Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Clin Endocrinol Metab ; 97(3): E329-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238402

ABSTRACT

CONTEXT: Indications of adipose tissue dysfunction correlate with systemic insulin resistance and type 2 diabetes. It has been suggested that a defect in adipose tissue turnover may be involved in the development of these disorders. Whether this dysfunction causes or exacerbates systemic insulin resistance is not fully understood. OBJECTIVES, PARTICIPANTS, AND MEASURES: We tested whether the expression of members of the mitogenic ErbB family was reduced in adipose tissue of insulin-resistant individuals and whether ErbB1 and ErbB2 were involved in adipogenesis. Thirty-two women covering a wide range of body mass index values and insulin sensitivity participated in the cross-sectional portion of this study. We also studied preadipocytes isolated from 12 insulin-sensitive individuals to evaluate the impact of ErbB1 or ErbB2 inhibition on adipogenesis in vitro. For this purpose, we measured phospho-ErbB1 and phospho-ErbB2 levels using ELISA and the expression of peroxisome proliferator-activated receptor γ (PPARγ) and PPARγ-regulated genes by real-time PCR. RESULTS: Among the ErbB family members, only ErbB1 expression was correlated with insulin sensitivity. Additionally, ErbB1 levels correlated positively with PPARγ and several PPARγ-regulated genes including acyl-coenzyme A synthetase long-chain family member 1 (ACSL1), adiponectin, adipose tissue triacylglycerol lipase (ATGL), diacylglycerol acyl transferase 1 (DGAT1), glycerol-3-phosphate dehydrogenase 1 (GPD1), and lipoprotein lipase (LPL), but negatively with CD36 and fatty acid-binding protein 4 (FABP4). In preadipocyte culture, ErbB1, but not ErbB2, inhibition was associated with a reduction in the expression of all the above-mentioned genes. CONCLUSIONS: These findings demonstrate a key role for ErbB1 in adipogenesis and suggest that lower ErbB1 protein abundance may lead to adipose tissue dysfunction.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , ErbB Receptors/metabolism , Insulin Resistance/physiology , Adipocytes/cytology , Adipocytes/metabolism , Adult , Body Mass Index , Cells, Cultured , Cross-Sectional Studies , Diabetes Mellitus, Type 2/genetics , ErbB Receptors/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Humans , Insulin/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Middle Aged , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation
2.
Endocrinology ; 150(7): 3040-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19246534

ABSTRACT

Uncoupling protein (UCP) 2 is a widely expressed mitochondrial protein whose precise function is still unclear but has been linked to mitochondria-derived reactive oxygen species production. Thus, the chronic absence of UCP2 has the potential to promote persistent reactive oxygen species accumulation and an oxidative stress response. Here, we show that Ucp2-/- mice on three highly congenic (N >10) strain backgrounds (C57BL/6J, A/J, 129/SvImJ), including two independently generated sources of Ucp2-null animals, all exhibit increased oxidative stress. Ucp2-null animals exhibit a decreased ratio of reduced glutathione to its oxidized form in blood and tissues that normally express UCP2, including pancreatic islets. Islets from Ucp2-/- mice exhibit elevated levels of numerous antioxidant enzymes, increased nitrotyrosine and F4/80 staining, but no change in insulin content. Contrary to results in Ucp2-/- mice of mixed 129/B6 strain background, glucose-stimulated insulin secretion in Ucp2-/- islets of each congenic strain was significantly decreased. These data show that the chronic absence of UCP2 causes oxidative stress, including in islets, and is accompanied by impaired glucose-stimulated insulin secretion.


Subject(s)
Insulin-Secreting Cells/metabolism , Ion Channels/deficiency , Mitochondrial Proteins/deficiency , Oxidative Stress/genetics , Animals , Glucose/pharmacology , Glutathione/blood , Glutathione Disulfide/blood , Insulin/metabolism , Insulin Secretion , Mice , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
3.
J Lipid Res ; 50(1): 59-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18698091

ABSTRACT

Vascular dysfunction in response to reactive oxygen species (ROS) plays an important role in the development and progression of atherosclerotic lesions. In most cells, mitochondria are the major source of cellular ROS during aerobic respiration. Under most conditions the rates of ROS formation and elimination are balanced through mechanisms that sense relative ROS levels. However, a chronic imbalance in redox homeostasis is believed to contribute to various chronic diseases, including atherosclerosis. Uncoupling protein-2 (UCP2) is a mitochondrial inner membrane protein shown to be a negative regulator of macrophage ROS production. In response to a cholesterol-containing atherogenic diet, C57BL/6J mice significantly increased expression of UCP2 in the aorta, while mice lacking UCP2, in the absence of any other genetic modification, displayed significant endothelial dysfunction following the atherogenic diet. Compared with wild-type mice, Ucp2(-/-) mice had decreased endothelial nitric oxide synthase, an increase in vascular cell adhesion molecule-1 expression, increased ROS production, and an impaired ability to increase total antioxidant capacity. These changes in Ucp2(-/-) mice were associated with increased aortic macrophage infiltration and more numerous and larger atherosclerotic lesions. These data establish that in the vasculature UCP2 functions as an adaptive antioxidant defense to protect against the development of atherosclerosis in response to a fat and cholesterol diet.


Subject(s)
Antioxidants/metabolism , Atherosclerosis/metabolism , Ion Channels/genetics , Ion Channels/physiology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Animals , Aorta/metabolism , Diet , Female , Homeostasis , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Oxidation-Reduction , Reactive Oxygen Species , Uncoupling Protein 2
4.
Mol Cell Biol ; 28(7): 2187-200, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18195045

ABSTRACT

The adipocyte integrates crucial information about metabolic needs in order to balance energy intake, storage, and expenditure. Whereas white adipose tissue stores energy, brown adipose tissue is a major site of energy dissipation through adaptive thermogenesis mediated by uncoupling protein 1 (UCP1) in mammals. In both white and brown adipose tissue, nuclear receptors and their coregulators, such as peroxisome proliferator-activated receptor gamma (PPARgamma) and PPARgamma coactivator 1alpha (PGC-1alpha), play key roles in regulating their development and metabolic functions. Here we show the unexpected role of liver X receptor alpha (LXRalpha) as a direct transcriptional inhibitor of beta-adrenergic receptor-mediated, cyclic AMP-dependent Ucp1 gene expression through its binding to the critical enhancer region of the Ucp1 promoter. The mechanism of inhibition involves the differential recruitment of the corepressor RIP140 to an LXRalpha binding site that overlaps with the PPARgamma/PGC-1alpha response element, resulting in the dismissal of PPARgamma. The ability of LXRalpha to dampen energy expenditure in this way provides another mechanism for maintaining a balance between energy storage and utilization.


Subject(s)
Adipocytes, Brown/metabolism , DNA-Binding Proteins/physiology , Ion Channels/physiology , Mitochondrial Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Body Temperature/genetics , Body Temperature/physiology , Cell Differentiation/drug effects , Cells, Cultured/cytology , Cells, Cultured/metabolism , Colforsin/pharmacology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Ion Channels/biosynthesis , Ion Channels/genetics , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Orphan Nuclear Receptors , Oxygen Consumption , PPAR gamma/metabolism , RNA, Small Interfering/pharmacology , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Uncoupling Protein 1
5.
J Biol Chem ; 282(12): 9244-50, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17251187

ABSTRACT

Catecholamine stimulation of beta-adrenergic receptors (betaAR) in adipocytes activates the cAMP-dependent protein kinase to promote liberation of fatty acids as a fuel source. The adipocyte beta3AR also activates extracellular signal-regulated kinases (ERK)-1 and -2 through direct recruitment and activation of Src kinase. This pathway together with cAMP-dependent protein kinase contributes to maximal beta3AR-stimulated lipolysis. In a search for other molecules that might associate with beta3AR upon agonist stimulation, we identified vimentin using a proteomics approach. Immunoprecipitation of beta3AR from adipocytes in the absence or presence of the beta3AR agonist CL316,243, followed by Western blotting for vimentin confirmed this specific interaction. Since vimentin has also been identified on lipid droplets, the functional consequences of blocking the expression or structural integrity of vimentin intermediate filaments on beta3AR regulation of ERK activation and lipolysis was assessed. Following disruption of intermediate filaments with beta,beta'-iminodipropionitrile, as confirmed by confocal microscopy, beta3AR-stimulated ERK activation was blocked, and lipolysis was reduced by more than 40%. Independently, depletion of vimentin by small hairpin RNA (shRNA) completely inhibited beta3AR-mediated ERK activation and significantly reduced lipolysis. By contrast, disruption of actin-containing microfilaments by cytochalasin D or microtubules by nocodazole had no effect on either lipolysis or ERK activation. These results indicate that vimentin plays an essential role in the signal transduction pathway from beta3AR to the activation ERK and its contribution to lipolysis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Adrenergic, beta-3/metabolism , Vimentin/chemistry , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Blotting, Western , Cyclic AMP-Dependent Protein Kinases/metabolism , Dioxoles/pharmacology , Enzyme Activation , Mice , Mice, Inbred C3H , Protein Binding , Signal Transduction , Vimentin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
J Biol Chem ; 281(49): 37794-802, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17032647

ABSTRACT

Catecholamine-stimulated lipolysis is primarily a beta-adrenergic and cAMP-dependent event. In previous studies we established that the beta(3)-adrenergic receptor (beta(3)AR) in adipocytes utilizes a unique mechanism to stimulate extracellular signal-regulated kinases 1 and 2 (ERK) by direct recruitment and activation of Src kinase. Therefore, we investigated the role of the ERK pathway in adipocyte metabolism and found that the beta(3)AR agonist CL316,243 regulates lipolysis through both cAMP-dependent protein kinase (PKA) and ERK. Inhibition of PKA activity completely eliminated lipolysis at low (subnanomolar) CL316,243 concentrations and by 75-80% at higher nanomolar concentrations. The remaining 20-25% of PKA-independent lipolysis, as well as ERK activation, was abolished by inhibiting the activity of either Src (PP2 or small interfering RNA), epidermal growth factor receptor (EGFR with AG1478 or small interfering RNA), or mitogen-activated protein kinase kinase 1 or 2 (MKK1/2 with PD098059). PD098059 inhibited lipolysis by 53% in mice as well. Finally, the effect of estradiol, a reported acute activator of ERK and lipolysis, was also totally prevented by PP2, AG1478, and PD098059. These results suggest that ERK activation by beta(3)AR depends upon Src and epidermal growth factor receptor kinase activities and is responsible for the PKA-independent portion of the lipolytic response. Together these results illustrate the distinct and complementary roles for PKA and ERK in catecholamine-stimulated lipolysis.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lipolysis/physiology , Receptors, Adrenergic, beta-3/metabolism , src-Family Kinases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adrenergic beta-3 Receptor Agonists , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dioxoles/pharmacology , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Isoquinolines/pharmacology , Lipolysis/drug effects , Mice , Quinazolines , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Sulfonamides/pharmacology , Tyrphostins/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
7.
J Biol Chem ; 280(52): 42731-7, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16272151

ABSTRACT

Hepatic gluconeogenesis is essential for maintaining blood glucose levels during fasting and is the major contributor to postprandial and fasting hyperglycemia in diabetes. Gluconeogenesis is a classic cAMP/protein kinase A-dependent process initiated by glucagon, which is elevated in the blood during fasting and in diabetes. In this study, we have shown that p38 mitogen-activated protein kinase (p38) was activated in liver by fasting and in primary hepatocytes by glucagon or forskolin. Fasting plasma glucose levels were reduced upon blockade of p38 with either a chemical inhibitor or small interference RNA in mice. In examining the mechanism, inhibition of p38 suppressed gluconeogenesis in liver, along with expression of key gluconeogenic genes, including phosphoenolpyruvate carboxykinase and glucose-6-phosphatase. Peroxisome proliferator-activated receptor gamma coactivator 1alpha and cAMP-response element-binding protein have been shown to be important mediators of hepatic gluconeogenesis. We have shown that inhibition of p38 prevented transcription of the PPARgamma coactivator 1alpha gene as well as phosphorylation of cAMP-response element-binding protein. Together, our results from in vitro and in vivo studies define a model in which cAMP-dependent activation of genes involved in gluconeogenesis is dependent upon the p38 pathway, thus adding a new player to our evolving understanding of this physiology.


Subject(s)
Liver/metabolism , p38 Mitogen-Activated Protein Kinases/physiology , Acetylcysteine/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Silencing , Glucagon/metabolism , Gluconeogenesis , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , Hepatocytes/metabolism , Imidazoles/pharmacology , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Phosphorylation , Promoter Regions, Genetic , Pyridines/pharmacology , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Streptozocin/pharmacology , Trans-Activators/metabolism , Transcription Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mol Cell Biol ; 25(13): 5466-79, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15964803

ABSTRACT

The sympathetic nervous system regulates the activity and expression of uncoupling protein 1 (UCP1) through the three beta-adrenergic receptor subtypes and their ability to raise intracellular cyclic AMP (cAMP) levels. Unexpectedly, we recently discovered that the cAMP-dependent regulation of multiple genes in brown adipocytes, including Ucp1, occurred through the p38 mitogen-activated protein kinases (MAPK) (W. Cao, K. W. Daniel, J. Robidoux, P. Puigserver, A. V. Medvedev, X. Bai, L. M. Floering, B. M. Spiegelman, and S. Collins, Mol. Cell. Biol. 24:3057-3067, 2004). However, no well-defined pathway linking cAMP accumulation or cAMP-dependent protein kinase (PKA) to p38 MAPK has been described. Therefore, in the present study using both in vivo and in vitro models, we have initiated a retrograde approach to define the required components, beginning with the p38 MAPK isoforms themselves and the MAP kinase kinase(s) that regulates them. Our strategy included ectopic expression of wild-type and mutant kinases as well as targeted inhibition of gene expression using small interfering RNA. The results indicate that the beta-adrenergic receptors and PKA lead to a highly selective activation of the p38alpha isoform of MAPK, which in turn promotes Ucp1 gene transcription. In addition, this specific activation of p38alpha relies solely on the presence of MAP kinase kinase 3, despite the expression in brown fat of MKK3, -4, and -6. Finally, of the three scaffold proteins of the JIP family expressed in brown adipocytes, only JIP2 co-immunoprecipitates p38alpha MAPK and MKK3. Therefore, in the brown adipocyte the recently described scaffold protein JIP2 assembles the required factors MKK3 and p38alpha MAPK linking PKA to the control of thermogenic gene expression.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation , MAP Kinase Kinase 3/metabolism , Membrane Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/cytology , Animals , Blotting, Western , Cell Line , Enzyme Activation , Genes, Reporter , Ion Channels , Isoenzymes/metabolism , Luciferases/metabolism , MAP Kinase Kinase 3/genetics , Mice , Mice, Inbred Strains , Mitochondrial Proteins , Mutation , Phosphorylation , Precipitin Tests , Thermogenesis/physiology , Uncoupling Protein 1 , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/genetics
9.
J Biol Chem ; 280(19): 19062-9, 2005 May 13.
Article in English | MEDLINE | ID: mdl-15757894

ABSTRACT

One of the phenotypes of mice with targeted disruption of the uncoupling protein-2 gene (Ucp2-/-) is greater macrophage phagocytic activity and free radical production, resulting in a striking resistance to infectious microorganisms. In this study, the molecular mechanisms of this enhanced immune response were investigated. We found that levels of nitric oxide measured in either plasma or isolated macrophages from Ucp2-/- mice are significantly elevated in response to bacterial lipopolysaccharide challenge compared with similarly treated Ucp2+/+ mice. Likewise, expression of inducible nitric-oxide synthase and inflammatory cytokines is higher in Ucp2-/- mice in vivo and in vitro. Key steps in the activation cascade of nuclear factor (NF)-kappa B, including I kappa B kinase and nuclear translocation of NF-kappa B subunits, are all remarkably enhanced in Ucp2-/- mice, most notably even under basal conditions. The elevated basal activity of I kappa B kinase in macrophages from Ucp2-/- mice can be blocked by cell-permeable inhibitors of superoxide and hydrogen peroxide generation, but not by a specific inhibitor for inducible nitric-oxide synthase. Isolated mitochondria from Ucp2-/- cells produced more superoxide/hydrogen peroxide. We conclude that mitochrondrially derived reactive oxygen from Ucp2-/- cells constitutively activates NF-kappa B, resulting in a "primed" state to both potentiate and amplify the inflammatory response upon subsequent stimulation.


Subject(s)
Cytokines/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , NF-kappa B/metabolism , Nitric Oxide/metabolism , Algorithms , Animals , Blotting, Northern , Blotting, Western , Cyclooxygenase 2 , Cytosol/metabolism , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Free Radicals , Genotype , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , I-kappa B Kinase , Inflammation , Ion Channels , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Models, Biological , NADPH Oxidases/chemistry , Nitrates/chemistry , Nitrates/metabolism , Nitric Oxide/chemistry , Nitric Oxide Synthase/metabolism , Nitrites/chemistry , Nitrites/metabolism , Oligonucleotide Array Sequence Analysis , Oxygen/metabolism , Phenotype , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Serine-Threonine Kinases/chemistry , Spleen/metabolism , Subcellular Fractions , Superoxides/metabolism , Time Factors , Uncoupling Protein 2
10.
Mol Cell Biol ; 24(7): 3057-67, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15024092

ABSTRACT

It is well established that catecholamine-stimulated thermogenesis in brown fat requires beta-adrenergic elevations in cyclic AMP (cAMP) to increase expression of the uncoupling protein 1 (UCP1) gene. However, little is known about the downstream components of the signaling cascade or the relevant transcription factor targets thereof. Here we demonstrate that cAMP- and protein kinase A-dependent activation of p38 mitogen-activated protein kinase (MAPK) in brown adipocytes is an indispensable step in the transcription of the UCP1 gene in mice. By phosphorylating activating transcription factor 2 (ATF-2) and peroxisome proliferator-activated receptor gamma (PPARgamma) coativator 1alpha (PGC-1alpha), members of two distinct nuclear factor families, p38 MAPK controls the expression of the UCP1 gene through their respective interactions with a cAMP response element and a PPAR response element that both reside within a critical enhancer motif of the UCP1 gene. Activation of ATF-2 by p38 MAPK additionally serves as the cAMP sensor that increases expression of the PGC-1alpha gene itself in brown adipose tissue. In conclusion, our findings illustrate that by orchestrating the activity of multiple transcription factors, p38 MAPK is a central mediator of the cAMP signaling mechanism of brown fat that promotes thermogenesis.


Subject(s)
Adipose Tissue, Brown/physiology , Carrier Proteins/genetics , Cyclic AMP/metabolism , Gene Expression Regulation , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Transcription, Genetic , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cells, Cultured , Cold Temperature , Enhancer Elements, Genetic , Enzyme Activation , Enzyme Inhibitors/metabolism , Humans , Ion Channels , Mice , Mice, Inbred C57BL , Mitochondrial Proteins , Random Allocation , Signal Transduction/physiology , Thermogenesis/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Agents , Uncoupling Protein 1 , p38 Mitogen-Activated Protein Kinases
11.
J Biol Chem ; 277(45): 42639-44, 2002 Nov 08.
Article in English | MEDLINE | ID: mdl-12205102

ABSTRACT

Current evidence suggests that uncoupling protein-2 (UCP2) is a regulator of insulin secretion. It is also known that chronic exposure of pancreatic islets to free fatty acids (FFAs) blunts glucose-stimulated insulin secretion and is accompanied by elevated levels of UCP2. However, the mechanisms regulating expression of UCP2 in beta-cells are unknown. Here, we show that UCP2 mRNA and protein levels were increased after a 48-h exposure of INS-1(832/13) beta-cells to oleic acid (0.5 mm) by activation of the UCP2 promoter. Furthermore, progressive deletions of the mouse UCP2 promoter (from -7.3 kb to +12 bp) indicated that an enhancer region (-86/-44) was responsible for both basal and FFA-stimulated UCP2 gene transcription. This enhancer contains tightly clustered Sp1, sterol regulatory element (SRE), and double E-Box elements. While all three sequence motifs were required for basal activity of the UCP2 promoter, the mutations in either the SRE or the E-Box elements eliminated the response to FFAs. The SRE and sterol regulatory element binding protein-1 (SREBP1) appear to be crucial for the response of the UCP2 gene to FFAs, since overexpression of the nuclear forms of the SREBPs increased UCP2 promoter activity by 7-10-fold and restored the ability of E-Box mutants to respond to oleic acid. These data support a model in which SREBP is the major modulator of UCP2 gene transcription by FFA, while E-Box binding factors play a supportive role.


Subject(s)
Islets of Langerhans/physiology , Membrane Transport Proteins , Mitochondrial Proteins , Oleic Acid/pharmacology , Proteins/genetics , Transcription Factors , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Ion Channels , Islets of Langerhans/drug effects , Mutagenesis , Mutagenesis, Site-Directed , Point Mutation , Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Deletion , Sterol Regulatory Element Binding Protein 1 , Transfection , Uncoupling Agents/metabolism , Uncoupling Protein 2
12.
Bioorg Med Chem Lett ; 12(2): 209-12, 2002 Jan 21.
Article in English | MEDLINE | ID: mdl-11755356

ABSTRACT

A series of 13 novel hybrid molecules designed to possess both free radical scavenging activity and to stimulate the beta(3)-adrenoceptors in order to improve antidiabetic effect and to restore insulin sensitivity was synthesized and evaluated. Compounds were of quinolyl-, isoquinolyl-, pyridoindolyl- or carbazolyloxypropanolamine structure with a terminal amino group of benzopyranolyl-, di-tert-butylphenolyl- or methoxyindolyl-type. Some of the products possessed both the expected activities.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Free Radical Scavengers/pharmacology , Propanolamines/pharmacology , Adrenergic beta-Agonists/chemistry , Animals , COS Cells , Free Radical Scavengers/chemistry , Humans , Propanolamines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...