, free fatty acid oxidation and mitochondrial function in skeletal muscle. METHODS: Sixteen participants with Type 2 diabetes received muraglitazar, 5 mg/day (n = 12) or placebo (n = 4). Before and after 16 weeks, participants had vastus lateralis muscle biopsy followed by 180 min euglycaemic hyperinsulinaemic clamp. RESULTS: Muraglitazar increased plasma adiponectin (9.0 ± 1.1 to 17.8 ± 1.5 µg/ml, P < 0.05), while no significant change was observed with placebo. After 16 weeks with muraglitazar, fasting plasma glucose declined by 31%, fasting plasma insulin decreased by 44%, insulin-stimulated glucose disposal increased by 81%, HbA1c decreased by 21% and plasma triglyceride decreased by 39% (all P < 0.05). Muraglitazar increased mRNA levels of 5'-monophosphate-activated protein kinase, adiponectin receptor 1, adiponectin receptor 2, peroxisome proliferator-activated receptor gamma coactivator-1 alpha and multiple genes involved in mitochondrial function and fat oxidation. In the placebo group, there were no significant changes in expression of these genes. CONCLUSIONS: Muraglitazar increases plasma adiponectin, stimulates muscle 5'-monophosphate-activated protein kinase expression and increases expression of genes involved in adiponectin signalling, mitochondrial function and fat oxidation. These changes represent important cellular mechanisms by which dual peroxisome proliferator-activated receptor agonists improve skeletal muscle insulin sensitivity.


Subject(s)
Adiponectin/physiology , Diabetes Mellitus, Type 2/physiopathology , Fatty Acids, Nonesterified/metabolism , Glycine/analogs & derivatives , Mitochondria, Muscle/drug effects , Muscle, Skeletal/metabolism , Oxazoles/pharmacology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/physiology , Adiponectin/blood , Biopsy , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Glycine/pharmacology , Glycine/therapeutic use , Humans , Insulin/blood , Insulin Resistance/physiology , Male , Middle Aged , Mitochondria, Muscle/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Oxazoles/therapeutic use , Oxidation-Reduction , PPAR alpha/agonists , PPAR gamma/agonists , Signal Transduction/physiology , Triglycerides/blood
4.
Nutr Diabetes ; 3: e74, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23752133

ABSTRACT

INTRODUCTION: Individuals with obesity and type 2 diabetes (T2D) are typically insulin resistant, exhibiting impaired skeletal muscle glucose uptake. Animal and cell culture experiments have shown that site-specific phosphorylation of the Rab-GTPase-activating proteins AS160 and TBC1D1 is critical for GLUT4 translocation facilitating glucose uptake, but their regulation in human skeletal muscle is not well understood. METHODS: Here, lean, obese and T2D subjects underwent a euglycemic-hyperinsulinemic clamp, and vastus lateralis muscle biopsies were obtained before, and at 30 and 180 min post insulin infusion. RESULTS: Obese and T2D subjects had higher body mass indexes and fasting insulin concentrations, and T2D subjects showed insulin resistance. Consistent with the clamp findings, T2D subjects had impaired insulin-stimulated phosphorylation of AS160 Thr(642), a site previously shown to be important in glucose uptake in rodents. Interestingly, insulin-stimulated phosphorylation of TBC1D1 Thr(590), a site shown to be regulated by insulin in rodents, was only increased in T2D subjects, although the functional significance of this difference is unknown. CONCLUSION: These data show that insulin differentially regulates AS160 and TBC1D1 phosphorylation in human skeletal muscle. Impaired insulin-stimulated glucose uptake in T2D subjects is accompanied by dysregulation of AS160 and TBC1D1 phosphorylation in skeletal muscle, suggesting that these proteins may regulate glucose uptake in humans.

5.
Diabetes Obes Metab ; 15(10): 931-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23551856

ABSTRACT

AIMS: This study examined the effects of pioglitazone on body weight and bone mineral density (BMD) prospectively in patients with impaired glucose tolerance as pioglitazone (TZD) increases body weight and body fat in diabetic patients and increases the risk of bone fractures. METHODS: A total of 71 men and 163 women aged 49.3 (10.7) years [mean (s.d.)]; body mass index (BMI), 34.5 (5.9) kg/m(2) were recruited at five sites for measurements of body composition by dual energy X-ray absorptiometry at baseline and at conversion to diabetes or study end, if they had not converted. RESULTS: Mean follow-up was 33.6 months in the pioglitazone group and 32.1 months in the placebo group. Body weight increased 4.63 ± 0.60 (m ± s.e.) kg in the pioglitazone group compared to 0.98 ± 0.62 kg in the PIO group (p < 0.0001). Body fat rose 4.89 ± 0.42 kg in the pioglitazone group compared to 1.41 ± 0.44 kg, (p < 0.0001) in placebo-treated subjects. The increase in fat was greater in legs and trunk than in the arms. BMD was higher in all regions in men and significantly so in most. PIO decreased BMD significantly in the pelvis in men and women, decreased BMD in the thoracic spine and ribs of women and the lumbar spine and legs of men. Bone mineral content also decreased significantly in arms, legs, trunk and in the total body. CONCLUSIONS: Pioglitazone increased peripheral fat more than truncal fat and decreased BMD in several regions of the body.


Subject(s)
Bone Density/drug effects , Diabetes Mellitus, Type 2/prevention & control , Fractures, Bone/pathology , Hypoglycemic Agents/therapeutic use , Prediabetic State/drug therapy , Thiazolidinediones/therapeutic use , Absorptiometry, Photon , Adipose Tissue , Body Mass Index , Body Weight/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Female , Follow-Up Studies , Fractures, Bone/chemically induced , Fractures, Bone/epidemiology , Glucose Tolerance Test , Humans , Male , Middle Aged , Pioglitazone , Prediabetic State/blood , Prediabetic State/epidemiology , Prospective Studies , Treatment Outcome
6.
Diabetologia ; 54(10): 2660-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21779871

ABSTRACT

AIMS/HYPOTHESIS: Chemokines and their receptors such as chemokine (C-C motif) receptor 2 (CCR2) may contribute to the pathogenesis of the metabolic syndrome via their effects on inflammatory monocytes. Increased accumulation of CCR2-driven inflammatory monocytes in epididymal fat pads is thought to favour the development of insulin resistance. Ultimately, the resulting hyperglycaemia and dyslipidaemia contribute to development of the metabolic syndrome complications such as cardiovascular disease and diabetic nephropathy. Our goal was to elucidate the role of CCR2 and inflammatory monocytes in a mouse model that resembles the human metabolic syndrome. METHODS: We generated a model of the metabolic syndrome by backcrossing KKAy ( + ) with Apoe ( -/- ) mice (KKAy ( + ) Apoe ( -/- )) and studied the role of CCR2 in this model system. RESULTS: KKAy ( + ) Apoe ( -/- ) mice were characterised by the presence of obesity, insulin resistance, dyslipidaemia and increased systemic inflammation. This model also manifested two complications of the metabolic syndrome: atherosclerosis and diabetic nephropathy. Inactivation of Ccr2 in KKAy (+) Apoe ( -/- ) mice protected against the metabolic syndrome, as well as atherosclerosis and diabetic nephropathy. This protective phenotype was associated with a reduced number of inflammatory monocytes in the liver and muscle, but not in the epididymal fat pads; circulating levels of adipokines such as leptin, resistin and adiponectin were also not reduced. Interestingly, the proportion of inflammatory monocytes in the liver, pancreas and muscle, but not in the epididymal fat pads, correlated significantly with peripheral glucose levels. CONCLUSIONS/INTERPRETATION: CCR2-driven inflammatory monocyte accumulation in the liver and muscle may be a critical pathogenic factor in the development of the metabolic syndrome.


Subject(s)
Apolipoproteins E/metabolism , Metabolic Syndrome/metabolism , Receptors, CCR2/metabolism , Animals , Apolipoproteins E/genetics , Blood Pressure/genetics , Blood Pressure/physiology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Dyslipidemias/genetics , Dyslipidemias/metabolism , Eating/genetics , Eating/physiology , Flow Cytometry , Humans , Immunohistochemistry , Insulin Resistance/genetics , Insulin Resistance/physiology , Interleukin-6/metabolism , Metabolic Syndrome/genetics , Mice , Mice, Knockout , Receptors, CCR2/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Diabetes Obes Metab ; 13(10): 893-902, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21615671

ABSTRACT

AIM: To assess the effect of muraglitazar, a dual peroxisome proliferator-activated receptor (PPAR)γ-α agonist, versus placebo on metabolic parameters and body composition in subjects with type 2 diabetes mellitus (T2DM). METHODS: Twenty-seven T2DM subjects received oral glucose tolerance test (OGTT), euglycaemic insulin clamp with deuterated glucose, measurement of total body fat (DEXA), quantitation of muscle/liver (MRS) and abdominal subcutaneous and visceral (MRI) fat, and then were randomized to receive, in addition to diet, muraglitazar (MURA), 5 mg/day, or placebo (PLAC) for 4 months. RESULTS: HbA1c(c) decreased similarly (2.1%) during both MURA and PLAC treatments despite significant weight gain with MURA (+2.5 kg) and weight loss with PLAC (-0.7 kg). Plasma triglyceride, LDL cholesterol, free fatty acid (FFA), hsCRP levels all decreased with MURA while plasma adiponectin and HDL cholesterol increased (p < 0.05-0.001). Total body (muscle), hepatic and adipose tissue sensitivity to insulin and ß cell function all improved with MURA (p < 0.05-0.01). Intramyocellular, hepatic and abdominal visceral fat content decreased, while total body and subcutaneous abdominal fat increased with MURA (p < 0.05-0.01). CONCLUSIONS: Muraglitazar (i) improves glycaemic control by enhancing insulin sensitivity and ß cell function in T2DM subjects, (ii) improves multiple cardiovascular risk factors, (iii) reduces muscle, visceral and hepatic fat content in T2DM subjects. Despite similar reduction in A1c with PLAC/diet, insulin sensitivity and ß cell function did not improve significantly.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin/drug effects , Glycine/analogs & derivatives , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Intra-Abdominal Fat/drug effects , Oxazoles/pharmacology , Peroxisome Proliferator-Activated Receptors/agonists , Body Mass Index , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Clamp Technique , Glucose Tolerance Test , Glycated Hemoglobin/metabolism , Glycine/administration & dosage , Glycine/pharmacology , Humans , Hypoglycemic Agents/administration & dosage , Insulin Resistance , Insulin-Secreting Cells/metabolism , Intra-Abdominal Fat/metabolism , Male , Middle Aged , Oxazoles/administration & dosage , Peroxisome Proliferator-Activated Receptors/metabolism
8.
J Clin Endocrinol Metab ; 96(6): 1763-70, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21411546

ABSTRACT

OBJECTIVE: Our objective was to examine the mechanisms via which exenatide attenuates postprandial hyperglycemia in type 2 diabetes mellitus (T2DM). STUDY DESIGN: Seventeen T2DM patients (44 yr; seven females, 10 males; body mass index = 33.6 kg/m(2); glycosylated hemoglobin = 7.9%) received a mixed meal followed for 6 h with double-tracer technique ([1-(14)C]glucose orally; [3-(3)H]glucose i.v.) before and after 2 wk of exenatide. In protocol II (n = 5), but not in protocol I (n = 12), exenatide was given in the morning of the repeat meal. Total and oral glucose appearance rates (RaT and RaO, respectively), endogenous glucose production (EGP), splanchnic glucose uptake (75 g - RaO), and hepatic insulin resistance (basal EGP × fasting plasma insulin) were determined. RESULTS: After 2 wk of exenatide (protocol I), fasting plasma glucose decreased (from 10.2 to 7.6 mm) and mean postmeal plasma glucose decreased (from 13.2 to 11.3 mm) (P < 0.05); fasting and meal-stimulated plasma insulin and glucagon did not change significantly. After exenatide, basal EGP decreased (from 13.9 to 10.8 µmol/kg · min, P < 0.05), and hepatic insulin resistance declined (both P < 0.05). RaO, gastric emptying (acetaminophen area under the curve), and splanchnic glucose uptake did not change. In protocol II (exenatide given before repeat meal), fasting plasma glucose decreased (from 11.1 to 8.9 mm) and mean postmeal plasma glucose decreased (from 14.2 to 10.1 mm) (P < 0.05); fasting and meal-stimulated plasma insulin and glucagon did not change significantly. After exenatide, basal EGP decreased (from 13.4 to 10.7 µmol/kg · min, P = 0.05). RaT and RaO decreased markedly from 0-180 min after meal ingestion, consistent with exenatide's action to delay gastric emptying. CONCLUSIONS: Exenatide improves 1) fasting hyperglycemia by reducing basal EGP and 2) postmeal hyperglycemia by reducing the appearance of oral glucose in the systemic circulation.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Peptides/pharmacology , Postprandial Period/drug effects , Venoms/pharmacology , Adult , Area Under Curve , Blood Glucose/metabolism , Exenatide , Female , Glucagon/blood , Glycated Hemoglobin/metabolism , Humans , Insulin/blood , Male , Middle Aged , Postprandial Period/physiology
9.
Diabetes Metab Res Rev ; 27(4): 373-82, 2011 May.
Article in English | MEDLINE | ID: mdl-21309063

ABSTRACT

BACKGROUND: To test potential differences between the actions of anti-diabetic medications, we examined the effects of oral hypoglycaemic agents versus glargine-apidra insulin therapy in T2DM. METHODS: T2DM subjects were randomized to either oral hypoglycaemic agents (pioglitazone, metformin and glipizide, n = 9) or insulin therapy (n = 12) for 6 months. Carotid intimal media thickness, vascular reactivity (flow-mediated vasodilatation; percent change in brachial artery basal diameter post-ischaemia) and sublingual nitrate were measured with ultrasonography. Euglycemic hyperinsulinemic (80 mU/m(2) ) clamp with [3]-3H-glucose and muscle biopsies were performed. RESULTS: Fasting plasma glucose (~257 to ~124 mg/dL, oral hypoglycaemic agents and ~256 to ~142 mg/dL, IT) and HbA(1c) (~10.3 to ~6.4%, OHA and ~10.7 to ~7.1%, IT) improved comparably. Endogenous glucose production (~2.1 to ~1.7 mg/kg/min, oral hypoglycaemic agents and ~2.3 to ~2.0 mg/kg/min, insulin therapy) and endogenous glucose production suppression by insulin (~0.4 to ~0.3 mg/kg min, oral hypoglycaemic agents and ~0.5 to ~0.7 mg/kg min, insulin therapy) were different. Total glucose disposal × 100 increased in the oral hypoglycaemic agents group (~5.2 to ~8.1; p = 0.03), but not in insulin therapy (~6.0 to ~5.4 mg/kg/min/µU/mL × 100). OHA reduced CIMT (~0.080 to ~0.068 cm; p < 0.05), whereas insulin therapy did not (~0.075 to ~0.072 cm). After sublingual nitrate, brachial artery basal diameter increased in the OHA group (~8.7 to ~18.2%), but not in insulin therapy (~11.2 to ~15.0%; p < 0.02). Except for plasma adiponectin (~7 to ~15, oral hypoglycaemic agents versus ~6 to ~10, IT), changes in inflammatory markers in the circulation and in muscle (IκBα, super-oxidase dismutase 2, monocyte-chemo-attractant protein 1, p-ERK and JNK) were equivalent. CONCLUSIONS: Oral hypoglycaemic agents and insulin therapy treated patients achieved adequate glycemic control and the effects on circulating and muscle inflammatory biomarkers were similar, but only oral hypoglycaemic agents improved insulin sensitivity, vascular function and carotid intimal media thickness. These findings in a small sample suggest that the use of oral hypoglycaemic agents provides additional benefits to patients with T2DM.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Insulin/therapeutic use , Muscle, Skeletal/drug effects , Myositis/drug therapy , Adult , Body Mass Index , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Diseases/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Drug Combinations , Female , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/administration & dosage , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Insulin/analogs & derivatives , Insulin Glargine , Insulin, Long-Acting , Male , Metformin/administration & dosage , Metformin/therapeutic use , Mexican Americans , Middle Aged , Muscle, Skeletal/metabolism , Myositis/complications , Pioglitazone , Sulfonylurea Compounds/administration & dosage , Sulfonylurea Compounds/therapeutic use , Thiazolidinediones/administration & dosage , Thiazolidinediones/therapeutic use , Tunica Intima/drug effects , Tunica Intima/pathology
10.
Diabetologia ; 53(3): 435-45, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20012012

ABSTRACT

AIMS/HYPOTHESIS: The aim of the study was to examine the determinants of oral glucose tolerance in 602 persons with impaired glucose tolerance (IGT) who participated in the Actos Now for Prevention of Diabetes (ACT NOW) study. METHODS: In addition to the 602 IGT participants, 115 persons with normal glucose tolerance (NGT) and 50 with impaired fasting glucose (IFG) were identified during screening and included in this analysis. Insulin secretion and insulin sensitivity indices were derived from plasma glucose and insulin during an OGTT. The acute insulin response (AIR) (0-10 min) and insulin sensitivity (S(I)) were measured with the frequently sampled intravenous glucose tolerance test (FSIVGTT) in a subset of participants. RESULTS: At baseline, fasting plasma glucose, 2 h postprandial glucose (OGTT) and HbA(1c) were 5.8 +/- 0.02 mmol/l, 10.5 +/- 0.05 mmol/l and 5.5 +/- 0.04%, respectively, in participants with IGT. Participants with IGT were characterised by defects in early (DeltaI (0-30)/DeltaG (0-30) x Matsuda index, where DeltaI is change in insulin in the first 30 min and DeltaG is change in glucose in the first 30 min) and total (DeltaI(0-120)/DeltaG(0-120) x Matsuda index) insulin secretion and in insulin sensitivity (Matsuda index and S(I)). Participants with IGT in whom 2 h plasma glucose was 7.8-8.3 mmol/l had a 63% decrease in the insulin secretion/insulin resistance (disposition) index vs participants with NGT and this defect worsened progressively as 2 h plasma glucose rose to 8.9-9.94 mmol/l (by 73%) and 10.0-11.05 mmol/l (by 80%). The Matsuda insulin sensitivity index was reduced by 40% in IGT compared with NGT (p < 0.005). In multivariate analysis, beta cell function was the primary determinant of glucose AUC during OGTT, explaining 62% of the variance. CONCLUSION: Our results strongly suggest that progressive beta cell failure is the main determinant of progression of NGT to IGT.


Subject(s)
Blood Glucose/analysis , Glucose Tolerance Test/methods , Algorithms , Area Under Curve , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Double-Blind Method , Female , Humans , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/cytology , Male , Middle Aged , Placebos , Prospective Studies
11.
Diabetologia ; 52(10): 2169-81, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19633828

ABSTRACT

AIMS/HYPOTHESIS: TNF-alpha levels are increased in obesity and type 2 diabetes. The regulation of TNF-alpha converting enzyme (TACE) and its inhibitor, tissue inhibitor of metalloproteinase 3 (TIMP3), in human type 2 diabetes is unknown. METHODS: We examined TACE/TIMP3 regulation: (1) in lean and obese normal glucose tolerant (NGT) individuals and in type 2 diabetes patients; (2) following 6 h of lipid/saline infusion in NGT individuals; and (3) in cultured human myotubes from lean NGT individuals incubated with palmitate. Insulin sensitivity was assessed by a euglycaemic clamp and TACE/TIMP3 was evaluated by confocal microscopy, RT-PCR, western blotting and an in vitro activity assay. Circulating TNF-alpha, TNF-alpha-receptor 1 (TNFR1), TNF-alpha-receptor 2 (TNFR2), IL-6 receptor (IL-6R), vascular cell adhesion molecule (VCAM) and intercellular adhesion molecule (ICAM) levels were evaluated. RESULTS: TIMP3 levels were reduced and TACE enzymatic activity was increased in type 2 diabetes skeletal muscle. TACE expression, and TACE, TNF-alpha, TNFR1 and IL-6R levels were increased in type 2 diabetes, and positively correlated with insulin resistance. A 6 h lipid infusion into NGT individuals decreased insulin-stimulated glucose metabolism by 25% with increased TACE, decreased expression of the gene encoding TIMP3 and increased IL-6R release. Palmitate induced a dramatic reduction of TIMP3 and increased the TACE/TIMP3 ratio in cultured myotubes. CONCLUSIONS/INTERPRETATION: TACE activity was increased in skeletal muscle of obese type 2 diabetes patients and in lipid-induced insulin resistance. We propose that dysregulation of membrane proteolysis by TACE/TIMP3 of TNF-alpha and IL-6R is an important factor for the development of skeletal muscle insulin resistance in obese type 2 diabetes patients by a novel autocrine/paracrine mechanism.


Subject(s)
ADAM Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Adult , Blotting, Western , Diabetes Mellitus, Type 2/genetics , Enzyme-Linked Immunosorbent Assay , Female , Humans , Insulin Resistance/genetics , Male , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics
12.
Diabetologia ; 52(4): 723-32, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19169664

ABSTRACT

AIMS/HYPOTHESIS: The molecular mechanisms by which thiazolidinediones improve insulin sensitivity in type 2 diabetes are not fully understood. We hypothesised that pioglitazone would activate the adenosine 5'-monophosphate-activated protein kinase (AMPK) pathway and increase the expression of genes involved in adiponectin signalling, NEFA oxidation and mitochondrial function in human skeletal muscle. METHODS: A randomised, double-blind, parallel study was performed in 26 drug-naive type 2 diabetes patients treated with: (1) pioglitazone (n = 14) or (2) aggressive nutritional therapy (n = 12) to reduce HbA(1c) to levels observed in the pioglitazone-treated group. Participants were assigned randomly to treatment using a table of random numbers. Before and after 6 months, patients reported to the Clinical Research Center of the Texas Diabetes Institute for a vastus lateralis muscle biopsy followed by a 180 min euglycaemic-hyperinsulinaemic (80 mU m(-2) min(-1)) clamp. RESULTS: All patients in the pioglitazone (n = 14) or nutritional therapy (n = 12) group were included in the analysis. Pioglitazone significantly increased plasma adiponectin concentration by 79% and reduced fasting plasma NEFA by 35% (both p < 0.01). Following pioglitazone, insulin-stimulated glucose disposal increased by 30% (p < 0.01), and muscle AMPK and acetyl-CoA carboxylase (ACC) phosphorylation increased by 38% and 53%, respectively (p < 0.05). Pioglitazone increased mRNA levels for adiponectin receptor 1 and 2 genes (ADIPOR1, ADIPOR2), peroxisome proliferator-activated receptor gamma, coactivator 1 gene (PPARGC1) and multiple genes involved in mitochondrial function and fat oxidation. Despite a similar reduction in HbA(1c) and similar improvement in insulin sensitivity with nutritional therapy, there were no significant changes in muscle AMPK and ACC phosphorylation, or the expression of ADIPOR1, ADIPOR2, PPARGC1 and genes involved in mitochondrial function and fat oxidation. No adverse (or unexpected) effects or side effects were reported from the study. CONCLUSIONS/INTERPRETATIONS: Pioglitazone increases plasma adiponectin levels, stimulates muscle AMPK signalling and increases the expression of genes involved in adiponectin signalling, mitochondrial function and fat oxidation. These changes may represent an important cellular mechanism by which thiazolidinediones improve skeletal muscle insulin sensitivity. TRIAL REGISTRATION: NCT 00816218 FUNDING: This trial was funded by National Institutes of Health Grant DK24092, VA Merit Award, GCRC Grant RR01346, Executive Research Committee Research Award from the University of Texas Health Science Center at San Antonio, American Diabetes Association Junior Faculty Award, American Heart Association National Scientist Development Grant, Takeda Pharmaceuticals North America Grant and Canadian Institute of Health Research Grant.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/blood , Diabetes Mellitus, Type 2/drug therapy , Gene Expression Regulation/drug effects , Hypoglycemic Agents/therapeutic use , Mitochondria, Muscle/metabolism , Thiazolidinediones/therapeutic use , AMP-Activated Protein Kinases/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , DNA Primers , Diet, Diabetic , Double-Blind Method , Fatty Acids, Nonesterified/blood , Female , Glucose Clamp Technique , Humans , Hyperinsulinism , Male , Malonyl Coenzyme A/metabolism , Middle Aged , Pioglitazone , Polymerase Chain Reaction , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Diabetologia ; 50(8): 1723-31, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17520238

ABSTRACT

AIMS/HYPOTHESIS: The aim of the study was to examine the effects of pioglitazone (PIO), a peroxisome proliferator-activated receptor (PPAR)-gamma agonist, and fenofibrate (FENO), a PPAR-alpha agonist, as monotherapy and in combination on glucose and lipid metabolism. SUBJECTS AND METHODS: Fifteen type 2 diabetic patients received FENO (n = 8) or PIO (n = 7) for 3 months, followed by the addition of the other agent for 3 months in an open-label study. Subjects received a 4 h hyperinsulinaemic-euglycaemic clamp and a hepatic fat content measurement at 0, 3 and 6 months. RESULTS: Following PIO, fasting plasma glucose (FPG) (p < 0.05) and HbA(1c) (p < 0.01) decreased, while plasma adiponectin (AD) (5.5 +/- 0.9 to 13.8 +/- 3.5 microg/ml [SEM], p < 0.03) and the rate of insulin-stimulated total-body glucose disposal (R (d)) (23.8 +/- 3.8 to 40.5 +/- 4.4 micromol kg(-1) min(-1), p < 0.005) increased. After FENO, FPG, HbA(1c), AD and R (d) did not change. PIO reduced fasting NEFA (784 +/- 53 to 546 +/- 43 micromol/l, p < 0.05), triacylglycerol (2.12 +/- 0.28 to 1.61 +/- 0.22 mmol/l, p < 0.05) and hepatic fat content (20.4 +/- 4.8 to 10.2 +/- 2.5%, p < 0.02). Following FENO, fasting NEFA and hepatic fat content did not change, while triacylglycerol decreased (2.20 +/- 0.14 to 1.59 +/- 0.13 mmol/l, p < 0.01). Addition of FENO to PIO had no effect on R (d), FPG, HbA(1c), NEFA, hepatic fat content or AD, but triacylglycerol decreased (1.61 +/- 0.22 to 1.00 +/- 0.15 mmol/l, p < 0.05). Addition of PIO to FENO increased R (d) (24.9 +/- 4.4 to 36.1 +/- 2.2 micromol kg(-1) min(-1), p < 0.005) and AD (4.1 +/- 0.8 to 13.1 +/- 2.5 microg/ml, p < 0.005) and reduced FPG (p < 0.05), HbA(1c) (p < 0.05), NEFA (p < 0.01), hepatic fat content (18.3 +/- 3.1 to 13.5 +/- 2.1%, p < 0.03) and triacylglycerol (1.59 +/- 0.13 to 0.96 +/- 0.9 mmol/l, p < 0.01). Muscle adenosine 5'-monophosphate-activated protein kinase (AMPK) activity did not change following FENO; following the addition of PIO, muscle AMPK activity increased significantly (phosphorylated AMPK:total AMPK ratio 1.2 +/- 0.2 to 2.2 +/- 0.3, p < 0.01). CONCLUSIONS/INTERPRETATION: We conclude that PPAR-alpha therapy has no effect on NEFA or glucose metabolism and that addition of a PPAR-alpha agonist to a PPAR-gamma agent causes a further decrease in plasma triacylglycerol, but has no effect on NEFA or glucose metabolism.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucose/metabolism , Hypoglycemic Agents/therapeutic use , Lipid Metabolism/drug effects , PPAR alpha/agonists , PPAR gamma/agonists , AMP-Activated Protein Kinases , Adiponectin/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Drug Therapy, Combination , Female , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Humans , Hypoglycemic Agents/pharmacology , Insulin/blood , Insulin Resistance , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Multienzyme Complexes/metabolism , Phosphorylation/drug effects , Pioglitazone , Protein Serine-Threonine Kinases/metabolism , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Treatment Outcome
14.
Acta Physiol Scand ; 178(4): 337-45, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12864738

ABSTRACT

The AMP-activated protein kinase (AMPK) is an enzyme that is activated in situations where there are changes in the cellular energy status such as muscle contraction and hypoxia. AMPK can also be pharmacologically activated by the compound 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) and the antidiabetic agent metformin. Several studies support the hypothesis that AMPK plays an important role in the stimulation of muscle glucose uptake by these physiological and pharmacological stimuli. In isolated rat muscles, activation of AMPK is associated with increases in glucose uptake through an insulin-independent mechanism. Studies done in rodents have shown that the activation of AMPK by AICAR is accompanied by decreases in blood glucose concentrations, in part due to enhanced muscle glucose uptake. Similar to exercise, AICAR not only directly stimulates glucose uptake into the skeletal muscle, but also enhances insulin sensitivity. The activation of AMPK and associated increases in muscle glucose uptake are affected by factors such as glycogen content, exercise training and fibre type. The effects of AMPK on muscle glucose uptake makes this protein a promising pharmacological target for the treatment of type 2 diabetes.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Glucose/metabolism , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/metabolism , Animals , Exercise/physiology , Glycogen/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Metformin/pharmacology , Muscle Contraction/physiology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/drug effects , Nutritional Status/physiology , Rats , Ribonucleotides/metabolism
15.
Biochem Soc Trans ; 31(Pt 1): 191-5, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12546683

ABSTRACT

Physical exercise increases muscle glucose uptake, enhances insulin sensitivity and leads to fatty acid oxidation in muscle. The AMP-activated protein kinase (AMPK) is an energy-sensing enzyme that is strongly activated during muscle contraction due to acute decreases in ATP/AMP and phosphocreatine/creatine ratios. Accumulating evidence suggests that AMPK plays an important role in mediating these metabolic processes. Furthermore, AMPK has been implicated in regulating gene transcription and therefore may play a role in some of the cellular adaptations to training exercise. There is also evidence that changes in AMPK activity result in altered cellular glycogen content, suggesting that this enzyme regulates glycogen metabolism. Recent studies have shown that the magnitude of AMPK activation and associated metabolic responses are affected by factors such as glycogen content, exercise training and fibre type. In summary, AMPK regulates several metabolic pathways during acute exercise and modifies the expression of many genes involved in the adaptive changes to exercise training.


Subject(s)
Exercise , Multienzyme Complexes/physiology , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases , Animals , Enzyme Activation , Gene Expression Regulation, Enzymologic , Glycogen/metabolism , Humans , Insulin/metabolism , Multienzyme Complexes/metabolism , Mutation , Protein Serine-Threonine Kinases/metabolism , Rats
16.
Circulation ; 104(14): 1664-9, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11581146

ABSTRACT

BACKGROUND: Recent reports suggest that activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK), in response to acute changes in cellular energy status in cardiac and skeletal muscles, results in altered substrate utilization. We hypothesized that chronic alterations in myocardial energetics in hypertrophied hearts (left ventricular hypertrophy, LVH) will lead to elevated AMPK activity, which in turn regulates substrate utilization. METHODS AND RESULTS: Using (31)P NMR spectroscopy and biochemical assays, we found that in LVH hearts, adenosine triphosphate (ATP) concentration decreased by 10%, phosphocreatine concentration decreased by 30%, and total creatine concentration was unchanged. Thus, the ratio of phosphocreatine/creatine decreased to one third of controls, and the ratio of AMP/ATP increased to 5 times above controls. These changes were associated with increased alpha(1) and alpha(2) AMPK activity (3.5- and 4.8-fold above controls, respectively). The increase in AMPK alpha(1) activity was accompanied by a 2-fold increase in alpha(1) expression, whereas alpha(2) expression was decreased by 30% in LVH. The basal rate of 2-deoxyglucose uptake increased by 3-fold in LVH, which was associated with an increased amount of glucose transporters present on the plasma membrane. CONCLUSIONS: These results demonstrate for the first time that chronic changes in myocardial energetics in hypertrophied hearts are accompanied by significant elevations in AMPK activity and isoform-specific alterations in AMPK expression. It also raises the possibility that AMPK signaling plays an important role in regulating substrate utilization in hypertrophied hearts.


Subject(s)
Hypertrophy, Left Ventricular/metabolism , Multienzyme Complexes/metabolism , Muscle Proteins , Myocardium/enzymology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Acyl-CoA Dehydrogenase , Animals , Biological Transport , Carnitine O-Palmitoyltransferase/biosynthesis , Carnitine O-Palmitoyltransferase/genetics , Enzyme Activation , Fatty Acid Desaturases/biosynthesis , Fatty Acid Desaturases/genetics , Fatty Acids/metabolism , Glucose/metabolism , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Hypertrophy, Left Ventricular/genetics , In Vitro Techniques , Male , Models, Cardiovascular , Monosaccharide Transport Proteins/metabolism , Oxidation-Reduction , Phosphates/metabolism , Pressure , RNA, Messenger/biosynthesis , Rats , Rats, Wistar
17.
J Clin Invest ; 108(8): 1167-74, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11602624

ABSTRACT

Metformin is a widely used drug for treatment of type 2 diabetes with no defined cellular mechanism of action. Its glucose-lowering effect results from decreased hepatic glucose production and increased glucose utilization. Metformin's beneficial effects on circulating lipids have been linked to reduced fatty liver. AMP-activated protein kinase (AMPK) is a major cellular regulator of lipid and glucose metabolism. Here we report that metformin activates AMPK in hepatocytes; as a result, acetyl-CoA carboxylase (ACC) activity is reduced, fatty acid oxidation is induced, and expression of lipogenic enzymes is suppressed. Activation of AMPK by metformin or an adenosine analogue suppresses expression of SREBP-1, a key lipogenic transcription factor. In metformin-treated rats, hepatic expression of SREBP-1 (and other lipogenic) mRNAs and protein is reduced; activity of the AMPK target, ACC, is also reduced. Using a novel AMPK inhibitor, we find that AMPK activation is required for metformin's inhibitory effect on glucose production by hepatocytes. In isolated rat skeletal muscles, metformin stimulates glucose uptake coincident with AMPK activation. Activation of AMPK provides a unified explanation for the pleiotropic beneficial effects of this drug; these results also suggest that alternative means of modulating AMPK should be useful for the treatment of metabolic disorders.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Multienzyme Complexes/metabolism , Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/pharmacology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Enzyme Activation/drug effects , Fatty Acids/metabolism , Gene Expression/drug effects , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , In Vitro Techniques , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Protein Kinase Inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Sterol Regulatory Element Binding Protein 1
18.
Diabetes ; 50(5): 921-7, 2001 May.
Article in English | MEDLINE | ID: mdl-11334434

ABSTRACT

Insulin-stimulated GLUT4 translocation is impaired in people with type 2 diabetes. In contrast, exercise results in a normal increase in GLUT4 translocation and glucose uptake in these patients. Several groups have recently hypothesized that exercise increases glucose uptake via an insulin-independent mechanism mediated by the activation of AMP-activated protein kinase (AMPK). If this hypothesis is correct, people with type 2 diabetes should have normal AMPK activation in response to exercise. Seven subjects with type 2 diabetes and eight matched control subjects exercised on a cycle ergometer for 45 min at 70% of maximum workload. Biopsies of vastus lateralis muscle were taken before exercise, after 20 and 45 min of exercise, and at 30 min postexercise. Blood glucose concentrations decreased from 7.6 to 4.77 mmol/l with 45 min of exercise in the diabetic group and did not change in the control group. Exercise significantly increased AMPK alpha2 activity 2.7-fold over basal at 20 min in both groups and remained elevated throughout the protocol, but there was no effect of exercise on AMPK alpha1 activity. Subjects with type 2 diabetes had similar protein expression of AMPK alpha1, alpha2, and beta1 in muscle compared with control subjects. AMPK alpha2 was shown to represent approximately two-thirds of the total alpha mRNA in the muscle from both groups. In conclusion, people with type 2 diabetes have normal exercise-induced AMPK alpha2 activity and normal expression of the alpha1, alpha2 and beta1 isoforms. Pharmacological activation of AMPK may be an attractive target for the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Exercise/physiology , Multienzyme Complexes/metabolism , Muscle Proteins , Muscle, Skeletal/physiopathology , Physical Exertion/physiology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/enzymology , Enzyme Activation , Gene Expression Regulation, Enzymologic , Glucose Transporter Type 4 , Glycated Hemoglobin/analysis , Glycogen/metabolism , Humans , Kinetics , Male , Middle Aged , Monosaccharide Transport Proteins/metabolism , Multienzyme Complexes/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Reference Values , Rest/physiology , Transcription, Genetic
19.
Am J Physiol Endocrinol Metab ; 280(5): E677-84, 2001 May.
Article in English | MEDLINE | ID: mdl-11287349

ABSTRACT

The AMP-activated protein kinase (AMPK) has been hypothesized to mediate contraction and 5-aminoimidazole-4-carboxamide 1-beta-D-ribonucleoside (AICAR)-induced increases in glucose uptake in skeletal muscle. The purpose of the current study was to determine whether treadmill exercise and isolated muscle contractions in rat skeletal muscle increase the activity of the AMPK alpha 1 and AMPK alpha 2 catalytic subunits in a dose-dependent manner and to evaluate the effects of the putative AMPK inhibitors adenine 9-beta-D-arabinofuranoside (ara-A), 8-bromo-AMP, and iodotubercidin on AMPK activity and 3-O-methyl-D-glucose (3-MG) uptake. There were dose-dependent increases in AMPK alpha 2 activity and 3-MG uptake in rat epitrochlearis muscles with treadmill running exercise but no effect of exercise on AMPK alpha1 activity. Tetanic contractions of isolated epitrochlearis muscles in vitro significantly increased the activity of both AMPK isoforms in a dose-dependent manner and at a similar rate compared with increases in 3-MG uptake. In isolated muscles, the putative AMPK inhibitors ara-A, 8-bromo-AMP, and iodotubercidin fully inhibited AICAR-stimulated AMPK alpha 2 activity and 3-MG uptake but had little effect on AMPK alpha 1 activity. In contrast, these compounds had absent or minimal effects on contraction-stimulated AMPK alpha 1 and -alpha 2 activity and 3-MG uptake. Although the AMPK alpha 1 and -alpha 2 isoforms are activated during tetanic muscle contractions in vitro, in fast-glycolytic fibers, the activation of AMPK alpha 2-containing complexes may be more important in regulating exercise-mediated skeletal muscle metabolism in vivo. Development of new compounds will be required to study contraction regulation of AMPK by pharmacological inhibition.


Subject(s)
Glucose/pharmacokinetics , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-O-Methylglucose/pharmacokinetics , AMP-Activated Protein Kinases , Animals , Enzyme Inhibitors/pharmacology , Glucose/antagonists & inhibitors , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Motor Activity/physiology , Multienzyme Complexes/antagonists & inhibitors , Muscle Contraction/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...