Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Diabetologia ; 54(6): 1457-67, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21442160

ABSTRACT

AIMS/HYPOTHESIS: We examined in skeletal muscle (1) whether fatty acid transport protein (FATP) 1 channels long-chain fatty acid (LCFA) to specific metabolic fates in rats; and (2) whether FATP1-mediated increases in LCFA uptake exacerbate the development of diet-induced insulin resistance in mice. We also examined whether FATP1 is altered in insulin-resistant obese Zucker rats. METHODS: LCFA uptake, oxidation and triacylglycerol esterification rates were measured in control and Fatp1-transfected soleus muscles to determine FATP1-mediated lipid handling. The effects of FATP1 on insulin sensitivity and triacylglycerol accumulation were determined in high-fat diet-fed wild-type mice and in muscle-specific Fatp1 (also known as Slc27a1) overexpressing transgenic mice driven by the muscle creatine kinase (Mck [also known as Ckm]) promoter. We also examined the relationship between FATP1 and both fatty acid transport and metabolism in insulin-resistant obese Zucker rats. RESULTS: Transient Fatp1 overexpression in soleus muscle increased (p < 0.05) palmitate transport (24%) and oxidation (35%), without altering triacylglycerol esterification or the intrinsic rate of palmitate oxidation in isolated mitochondria. In Mck/Fatp1 animals, Fatp1 mRNA and 15-(p-iodophenyl)-3-R,S-methylpentadecanoic acid uptake in skeletal muscle were upregulated (75%). However, insulin sensitivity and intramuscular triacylglycerol content did not differ between wild-type and Mck/Fatp1 mice following a 16 week high-fat diet. In insulin-resistant obese Zucker rats, LCFA transport and triacylglycerol accumulation were increased (85% and 24%, respectively), but this was not attributable to Fatp1 expression, as neither total cellular nor sarcolemmal FATP1 content were altered. CONCLUSIONS/INTERPRETATION: Overexpression of Fatp1 in skeletal muscle increased the rate of LCFA transport and channelled these lipids to oxidation, not to intramuscular lipid accumulation. Therefore, skeletal muscle FATP1 overabundance does not predispose animals to diet-induced insulin resistance.


Subject(s)
Dietary Fats/adverse effects , Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Animals , Disease Models, Animal , Female , Mice , Mice, Transgenic , Mitochondria, Muscle/metabolism , Obesity/metabolism , Obesity/physiopathology , Oxidation-Reduction , Palmitates/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Triglycerides/metabolism
2.
Clin Pharmacol Ther ; 89(2): 234-42, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21191377

ABSTRACT

We studied the time course for the reversal of rifampin's effect on the pharmacokinetics of oral midazolam (a cytochrome P450 (CYP) 3A4 substrate) and digoxin (a P-glycoprotein (P-gp) substrate). Rifampin increased midazolam metabolism, greatly reducing the area under the concentration-time curve (AUC(0-∞)). The midazolam AUC(0-∞) returned to baseline with a half-life of ~8 days. Rifampin's effect on the AUC(0-3 h) of digoxin was biphasic: the AUC(0-3 h) increased with concomitant dosing of the two drugs but decreased when digoxin was administered after rifampin. Digoxin was found to be a weak substrate of organic anion-transporting polypeptide (OATP) 1B3 in transfected cells. Although the drug was transported into isolated hepatocytes, it is not likely that this transport was through OATP1B3 because the transport was not inhibited by rifampin. However, rifampin did inhibit the P-gp-mediated transport of digoxin with a half-maximal inhibitory concentration (IC(50)) below anticipated gut lumen concentrations, suggesting that rifampin inhibits digoxin efflux from the enterocyte to the intestinal lumen. Pharmacokinetic modeling suggested that the effects on digoxin are consistent with a combination of inhibitory and inductive effects on gut P-gp. These results suggest modifications to drug-drug interaction (DDI) trial designs.


Subject(s)
Digoxin/pharmacokinetics , Midazolam/pharmacokinetics , Research Design , Rifampin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Adult , Area Under Curve , Biological Transport , Drug Interactions , Humans , Male , Middle Aged , Models, Biological , Organic Anion Transporters, Sodium-Independent/physiology , Solute Carrier Organic Anion Transporter Family Member 1B3
3.
Nat Med ; 7(8): 941-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11479627

ABSTRACT

Adiponectin is an adipocyte-derived hormone. Recent genome-wide scans have mapped a susceptibility locus for type 2 diabetes and metabolic syndrome to chromosome 3q27, where the gene encoding adiponectin is located. Here we show that decreased expression of adiponectin correlates with insulin resistance in mouse models of altered insulin sensitivity. Adiponectin decreases insulin resistance by decreasing triglyceride content in muscle and liver in obese mice. This effect results from increased expression of molecules involved in both fatty-acid combustion and energy dissipation in muscle. Moreover, insulin resistance in lipoatrophic mice was completely reversed by the combination of physiological doses of adiponectin and leptin, but only partially by either adiponectin or leptin alone. We conclude that decreased adiponectin is implicated in the development of insulin resistance in mouse models of both obesity and lipoatrophy. These data also indicate that the replenishment of adiponectin might provide a novel treatment modality for insulin resistance and type 2 diabetes.


Subject(s)
Adipose Tissue/physiopathology , Insulin Resistance , Intercellular Signaling Peptides and Proteins , Obesity/physiopathology , Proteins/physiology , Adiponectin , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , Leptin/metabolism , Mice , Molecular Sequence Data , Oxidation-Reduction , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/physiology , Triglycerides/metabolism
4.
Biochem Soc Trans ; 29(Pt 2): 68-72, 2001 May.
Article in English | MEDLINE | ID: mdl-11356129

ABSTRACT

Leptin is a hormone that is secreted by adipose cells in proportion to adipose mass, and therefore a low leptin level signifies depletion of energy stores. It has been proposed that leptin is one of the signals controlling sexual maturation. For example, humans and rodents lacking leptin fail to undergo complete puberty, while overexpression of leptin in mice causes early puberty. The placenta also produces leptin in human pregnancy, increasing the amount in the maternal circulation. The effects of the increased leptin levels during pregnancy are not clear. In contrast, the mouse placenta does not produce endocrinologically significant amounts of leptin. The mouse placenta does secrete a leptin-binding protein, the production of which correlates with a large increase in maternal leptin levels. The physiology of leptin during pregnancy and fetal development differs significantly between species, and is not well understood in any.


Subject(s)
Embryonic and Fetal Development , Fetus/metabolism , Leptin/metabolism , Pregnancy/metabolism , Animals , Energy Metabolism , Female , Homeostasis , Humans , Sexual Maturation
5.
Diabetes ; 50(6): 1440-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375346

ABSTRACT

Lipoatrophic diabetes is caused by a deficiency of adipose tissue and is characterized by severe insulin resistance, hypoleptinemia, and hyperphagia. The A-ZIP/F-1 mouse (A-ZIPTg/+) is a model of severe lipoatrophic diabetes and is insulin resistant, hypoleptinemic, hyperphagic, and shows severe hepatic steatosis. We have also produced transgenic "skinny" mice that have hepatic overexpression of leptin (LepTg/+) and no adipocyte triglyceride stores, and are hypophagic and show increased insulin sensitivity. To explore the pathophysiological and therapeutic roles of leptin in lipoatrophic diabetes, we crossed LepTg/+ and A-ZIPTg/+ mice, producing doubly transgenic mice (LepTg/+:A-ZIPTg/+) virtually lacking adipose tissue but having greatly elevated leptin levels. The LepTg/+:A-ZIPTg/+ mice were hypophagic and showed improved hepatic steatosis. Glucose and insulin tolerance tests revealed increased insulin sensitivity, comparable to LepTg/+ mice. These effects were stable over at least 6 months of age. Pair-feeding the A-ZIPTg/+ mice to the amount of food consumed by LepTg/+:A-ZIPTg/+ mice did not improve their insulin resistance, diabetes, or hepatic steatosis, demonstrating that the beneficial effects of leptin were not due to the decreased food intake. Continuous leptin administration that elevates plasma leptin concentrations to those of LepTg/+:A-ZIPTg/+ mice also effectively improved hepatic steatosis and the disorder of glucose and lipid metabolism in A-ZIP/F-1 mice. These data demonstrate that leptin can improve the insulin resistance and diabetes of a mouse model of severe lipoatrophic diabetes, suggesting that leptin may be therapeutically useful in the long-term treatment of lipoatrophic diabetes.


Subject(s)
Diabetes Mellitus, Lipoatrophic/drug therapy , Diabetes Mellitus, Lipoatrophic/physiopathology , Insulin Resistance , Leptin/therapeutic use , Animals , Blood Glucose/analysis , Body Weight/drug effects , Diabetes Mellitus, Lipoatrophic/pathology , Eating , Gene Expression , Infusion Pumps , Injections , Leptin/administration & dosage , Leptin/blood , Leptin/genetics , Lipids/blood , Mice , Mice, Inbred Strains , Mice, Transgenic/genetics , Organ Size , Transgenes/genetics
6.
Proc Natl Acad Sci U S A ; 98(11): 6494-9, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11371650

ABSTRACT

Perilipin coats the lipid droplets of adipocytes and is thought to have a role in regulating triacylglycerol hydrolysis. To study the role of perilipin in vivo, we have created a perilipin knockout mouse. Perilipin null (peri(-/-)) and wild-type (peri(+/+)) mice consume equal amounts of food, but the adipose tissue mass in the null animals is reduced to approximately 30% of that in wild-type animals. Isolated adipocytes of perilipin null mice exhibit elevated basal lipolysis because of the loss of the protective function of perilipin. They also exhibit dramatically attenuated stimulated lipolytic activity, indicating that perilipin is required for maximal lipolytic activity. Plasma leptin concentrations in null animals were greater than expected for the reduced adipose mass. The peri(-/-) animals have a greater lean body mass and increased metabolic rate but they also show an increased tendency to develop glucose intolerance and peripheral insulin resistance. When fed a high-fat diet, the perilipin null animals are resistant to diet-induced obesity but not to glucose intolerance. The data reveal a major role for perilipin in adipose lipid metabolism and suggest perilipin as a potential target for attacking problems associated with obesity.


Subject(s)
Adipocytes/metabolism , Leptin/biosynthesis , Obesity/metabolism , Phosphoproteins/physiology , Adipocytes/cytology , Adipose Tissue/metabolism , Animals , Blood Glucose/analysis , Carrier Proteins , Cell Differentiation , Dietary Fats/metabolism , Female , Lipolysis , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption , Peptides/metabolism , Perilipin-1 , Perilipin-2 , Phosphoproteins/genetics , Sterol Esterase/metabolism , Thinness , Triglycerides/metabolism
7.
Int J Obes Relat Metab Disord ; 24 Suppl 4: S11-4, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11126232

ABSTRACT

The A-ZIP/F-1 mouse is lacking virtually all white adipose tissue. Like humans with extensive deficiencies of adipose tissue, the A-ZIP/F-1 mice develop a severe form of insulin resistant diabetes. We have studied the physiology of the A-ZIP/F-1 mice. Their adaptation to fasting is notable for its rapidity and the use of torpor, a hibernation-like state, to minimize energy needs. Transplantation of adipose tissue reversed the metabolic manifestations in the mice, demonstrating that the lack of adipose tissue is the cause of the insulin resistance. Leptin replacement is not very effective in reversing the diabetes of the A-ZIP/F-1 mice, which contrasts with its efficacy in the aP2-SREBP-lc mouse.


Subject(s)
Adipose Tissue , Diabetes Mellitus, Lipoatrophic , Disease Models, Animal , Insulin Resistance , Mice, Transgenic/physiology , Adipose Tissue/pathology , Adipose Tissue/transplantation , Animals , Body Composition , Diabetes Mellitus, Lipoatrophic/etiology , Diabetes Mellitus, Lipoatrophic/surgery , Fasting/physiology , Humans , Leptin/physiology , Mice , Phenotype , Transcription Factors
8.
Diabetes ; 49(11): 1910-6, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078459

ABSTRACT

Stimulation of beta3-adrenergic receptors increases metabolic rate via lipolysis in white adipose tissue (WAT) and thermogenesis in brown adipose tissue (BAT). Other acute effects include decreased gastrointestinal motility and food intake and increased insulin secretion. Chronic treatment with a beta3 agonist ameliorates diabetes and obesity in rodents. We studied the effects of beta3 stimulation in A-ZIP/F-1 mice, which have virtually no WAT, a reduced amount of BAT, severe insulin resistance, and diabetes. In contrast with wild-type mice, treatment of A-ZIP/F-1 mice with CL316243, a beta3-adrenergic agonist, did not increase O2 consumption. A single dose of CL316243 produced a 2-fold increase in serum free fatty acids, a 53-fold increase in insulin, and a 2.4-fold decrease in glucose levels in wild-type mice but no change in A-ZIP/F-1 animals. The A-ZIP/F-1 mice also did not show reduced gastrointestinal motility or 24-h food intake during beta3 stimulation. Chronic administration of CL316243 to the A-ZIP/F-1 mice did not improve their thermogenesis, hyperglycemia, or hyperinsulinemia. Thus, all of the beta3 effects studied were absent in the lipoatrophic A-ZIP/F-1 mice, including the effects on nonadipose tissues. From these results, we suggest that all of the effects of beta3 agonists are initiated at the adipocyte with the nonadipose effects being secondary events presumably mediated by signals from adipose tissue.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/physiology , Adrenergic beta-Agonists/pharmacology , Adipose Tissue/pathology , Adipose Tissue, Brown/chemistry , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/pathology , Animals , Atrophy , Blood Glucose/metabolism , Carrier Proteins/genetics , Diabetes Mellitus/genetics , Dioxoles/pharmacology , Eating/drug effects , Fatty Acids, Nonesterified/blood , Female , Gastrointestinal Motility/drug effects , Insulin/blood , Insulin Resistance , Ion Channels , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mitochondrial Proteins , Oxygen Consumption , RNA, Messenger/analysis , Receptors, Adrenergic, beta-3/genetics , Thermogenesis/drug effects , Uncoupling Protein 1
9.
Trends Endocrinol Metab ; 11(10): 410-6, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11091118

ABSTRACT

The lipoatrophy syndromes are a heterogeneous group of syndromes characterized by a paucity of adipose tissue. Severe lipoatrophy is associated with insulin-resistant diabetes mellitus (DM). The loss of adipose tissue can have a genetic, immune, or infectious/drug-associated etiology. Causative mutations have been identified in patients for one form of partial lipoatrophy--Dunnigan-type familial partial lipodystrophy. Experiments using lipoatrophic mice demonstrate that the diabetes results from the lack of fat and that leptin deficiency is a contributing factor. Thiazolidinedione therapy improves metabolic control in lipoatrophic patients; the efficacy of leptin treatment is currently being investigated.


Subject(s)
Adipose Tissue/physiopathology , Diabetes Mellitus, Lipoatrophic/physiopathology , Adipose Tissue/pathology , Animals , Diabetes Mellitus, Lipoatrophic/genetics , Humans , Mice
10.
J Clin Invest ; 106(10): 1221-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11086023

ABSTRACT

There is uncertainty about the site(s) of action of the antidiabetic thiazolidinediones (TZDs). These drugs are agonist ligands of the transcription factor PPAR gamma, which is abundant in adipose tissue but is normally present at very low levels in liver and muscle. We have studied the effects of TZDs in A-ZIP/F-1 mice, which lack white adipose tissue. The A-ZIP/F-1 phenotype strikingly resembles that of humans with severe lipoatrophic diabetes, including the lack of fat, marked insulin resistance and hyperglycemia, hyperlipidemia, and fatty liver. Rosiglitazone or troglitazone treatment did not reduce glucose or insulin levels, suggesting that white adipose tissue is required for the antidiabetic effects of TZDs. However, TZD treatment was effective in lowering circulating triglycerides and increasing whole body fatty acid oxidation in the A-ZIP/F-1 mice, indicating that this effect occurs via targets other than white adipose tissue. A-ZIP/F-1 mice have markedly increased liver PPAR gamma mRNA levels, which may be a general property of fatty livers. Rosiglitazone treatment increased the triglyceride content of the steatotic livers of A-ZIP/F-1 and ob/ob mice, but not the "lean" livers of fat-transplanted A-ZIP/F-1 mice. In light of this evidence that rosiglitazone acts differently in steatotic livers, the effects of rosiglitazone, particularly on hepatic triglyceride levels, should be examined in humans with hepatic steatosis.


Subject(s)
Adipose Tissue/physiology , Chromans/therapeutic use , Diabetes Mellitus, Lipoatrophic/drug therapy , Hypoglycemic Agents/therapeutic use , Thiazoles/therapeutic use , Thiazolidinediones , Triglycerides/metabolism , Animals , Blood Glucose , Diabetes Mellitus, Lipoatrophic/metabolism , Disease Models, Animal , Female , Insulin/metabolism , Ligands , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Cytoplasmic and Nuclear/genetics , Respiratory Function Tests , Rosiglitazone , Transcription Factors/genetics , Troglitazone
11.
Ann Intern Med ; 133(4): 263-74, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10929166

ABSTRACT

BACKGROUND: Troglitazone promotes adipocyte differentiation in vitro and increases insulin sensitivity in vivo. Therefore, troglitazone may have therapeutic benefit in lipoatrophic diabetes. OBJECTIVE: To determine whether troglitazone ameliorates hyperglycemia and hypertriglyceridemia or increases fat mass in lipoatrophic patients. DESIGN: Open-labeled prospective study. SETTING: United States and Canada. PATIENTS: 20 patients with various syndromes associated with lipoatrophy or lipodystrophy. INTERVENTION: 6 months of therapy with troglitazone, 200 to 600 mg/d. MEASUREMENTS: Levels of hemoglobin A1c triglycerides, free fatty acids, and insulin; respiratory quotient; percentage of body fat; liver volume; and regional fat mass. RESULTS: In the 13 patients with diabetes who completed 6 months of troglitazone therapy, hemoglobin A1c levels decreased by a mean of 2.8% (95% CI, 1.9% to 3.7%; P < 0.001). In all 19 study patients, fasting triglyceride levels decreased by 2.6 mmol/L (230 mg/dL) (CI, 0.7 to 4.5 mmol/L [62 to 398 mg/dL]; P = 0.019) and free fatty acid levels decreased by 325 micromol/L (CI, 135 to 515 micromol/L; P = 0.035). The respiratory quotient decreased by a mean of 0.12 (CI, 0.08 to 0.16; P < 0.001), suggesting that troglitazone promoted oxidation of fat. Body fat increased by a mean of 2.4 percentage points (CI, 1.3 to 4.5 percentage points; P = 0.044). Magnetic resonance imaging showed an increase in subcutaneous adipose tissue but not in visceral fat. In one patient, the serum alanine aminotransferase level increased eightfold during the 10th months of troglitazone treatment but normalized 3 months after discontinuation of treatment Liver biopsy revealed an eosinophilic infiltrate, suggesting hypersensitivity reaction as a cause of hepatotoxicity. CONCLUSION: Troglitazone therapy improved metabolic control and increased body fat in patients with lipoatrophic diabetes. The substantial benefits of troglitazone must be balanced against the risk for hepatotoxicity, which can occur relatively late in the treatment course.


Subject(s)
Chromans/therapeutic use , Hypoglycemic Agents/therapeutic use , Lipodystrophy/drug therapy , Thiazoles/therapeutic use , Thiazolidinediones , Adipose Tissue/anatomy & histology , Adipose Tissue/drug effects , Adult , Aged , Body Composition/drug effects , Chemical and Drug Induced Liver Injury/etiology , Child , Chromans/adverse effects , Drug Administration Schedule , Fatty Acids, Nonesterified/blood , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/adverse effects , Insulin/blood , Insulin Resistance/physiology , Lipodystrophy/blood , Lipodystrophy/physiopathology , Liver/anatomy & histology , Liver/drug effects , Male , Middle Aged , Prospective Studies , Respiratory Function Tests , Statistics, Nonparametric , Syndrome , Thiazoles/adverse effects , Triglycerides/blood , Troglitazone
12.
Mol Cell Biol ; 20(14): 5119-28, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10866668

ABSTRACT

To determine the physiological roles of peroxisome proliferator-activated receptor beta (PPARbeta), null mice were constructed by targeted disruption of the ligand binding domain of the murine PPARbeta gene. Homozygous PPARbeta-null term fetuses were smaller than controls, and this phenotype persisted postnatally. Gonadal adipose stores were smaller, and constitutive mRNA levels of CD36 were higher, in PPARbeta-null mice than in controls. In the brain, myelination of the corpus callosum was altered in PPARbeta-null mice. PPARbeta was not required for induction of mRNAs involved in epidermal differentiation induced by O-tetradecanoylphorbol-13-acetate (TPA). The hyperplastic response observed in the epidermis after TPA application was significantly greater in the PPARbeta-null mice than in controls. Inflammation induced by TPA in the skin was lower in wild-type mice fed sulindac than in similarly treated PPARbeta-null mice. These results are the first to provide in vivo evidence of significant roles for PPARbeta in development, myelination of the corpus callosum, lipid metabolism, and epidermal cell proliferation.


Subject(s)
Adipose Tissue/abnormalities , Body Constitution/genetics , Brain/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Skin/pathology , Transcription Factors/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Body Temperature/genetics , Brain/abnormalities , Brain/physiology , CD36 Antigens/genetics , CD36 Antigens/metabolism , Embryonic and Fetal Development/genetics , Fasting , Female , Hyperplasia , Inflammation/chemically induced , Inflammation/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myelin Sheath/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Sulindac/pharmacology , Tetradecanoylphorbol Acetate/toxicity , Transcription Factors/drug effects , Transcription Factors/metabolism
13.
J Biol Chem ; 275(21): 16251-7, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10748195

ABSTRACT

Uncoupling protein-3 (UCP3) is a mitochondrial protein that can diminish the mitochondrial membrane potential. Levels of muscle Ucp3 mRNA are increased by thyroid hormone and fasting. Ucp3 has been proposed to influence metabolic efficiency and is a candidate obesity gene. We have produced a Ucp3 knockout mouse to test these hypotheses. The Ucp3 (-/-) mice had no detectable immunoreactive UCP3 by Western blotting. In mitochondria from the knockout mice, proton leak was greatly reduced in muscle, minimally reduced in brown fat, and not reduced at all in liver. These data suggest that UCP3 accounts for much of the proton leak in skeletal muscle. Despite the lack of UCP3, no consistent phenotypic abnormality was observed. The knockout mice were not obese and had normal serum insulin, triglyceride, and leptin levels, with a tendency toward reduced free fatty acids and glucose. Knockout mice showed a normal circadian rhythm in body temperature and motor activity and had normal body temperature responses to fasting, stress, thyroid hormone, and cold exposure. The base-line metabolic rate and respiratory exchange ratio were the same in knockout and control mice, as were the effects of fasting, a beta3-adrenergic agonist (CL316243), and thyroid hormone on these parameters. The phenotype of Ucp1/Ucp3 double knockout mice was indistinguishable from Ucp1 single knockout mice. These data suggest that Ucp3 is not a major determinant of metabolic rate but, rather, has other functions.


Subject(s)
Carrier Proteins/genetics , Obesity/genetics , Thyroid Hormones/pharmacology , Adipose Tissue, Brown/metabolism , Adrenergic beta-Agonists/pharmacology , Age Factors , Animals , Body Temperature/genetics , Fasting , Ion Channels , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria, Liver/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Proteins , Phenotype , Protons , RNA, Messenger/metabolism , Uncoupling Protein 1 , Uncoupling Protein 3
15.
J Clin Invest ; 105(5): 615-23, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712433

ABSTRACT

Heterozygous disruption of Gnas, the gene encoding the stimulatory G-protein alpha subunit (G(s)alpha), leads to distinct phenotypes depending on whether the maternal (m-/+) or paternal (+/p-) allele is disrupted. G(s)alpha is imprinted, with the maternal allele preferentially expressed in adipose tissue. Hence, expression is decreased in m-/+ mice but normal in +/p- mice. M-/+ mice become obese, with increased lipid per cell in white and brown adipose tissue, whereas +/p- mice are thin, with decreased lipid in adipose tissue. These effects are not due to abnormalities in thyroid hormone status, food intake, or leptin secretion. +/p- mice are hypermetabolic at both ambient temperature (21 degrees C) and thermoneutrality (30 degrees C). In contrast, m-/+ mice are hypometabolic at ambient temperature and eumetabolic at thermoneutrality M-/+ and wild-type mice have similar dose-response curves for metabolic response to a beta(3)-adrenergic agonist, CL316243, indicating normal sensitivity of adipose tissue to sympathetic stimulation. Measurement of urinary catecholamines suggests that +/p- and m-/+ mice have increased and decreased activation of the sympathetic nervous system, respectively. This is to our knowledge the first animal model in which a single genetic defect leads to opposite effects on energy metabolism depending on parental inheritance. This probably results from deficiency of maternal- and paternal-specific Gnas gene products, respectively.


Subject(s)
Energy Metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Obesity/genetics , Adrenergic beta-Agonists/pharmacology , Alleles , Animals , Body Weight , Dioxoles/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gs/deficiency , Genomic Imprinting , Histocytochemistry , Leptin/blood , Lipids/blood , Male , Mice , Mice, Knockout , Obesity/blood , Phenotype , Thyroid Hormones/blood , Thyrotropin/blood
16.
J Biol Chem ; 275(12): 8456-60, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10722680

ABSTRACT

Insulin resistance is a major factor in the pathogenesis of type 2 diabetes and may be related to alterations in fat metabolism. Fatless mice have been created using dominant-negative protein (A-ZIP/F-1) targeted gene expression in the adipocyte and shown to develop diabetes. To understand the mechanism responsible for the insulin resistance in these mice, we conducted hyperinsulinemic-euglycemic clamps in awake fatless and wild type littermates before the development of diabetes and examined insulin action and signaling in muscle and liver. We found the fatless mice to be severely insulin-resistant, which could be attributed to defects in insulin action in muscle and liver. Both of these abnormalities were associated with defects in insulin activation of insulin receptor substrate-1 and -2-associated phosphatidylinositol 3-kinase activity and a 2-fold increase in muscle and liver triglyceride content. We also show that upon transplantation of fat tissue into these mice, triglyceride content in muscle and liver returned to normal as does insulin signaling and action. In conclusion, these results suggest that the development of insulin resistance in type 2 diabetes may be due to alterations in the partitioning of fat between the adipocyte and muscle/liver leading to accumulation of triglyceride in the latter tissues with subsequent impairment of insulin signaling and action.


Subject(s)
Adipose Tissue , Insulin Resistance/genetics , Insulin/pharmacology , Liver/metabolism , Muscles/metabolism , Transcription Factors/genetics , Animals , Body Composition , Glucose Clamp Technique , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism
17.
J Clin Invest ; 105(3): 271-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10675352

ABSTRACT

In lipoatrophic diabetes, a lack of fat is associated with insulin resistance and hyperglycemia. This is in striking contrast to the usual association of diabetes with obesity. To understand the underlying mechanisms, we transplanted adipose tissue into A-ZIP/F-1 mice, which have a severe form of lipoatrophic diabetes. Transplantation of wild-type fat reversed the hyperglycemia, dramatically lowered insulin levels, and improved muscle insulin sensitivity, demonstrating that the diabetes in A-ZIP/F-1 mice is caused by the lack of adipose tissue. All aspects of the A-ZIP/F-1 phenotype including hyperphagia, hepatic steatosis, and somatomegaly were either partially or completely reversed. However, the improvement in triglyceride and FFA levels was modest. Donor fat taken from parametrial and subcutaneous sites was equally effective in reversing the phenotype. The beneficial effects of transplantation were dose dependent and required near-physiological amounts of transplanted fat. Transplantation of genetically modified fat into A-ZIP/F-1 mice is a new and powerful technique for studying adipose physiology and the metabolic and endocrine communication between adipose tissue and the rest of the body.


Subject(s)
Adipose Tissue/transplantation , Diabetes Mellitus, Lipoatrophic/surgery , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Lipoatrophic/blood , Diabetes Mellitus, Lipoatrophic/physiopathology , Fatty Acids/blood , Gene Expression Regulation , Gene Transfer Techniques , Insulin Resistance , Mice , Triglycerides/blood
19.
Proc Natl Acad Sci U S A ; 96(25): 14623-8, 1999 Dec 07.
Article in English | MEDLINE | ID: mdl-10588755

ABSTRACT

We tested the effect of chronic leptin treatment on fasting-induced torpor in leptin-deficient A-ZIP/F-1 and ob/ob mice. A-ZIP/F-1 mice have virtually no white adipose tissue and low leptin levels, whereas ob/ob mice have an abundance of fat but no leptin. These two models allowed us to examine the roles of adipose tissue and leptin in the regulation of entry into torpor. Torpor is a short-term hibernation-like state that allows conservation of metabolic fuels. We first characterized the A-ZIP/F-1 animals, which have a 10-fold reduction in total body triglyceride stores. Upon fasting, A-ZIP/F-1 mice develop a lower metabolic rate and decreased plasma glucose, insulin, and triglyceride levels, with no increase in free fatty acids or beta-hydroxybutyrate. Unlike control mice, by 24 hr of fasting, they have nearly exhausted their triglycerides and are catabolizing protein. To conserve energy supplies during fasting, A-ZIP/F-1 (but not control) mice entered deep torpor, with a minimum core body temperature of 24 degrees C, 2 degrees C above ambient. In ob/ob mice, fasting-induced torpor was completely reversed by leptin treatment. In contrast, neither leptin nor thyroid hormone prevented torpor in A-ZIP/F-1 mice. These data suggest that there are at least two signals for entry into torpor in mice, a low leptin level and another signal that is independent of leptin and thyroid hormone levels. Studying rodent torpor provides insight into human torpor-like states such as near drowning in cold water and induced hypothermia for surgery.


Subject(s)
Adaptation, Physiological , Fasting/physiology , Leptin/physiology , Animals , Energy Metabolism , Leptin/deficiency , Liver Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Testosterone/blood , Thyroid Hormones/blood , Triglycerides/metabolism
20.
Int J Obes Relat Metab Disord ; 23 Suppl 6: S56-9, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10454126

ABSTRACT

The role of the thyroid gland in the regulation of metabolic rate has been known since the last century. The knowledge that thyroid hormones increase energy expenditure, in part by lowering metabolic efficiency, dates from the 1950s. Presumably thyroid hormones regulate energy expenditure and efficiency by controlling the rate of transcription of specific genes. However, the number, identity, and relative contributions of these genes are not known. The uncoupling proteins (UCPs) are obvious candidates to mediate thyroid thermogenesis. UCP1 is not a major contributor, since thyrotoxicosis decreases UCP1 expression and inactivates brown fat. Discovery of UCP3 and its regulation by T3 in muscle is an exciting observation, consistent with a role for UCP3 in thyroid thermogenesis. Since free fatty acids appear to regulate UCP3 expression and T3 stimulates lipolysis, further experiments are required to determine if T3 regulation of UCP3 expression is direct or not.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , Membrane Transport Proteins , Mitochondrial Proteins , Proteins/genetics , Thyroid Hormones/physiology , Animals , Basal Metabolism/physiology , Carrier Proteins/physiology , Energy Metabolism/physiology , Humans , Ion Channels , Membrane Proteins/physiology , Proteins/physiology , Uncoupling Protein 1 , Uncoupling Protein 2 , Uncoupling Protein 3
SELECTION OF CITATIONS
SEARCH DETAIL
...