Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Geochem Geophys Geosyst ; 17(2): 300-323, 2016 Feb.
Article in English | MEDLINE | ID: mdl-30123099

ABSTRACT

Rock and fluid samples were collected from three hydrothermal chimneys at the Endeavour Segment, Juan de Fuca Ridge to evaluate linkages among mineralogy, fluid chemistry, and microbial community composition within the chimneys. Mössbauer, midinfrared thermal emission, and visible-near infrared spectroscopies were utilized for the first time to characterize vent mineralogy, in addition to thin-section petrography, X-ray diffraction, and elemental analyses. A 282°C venting chimney from the Bastille edifice was composed primarily of sulfide minerals such as chalcopyrite, marcasite, and sphalerite. In contrast, samples from a 300°C venting chimney from the Dante edifice and a 321°C venting chimney from the Hot Harold edifice contained a high abundance of the sulfate mineral anhydrite. Geochemical modeling of mixed vent fluids suggested the oxic-anoxic transition zone was above 100°C at all three vents, and that the thermodynamic energy available for autotrophic microbial redox reactions favored aerobic sulfide and methane oxidation. As predicted, microbes within the Dante and Hot Harold chimneys were most closely related to mesophilic and thermophilic aerobes of the Betaproteobacteria and Gammaproteobacteria and sulfide-oxidizing autotrophic Epsilonproteobacteria. However, most of the microbes within the Bastille chimney were most closely related to mesophilic and thermophilic anaerobes of the Deltaproteobacteria, especially sulfate reducers, and anaerobic hyperthermophilic archaea. The predominance of anaerobes in the Bastille chimney indicated that other environmental factors promote anoxic conditions. Possibilities include the maturity or fluid flow characteristics of the chimney, abiotic Fe2+ and S2- oxidation in the vent fluids, or O2 depletion by aerobic respiration on the chimney outer wall.

2.
Int J Obes (Lond) ; 32 Suppl 4: S49-54, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18719599

ABSTRACT

Energy balance is monitored by the hypothalamus. Malonyl-CoA, an intermediate in fatty acid synthesis, serves as an indicator of energy status in the hypothalamic neurons. The cellular malonyl-CoA level is determined by its rate of synthesis, catalyzed by acetyl-CoA carboxylase (ACC), and rate of removal, by fatty acid synthase (FAS). Malonyl-CoA functions in the hypothalamic neurons that express orexigenic and anorexigenic neuropeptides. Inhibitors of FAS, administered systemically or intracerebroventricularly to mice, increase hypothalamic malony-CoA and suppress food intake. Recent evidence suggests that the changes of hypothalamic malonyl-CoA during feeding and fasting cycles are caused by changes in the phosphorylation state and activity of ACC mediated via 5'-AMP-activated protein kinase (AMPK). Stereotactic delivery of a viral malonyl-CoA decarboxylase (MCD) vector into the ventral hypothalamus lowers malonyl-CoA and increases food intake. Fasting decreases hypothalamic malonyl-CoA and refeeding increases hypothalamic malonyl-CoA, to alter feeding behavior in the predicted manner. Malonyl-CoA level is under the control of AMP kinase which phosphorylates/inactivates ACC. Malonyl-CoA is an inhibitor of carnitine palmitoyl-CoA transferase-1 (CPT1), an outer mitochondrial membrane enzyme that regulates entry into, and oxidation of fatty acids, by mitochondria. CPT1c, a recently discovered, brain-specific enzyme expressed in the hypothalamus, has high sequence similarity to liver/muscle CPT1a/b and binds malonyl-CoA, but does not catalyze the prototypical reaction. This suggests that CPT1c has a unique function or activation mechanism. CPT1c knockout (KO) mice have lower food intake, weigh less and have less body fat, consistent with the role as an energy-sensing malonyl-CoA target. Paradoxically, CPT1c protects against the effects of a high-fat diet. CPT1cKO mice exhibit decreased rates of fatty acid oxidation, consistent with their increased susceptibility to diet-induced obesity. We suggest that CPT1c may be a downstream target of malonyl-CoA that regulates energy homeostasis.


Subject(s)
Eating/physiology , Energy Metabolism/physiology , Hypothalamus/enzymology , Malonyl Coenzyme A/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Carboxy-Lyases/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Fatty Acid Synthases/metabolism , Hypothalamus/physiology , Malonyl Coenzyme A/physiology , Mice
3.
Biochem Soc Trans ; 33(Pt 5): 1063-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16246046

ABSTRACT

The brain plays an important role in the regulation of energy balance in higher animals. Global energy balance is monitored by sets of neurons in the hypothalamus that respond to peripheral hormonal and afferent neural signals that sense the energy status. Malonyl-CoA, an intermediate in the biosynthesis of fatty acids, appears to function in this hypothalamic energy-sensing system. The steady-state level of malonyl-CoA is determined by its rate of synthesis catalysed by ACC (acetyl-CoA carboxylase) relative to its rate of turnover catalysed by FAS (fatty acid synthase). Changes in the level of malonyl-CoA in the hypothalamus alter the expression/secretion of key hypothalamic orexigenic and anorexigenic neuropeptides that regulate the feeding behaviour and energy expenditure. Inhibitors of FAS, administered i.c.v. (intracerebroventricularly) to lean or obese mice, cause a rapid rise in hypothalamic malonyl-CoA level, suppression of food intake, increased fatty acid oxidation in skeletal muscle and profound weight loss. Stereotactic delivery of a viral MCD (malonyl-CoA decarboxylase) expression vector into the ventral hypothalamus lowers malonyl-CoA levels and reverses the anorectic effect of the FAS inhibitors. Fasting decreases, whereas refeeding increases, hypothalamic malonyl-CoA and alters subsequent feeding behaviour accordingly. The level of malonyl-CoA in the hypothalamus appears to be under the control of 5'-AMP kinase, which phosphorylates and thereby inactivates ACC under conditions of energy surplus. Thus malonyl-CoA appears to link the energy-responsive fatty acid synthesis in the hypothalamus to feeding behaviour and peripheral energy expenditure.


Subject(s)
Energy Intake , Energy Metabolism , Feeding Behavior , Hypothalamus/physiology , Malonyl Coenzyme A/physiology , Animals , Mice , Models, Animal , Neurons/physiology
4.
Horm Metab Res ; 34(11-12): 607-15, 2002.
Article in English | MEDLINE | ID: mdl-12660869

ABSTRACT

Control of energy homeostasis requires communication between the brain and adipose tissue. The sympathetic nervous system plays an integral role in relaying information during this process. Recent investigations indicate that the contributions of the sympathetic nervous system to the regulation of adipose tissue are greater than initially appreciated. A recently developed co-culture system provides evidence that a local feedback loop may exist between sympathetic neurons and adipose tissue. The co-culture approach may prove useful in further investigations of the interaction between sympathetic neurons and adipocytes, and might be adapted to study interactions between other types of neurons and adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/innervation , Energy Metabolism/physiology , Homeostasis/physiology , Neurons/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Communication , Cells, Cultured , Coculture Techniques , Cricetinae , Feedback , Mice , Rats , Signal Transduction/physiology , Sympathetic Nervous System/physiology
5.
Biochem Biophys Res Commun ; 288(4): 752-6, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11688971

ABSTRACT

Activation of transcription of the C/EBPalpha (CCAAT/enhancer binding protein alpha) gene is a critical event in the differentiation of 3T3-L1 preadipocytes into adipocytes. The kinetics of this process parallels a decline of AP-2alpha protein (also referred to as CUP, C/EBP undifferentiated protein) and decreased binding of CUP/AP-2alpha to the C/EBPalpha promoter. Mutation of the CUP/AP-2 binding sites in the C/EBPalpha promoter results in increased C/EBPalpha expression. Based on these findings, it appears that decline in AP-2alpha expression is an important early event in the adipocyte differentiation program. In the studies presented here, we identify three mRNAs that encode the repressive CUP/AP-2alpha isoforms expressed in undifferentiated 3T3-L1 preadipocytes. We demonstrate that the kinetics of the decline of these isoforms' expression over the course of differentiation parallels both the decrease in CUP/AP-2alpha DNA binding activity and the increase in C/EBPalpha protein observed in previous studies.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation , DNA-Binding Proteins/genetics , Down-Regulation , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Line , Mice , Mice, Inbred C57BL , Nuclease Protection Assays , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Stem Cells/cytology , Transcription Factor AP-2
6.
Proc Natl Acad Sci U S A ; 98(22): 12385-90, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11606782

ABSTRACT

White adipose tissue plays an integral role in energy metabolism and is governed by endocrine, autocrine, and neural signals. Neural control of adipose metabolism is mediated by sympathetic neurons that innervate the tissue. To investigate the effects of this innervation, an ex vivo system was developed in which 3T3-L1 adipocytes are cocultured with sympathetic neurons isolated from the superior cervical ganglia of newborn rats. In coculture, both adipocytes and neurons exhibit appropriate morphology, express cell-type-specific markers, and modulate key metabolic processes in one another. Lipolysis (stimulated by beta-adrenergic agents) and leptin secretion by adipocytes are down-regulated by neurons in coculture, effects apparently mediated by neuropeptide Y (NPY). Secretion of NPY by neurons is up-regulated dramatically by the presence of adipocytes in coculture and appears to be mediated by an adipocyte-derived soluble factor. Insulin, an antilipolytic agent, down-regulates NPY secretion. Our findings suggest that an adipocyte-derived factor(s) up-regulates the secretion of NPY by sympathetic neurons, which, in turn, attenuates lipolytic energy mobilization by adipocytes.


Subject(s)
Adipocytes/physiology , Cell Communication , Ganglia, Sympathetic/physiology , 3T3 Cells , Animals , Coculture Techniques , Female , Insulin/pharmacology , Lipolysis , Mice , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
7.
J Biol Chem ; 275(52): 41325-32, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11018036

ABSTRACT

Many human immunodeficiency virus (HIV)-infected patients taking combination antiretroviral therapy that includes HIV protease inhibitors experience atrophy of peripheral subcutaneous adipose tissue. We investigated the effects of HIV protease inhibitors on adipogenesis and adipocyte survival using the 3T3-L1 preadipocyte cell line. Several HIV protease inhibitors were found either to inhibit preadipocyte differentiation or to promote adipocyte cell death. One protease inhibitor, nelfinavir, elicited both of these effects strongly. When induced to differentiate in the presence of nelfinavir, 3T3-L1 preadipocytes failed to accumulate cytoplasmic triacylglycerol and failed to express normal levels of the adipogenic transcription factors CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. The level of the proteolytically processed, active 68-kDa form of sterol regulatory element-binding protein-1, a transcription factor known to promote lipogenic gene expression, also was reduced markedly in nelfinavir-treated cells, whereas the level of the 125-kDa precursor form of this protein was unaffected. The inhibitory effect of nelfinavir occurred subsequent to critical early events in preadipocyte differentiation, expression of CCAAT/enhancer-binding protein beta and completion of the mitotic clonal expansion phase, because these events were unaffected by nelfinavir treatment. In addition, nelfinavir treatment of fully differentiated 3T3-L1 adipocytes resulted in DNA strand cleavage and severe loss of cell viability. In contrast, cell proliferation and viability of preadipocytes were unaffected by nelfinavir treatment. Thus, molecular or cellular changes that occur during acquisition of the adipocyte phenotype promote susceptibility to nelfinavir-induced cell death. When considered together, these results suggest that nelfinavir may promote adipose tissue atrophy by compromising adipocyte viability and preventing replacement of lost adipocytes by inhibiting preadipocyte differentiation.


Subject(s)
Adipocytes/drug effects , HIV Protease Inhibitors/pharmacology , Stem Cells/drug effects , Transcription Factors , 3T3 Cells , Adipocytes/physiology , Animals , CCAAT-Enhancer-Binding Proteins/biosynthesis , Cell Differentiation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/biosynthesis , In Situ Nick-End Labeling , Mice , Mitosis/drug effects , Nelfinavir/pharmacology , Stem Cells/physiology , Sterol Regulatory Element Binding Protein 1 , Triglycerides/metabolism
8.
Proc Natl Acad Sci U S A ; 97(23): 12446-50, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11050169

ABSTRACT

Hormone induction of growth-arrested preadipocytes triggers mitotic clonal expansion followed by expression of CCAAT/enhancer-binding protein (C/EBP)alpha and differentiation into adipocytes. The order of these events is critical because C/EBPalpha is antimitotic and its expression prematurely would block the mitotic clonal expansion required for differentiation. C/EBPbeta, a transcriptional activator of the C/EBPalpha gene, is expressed early in the differentiation program, but lacks DNA-binding activity and fails to localize to centromeres until preadipocytes traverse the G(1)-S checkpoint of mitotic clonal expansion. Evidence is presented that dominant-negative CHOP-10 expressed by growth-arrested preadipocytes transiently sequesters C/EBPbeta by heterodimerization. As preadipocytes reach S phase, CHOP-10 is down-regulated, apparently releasing C/EBPbeta from inhibitory constraint and allowing transactivation of the C/EBPalpha gene. In support of these findings, up-regulation of CHOP-10 with the protease inhibitor N-acetyl-Leu-Leu-norleucinal prevents activation of C/EBPbeta, expression of C/EBPalpha, and adipogenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Down-Regulation , Trans-Activators/metabolism , Transcription Factors/physiology , Up-Regulation , 3T3 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Centromere/metabolism , DNA/metabolism , Mice , Phenotype , Transcription Factor CHOP , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Proc Natl Acad Sci U S A ; 97(23): 12519-23, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11050170

ABSTRACT

CCAAT enhancer-binding protein-alpha (C/EBPalpha) functions as a pleiotropic transcriptional activator of adipocyte genes during adipogenesis. Nuclear factor C/EBP undifferentiated protein (CUP), an isoform of activator protein-2alpha (AP-2alpha), binds to repressive elements in the C/EBPalpha gene promoter, silencing the gene until late in the differentiation program. The CUP regulatory element overlaps a Sp (GT-box) element in the promoter to which Sp3 (or Sp1) can bind. Binding by Sp3 or Sp1 and CUP/AP2-alpha is mutually exclusive. Sp3 is a strong transcriptional activator of the C/EBPalpha gene promoter in 3T3-L1 preadipocytes and Schneider cells, this activation being repressed by CUP/AP-2alpha. Sp3 is expressed throughout differentiation, whereas CUP/AP-2alpha, which is expressed only by preadipocytes, is down-regulated during differentiation coincident with transcription of the C/EBPalpha gene. Thus, CUP/AP-2alpha delays access of Sp3 to the Sp regulatory element, preventing premature expression of C/EBPalpha and thereby interference by C/EBPalpha (which is antimitotic) with mitotic clonal expansion, an essential early event in the differentiation program.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes , Animals , Binding Sites , Cell Differentiation , Cell Line , DNA-Binding Proteins/genetics , Drosophila Proteins , Drosophila melanogaster , Electrophoresis, Polyacrylamide Gel/methods , Genes, Overlapping , Humans , Kinetics , Mice , Nuclear Proteins/genetics , Oligonucleotides , Sp3 Transcription Factor , Transcription Factor AP-2 , Transcription Factors/genetics , Transcriptional Activation
10.
J Biol Chem ; 275(44): 34344-52, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10926934

ABSTRACT

Differentiation of 3T3-L1 preadipocytes into adipocytes is induced by a combination of inducers, including a glucocorticoid, an agent that elevates cellular cAMP, and a ligand of the insulin-like growth factor-1 receptor. Previous studies have implicated protein-tyrosine phosphatase (PTPase) HA2, a homologue of PTPase 1B, in the signaling cascade initiated by the differentiation inducers. Vanadate, a potent PTPase inhibitor, blocks adipocyte differentiation at an early stage in the program, but has no effect on the mitotic clonal expansion required for differentiation. Exposure of preadipocytes to vanadate along with the inducing agents led to the accumulation of pp35, a phosphotyrosyl protein that is a substrate for PTPase HA2. pp35 was purified to homogeneity and shown by amino acid sequence and mass analyses of tryptic peptides to be c-Crk, a known cytoplasmic target of the insulin-like growth factor-1 receptor tyrosine kinase. Transfection of 3T3-L1 preadipocytes with a c-Crk antisense RNA expression vector markedly reduced c-Crk levels and prevented differentiation into adipocytes. Studies with C3G, a protein that binds to the SH3 domain in c-Crk, showed that phosphorylation of c-Crk rendered the SH3 domain inaccessible to C3G. Taken together, these findings indicate that locking c-Crk in the phosphorylated state with vanadate prevents its participation in the signaling system that initiates adipocyte differentiation.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/physiology , Proto-Oncogene Proteins/physiology , Receptor, IGF Type 1/metabolism , Signal Transduction , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Mice , Molecular Sequence Data , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/pharmacology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-crk , RNA, Antisense/genetics , Substrate Specificity , Tyrosine/metabolism , Vanadates/pharmacology , src Homology Domains
11.
Science ; 288(5475): 2379-81, 2000 Jun 30.
Article in English | MEDLINE | ID: mdl-10875926

ABSTRACT

With the escalation of obesity-related disease, there is great interest in defining the mechanisms that control appetite and body weight. We have identified a link between anabolic energy metabolism and appetite control. Both systemic and intracerebroventricular treatment of mice with fatty acid synthase (FAS) inhibitors (cerulenin and a synthetic compound C75) led to inhibition of feeding and dramatic weight loss. C75 inhibited expression of the prophagic signal neuropeptide Y in the hypothalamus and acted in a leptin-independent manner that appears to be mediated by malonyl-coenzyme A. Thus, FAS may represent an important link in feeding regulation and may be a potential therapeutic target.


Subject(s)
Appetite Depressants/pharmacology , Appetite/drug effects , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Weight Loss/drug effects , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/chemical synthesis , Cerulenin/pharmacology , Dose-Response Relationship, Drug , Eating/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemical synthesis , Fasting , Fatty Acid Synthases/metabolism , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Leptin/metabolism , Liver/drug effects , Liver/metabolism , Male , Malonyl Coenzyme A/metabolism , Mice , Mice, Inbred BALB C , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/administration & dosage , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
J Biol Chem ; 275(23): 17653-60, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10749891

ABSTRACT

Evidence is presented that calpain, a calcium-activated protease, degrades the cyclin-dependent kinase inhibitor, p27, during the mitotic clonal expansion phase of 3T3-L1 preadipocyte differentiation. Calpain activity is required during an early stage of the adipocyte differentiation program. Thus, inhibition of calpain with N-acetyl-Leu-Leu-norleucinal (ALLN) blocks clonal expansion and acquisition of the adipocyte phenotype only when added between 12 and 24 h after the induction of differentiation. Likewise, inhibition of calpain by overexpression of calpastatin, the specific endogenous inhibitor of calpain, prevents 2-day post-confluent preadipocytes from reentering the cell cycle triggered by the differentiation inducers. Inhibition of calpain with ALLN causes preadipocytes to arrest just prior to S phase and prevents phosphorylation of the retinoblastoma gene product, DNA replication, clonal expansion, and subsequent adipocyte differentiation but does not affect the expression of immediate early genes (i.e. fos, jun, C/EBPbeta, and C/EBPdelta). Inhibition of calpain by either ALLN or by overexpression of calpastatin blocks the degradation of p27. p27 is degraded in vitro by cell-free extracts from clonally expanding preadipocytes that contain "active" calpain but not by extracts from pre-mitotic preadipocytes that do not. This action is inhibited by calpastatin or ALLN. Likewise, p27 in preadipocyte extracts is a substrate for purified calpain; this proteolytic action was inhibited by heat inactivation, EGTA, or ALLN. Thus, extracellular signals from the differentiation inducers appear to activate calpain, which degrades p27 allowing density-dependent inhibited preadipocytes to reenter the cell cycle and undergo mitotic clonal expansion.


Subject(s)
Adipocytes/cytology , Adipocytes/physiology , Calpain/metabolism , Cell Cycle Proteins , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , 3T3 Cells , Animals , Cell Differentiation , Clone Cells , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/metabolism , Gene Expression Regulation/drug effects , Genes, Immediate-Early , Kinetics , Leupeptins/pharmacology , Mice , Mitosis , Recombinant Proteins/metabolism , Time Factors , Transfection , Triglycerides/metabolism
13.
Biochem Biophys Res Commun ; 266(3): 677-83, 1999 Dec 29.
Article in English | MEDLINE | ID: mdl-10603305

ABSTRACT

Members of the C/EBP family of transcription factors play essential roles in the adipocyte differentiation program. Treatment of growth-arrested 3T3-L1 preadipocytes with appropriate hormonal agents causes the cells to synchronously reenter the cell cycle and to undergo mitotic clonal expansion. Expression of C/EBPbeta and delta occur early in clonal expansion, later followed by C/EBPalpha (which is anti-mitotic) as the cells exit the cell cycle begin to express adipocyte genes. C/EBPalpha serves as transcriptional activator of many adipocyte genes whose expression produce the adipocyte phenotype. Recent work in this laboratory has focussed on the roles of C/EBPbeta and delta in the differentiation program, in particular the mechanisms by which they activate transcription of the C/EBPalpha gene. Several regulatory elements, both repressive and activating, in proximal promoter of the gene have been identified. The cognate transacting factors that interact with these elements have been characterized and their functions elucidated. These factors have been incorporated into a model for a cascade that leads to transcriptional activation of the C/EBPalpha gene and the terminal steps in the differentiation program.


Subject(s)
Adipocytes/cytology , Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Centromere , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Trans-Activators/metabolism
14.
Genes Dev ; 13(17): 2231-41, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10485846

ABSTRACT

Hormonal induction of 3T3-L1 preadipocytes triggers a cascade of events that initiate differentiation into adipocytes. CCAAT/enhancer-binding proteins beta and delta (C/EBPbeta/delta) are expressed early in the differentiation program, but are not immediately active. After a long lag, C/EBPbeta/delta become competent to bind to the C/EBP regulatory element in the C/EBPalpha gene promoter, C/EBPalpha being a transcriptional activator of numerous adipocyte genes. As C/EBPbeta/delta acquire binding activity, they become localized to centromeres as preadipocytes synchronously enter S phase at the onset of mitotic clonal expansion. Localization to centromeres occurs through C/EBP consensus-binding sites in centromeric satellite DNA. C/EBPalpha, which is antimitotic, becomes centromere-associated much later in the differentiation program as mitotic clonal expansion ceases and the cells become terminally differentiated.


Subject(s)
Adipocytes/cytology , Adipocytes/physiology , Cell Cycle/physiology , Centromere/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , 3T3 Cells , Animals , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , Cell Nucleus/physiology , DNA-Binding Proteins/analysis , Enhancer Elements, Genetic , Gene Expression Regulation , Kinetics , Mice , Mitosis , Molecular Sequence Data , Nuclear Proteins/analysis , Regulatory Sequences, Nucleic Acid , S Phase , Transcription Factors
15.
Biochem Biophys Res Commun ; 260(3): 600-4, 1999 Jul 14.
Article in English | MEDLINE | ID: mdl-10403812

ABSTRACT

Prolonged treatment of 3T3-L1 adipocytes with 8-Br-cAMP decreases expression of GLUT4, the insulin-responsive glucose transporter. Expression of a promoter-reporter gene construct that contained 785 base pairs of 5'-flanking region of the murine GLUT4 gene was down regulated by 8-Br-cAMP (p < 0.001), whereas expression of constructs that contained 641 or 469 base pairs of 5'-flanking region was not. A reporter gene construct in which bases -706 to -676 were deleted was not repressed by 8-Br-cAMP, thereby identifying a 30 bp region as necessary for repression of the GLUT4 promoter by 8-Br-cAMP. Mutations in this regulatory element that disrupt binding of the transcription factor NF1 abolish the 8-Br-cAMP-induced repression of the gene. Although insulin and cAMP both repress the GLUT4 promoter through this cis-element, they appear to do this through different mechanisms, as treatment with 8-Br-cAMP does not induce the phosphorylation of NF1 that is induced by insulin treatment.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Monosaccharide Transport Proteins/genetics , Muscle Proteins , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Base Sequence , Colforsin/pharmacology , Genes, Reporter/genetics , Glucose Transporter Type 4 , Insulin/pharmacology , Mice , Mutation , NFI Transcription Factors , Nuclear Proteins/metabolism , Oligonucleotides/genetics , Oligonucleotides/metabolism , Phosphorylation/drug effects , RNA, Messenger/analysis , RNA, Messenger/genetics , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Cell Biol ; 19(7): 4855-65, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373535

ABSTRACT

Expression of C/EBPalpha is required for differentiation of 3T3-L1 preadipocytes into adipocytes. Previous investigations indicated that transcription of the C/EBPalpha gene is sequentially activated during differentiation, initially by C/EBPbeta and C/EBPdelta and later by C/EBPalpha (autoactivation). These events are mediated by a C/EBP regulatory element in the promoter of the C/EBPalpha gene. This article presents evidence that members of the Sp family, notably Sp1, act repressively on the C/EBPalpha promoter prior to the induction of differentiation. Sp1 was shown to bind to a GC box at the 5' end of the C/EBP regulatory element in the C/EBPalpha promoter and, in so doing, to competitively prevent binding to and transactivation of the promoter by the C/EBPs. One of the differentiation inducers methylisobutylxanthine (a cAMP phosphodiesterase inhibitor) or Forskolin, both of which increase the cellular cAMP level, causes down-regulation of Sp1. This decrease in Sp1 level early in the differentiation program appears to facilitate access of C/EBPbeta and/or C/EBPdelta to the C/EBP regulatory element and, thereby, derepression of the C/EBPalpha gene.


Subject(s)
Adipocytes/cytology , DNA-Binding Proteins/genetics , Gene Expression Regulation , Nuclear Proteins/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , 3T3 Cells , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA Footprinting , DNA-Binding Proteins/metabolism , Deoxyribonuclease I , Genes, Overlapping , Mice , Nuclear Proteins/metabolism , Oligonucleotides , Phosphorylation , Transcriptional Activation
17.
Biochem Biophys Res Commun ; 258(1): 187-93, 1999 Apr 29.
Article in English | MEDLINE | ID: mdl-10222258

ABSTRACT

Mitochondrial uncoupling protein 3 (UCP3) is expressed in skeletal muscles. We have hypothesized that increased glucose flux in skeletal muscles may lead to increased UCP3 expression. Male transgenic mice harboring insulin-responsive glucose transporter (GLUT4) minigenes with differing lengths of 5'-flanking sequence (-3237, -2000, -1000 and -442 bp) express different levels of GLUT4 protein in various skeletal muscles. Expression of the GLUT4 transgenes caused an increase in UCP3 mRNA that paralleled the increase of GLUT4 protein in gastrocnemius muscle. The effects of increased intracellular GLUT4 level on the expression of UCP1, UCP2 and UCP3 were compared in several tissues of male 4 month-old mice harboring the -1000 GLUT4 minigene transgene. In the -1000 GLUT4 transgenic mice, expression of GLUT4 mRNA and protein in skeletal muscles, brown adipose tissue (BAT), and white adipose tissue (WAT) was increased by 1.4 to 4.0-fold. Compared with non-transgenic littermates, the -1000 GLUT4 mice exhibited about 4- and 1.8-fold increases of UCP3 mRNA in skeletal muscle and WAT, respectively, and a 38% decrease of UCP1 mRNA in BAT. The transgenic mice had a 16% increase in oxygen consumption and a 14% decrease in blood glucose and a 68% increase in blood lactate, but no change in FFA or beta-OHB levels. T3 and leptin concentrations were decreased in transgenic mice. Expression of UCP1 in BAT of the -442 GLUT4 mice, which did not overexpress GLUT4 in this tissue, was not altered. These findings indicate that overexpression of GLUT4 up-regulates UCP3 expression in skeletal muscle and down-regulates UCP1 expression in BAT, possibly by increasing the rate of glucose uptake into these tissues.


Subject(s)
Carrier Proteins/genetics , Monosaccharide Transport Proteins/genetics , Muscle Proteins , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , Up-Regulation , Adipose Tissue, Brown/metabolism , Animals , Glucagon/blood , Glucose/metabolism , Glucose Transporter Type 4 , Insulin/blood , Insulin/metabolism , Ion Channels , Male , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins , Uncoupling Protein 3
18.
Biochem Biophys Res Commun ; 258(2): 464-9, 1999 May 10.
Article in English | MEDLINE | ID: mdl-10329410

ABSTRACT

Uncoupling proteins (UCPs) are mitochondrial membrane proton transporters that uncouple oxidative phosphorylation by dissipating the proton gradient across the membrane. We have investigated regulation of the UCP3 gene in skeletal muscle and C2C12 muscle cells. UCP3 mRNA in mouse skeletal muscle is markedly increased by fasting and rapidly (within 4 h) decreased by re-feeding. Methyl palmoxirate, which inhibits fatty acid uptake by mitochondria and increases blood free fatty acids, prevents the fall in UCP3 message level induced by re-feeding. These findings suggest that fatty acid or a metabolite thereof, activates the UCP3 gene. Proof that fatty acid per se up-regulates UCP3 mRNA was obtained with C2C12 muscle cells in culture. Thus, oleic acid activated expression of UCP3 mRNA in differentiated C2C12 myotubes in a time and concentration-dependent manner. Moreover, BRL49653, a ligand for the nuclear hormone receptor PPARgamma induces expression of UCP3 mRNA suggesting that PPARgamma may regulate transcription of the UCP3 gene.


Subject(s)
Carrier Proteins/genetics , Fatty Acids/pharmacology , Mitochondria, Muscle/metabolism , Muscle, Skeletal/drug effects , Thiazolidinediones , Up-Regulation , Animals , Cell Line , Ion Channels , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rosiglitazone , Thiazoles/pharmacology , Uncoupling Protein 3
19.
J Biol Chem ; 274(18): 12917-24, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10212282

ABSTRACT

Insulin represses GLUT4 expression in 3T3-L1 adipocytes through an insulin response element located at bases -706 to -676 in the 5'-flanking sequence. Nuclear proteins related to the nuclear factor I (NF1) family of transcription factors bind to this insulin response element. Mutations that disrupt binding of NF1 proteins to the insulin response element impair the insulin response in reporter gene assays. Insulin treatment of 3T3-L1 adipocytes induces a rapid change in the level of phosphorylation of NF1 proteins, providing a potential mechanism for insulin's ability to regulate gene expression through NF1. Another as yet unidentified protein, not related to NF1, also binds to the GLUT4 insulin response element and is able to mediate partial repression of the GLUT4 promoter in reporter gene assays.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , Insulin/pharmacology , Monosaccharide Transport Proteins/genetics , Muscle Proteins , Promoter Regions, Genetic , Repressor Proteins/metabolism , Transcription Factors , 3T3 Cells , Animals , Base Sequence , DNA Probes , Glucose Transporter Type 4 , Mice , Molecular Sequence Data , NFI Transcription Factors , Nuclear Proteins , Phosphorylation , Y-Box-Binding Protein 1
20.
Proc Natl Acad Sci U S A ; 96(4): 1279-84, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-9990015

ABSTRACT

Evidence is presented that the calcium-activated protease, calpain, is required for differentiation of 3T3-L1 preadipocytes into adipocytes induced by methylisobutylxanthine (a cAMP phosphodiesterase inhibitor), dexamethasone, and insulin. Calpain is expressed by preadipocytes and its level falls during differentiation. Exposure of preadipocytes to the calpain inhibitor N-acetyl-Leu-Leu-norleucinal or overexpression of calpastatin, a specific endogenous inhibitor of calpain, blocks expression of adipocyte-specific genes, notably the CCAAT/enhancer-binding protein (C/EBP)alpha gene, and acquisition of the adipocyte phenotype. The inhibitor disrupts the differentiation-inducing effect of methylisobutylxanthine (by means of the cAMP-signaling pathway), but is without effect on differentiation induced by dexamethasone or insulin. N-acetyl-Leu-Leu-norleucinal, or overexpression of calpastatin, inhibits reporter gene expression mediated by the C/EBPalpha gene promoter by preventing C/EBPbeta, a transcriptional activator of the C/EBPalpha gene, from binding to the promoter. These findings implicate calpain in the transcriptional activation of the C/EBPalpha gene, a process required for terminal adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Calpain/metabolism , Leupeptins/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3 Cells , Adipocytes/drug effects , Animals , CCAAT-Enhancer-Binding Proteins , Calcium-Binding Proteins/metabolism , Calpain/antagonists & inhibitors , Cell Differentiation/drug effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Insulin/pharmacology , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...