Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Science ; 366(6464): 435, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31649192

Subject(s)
Lions , Sports , Animals , Biodiversity
2.
Diabetes Obes Metab ; 12(3): 195-203, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19895635

ABSTRACT

AIM: The insulin sensitizer rosiglitazone (RTZ) acts by activating peroxisome proliferator and activated receptor gamma (PPAR gamma), an effect accompanied in vivo in humans by an increase in fat storage. We hypothesized that this effect concerns PPARgamma(1) and PPARgamma(2) differently and is dependant on the origin of the adipose cells (subcutaneous or visceral). To this aim, the effect of RTZ, the PPARgamma antagonist GW9662 and lentiviral vectors expressing interfering RNA were evaluated on human pre-adipocyte models. METHODS: Two models were investigated: the human pre-adipose cell line Chub-S7 and primary pre-adipocytes derived from subcutaneous and visceral biopsies of adipose tissue (AT) obtained from obese patients. Cells were used to perform oil-red O staining, gene expression measurements and lentiviral infections. RESULTS: In both models, RTZ was found to stimulate the differentiation of pre-adipocytes into mature cells. This was accompanied by significant increases in both the PPARgamma(1) and PPARgamma(2) gene expression, with a relatively stronger stimulation of PPARgamma(2). In contrast, RTZ failed to stimulate differentiation processes when cells were incubated in the presence of GW9662. This effect was similar to the effect observed using interfering RNA against PPARgamma(2). It was accompanied by an abrogation of the RTZ-induced PPARgamma(2) gene expression, whereas the level of PPARgamma(1) was not affected. CONCLUSIONS: Both the GW9662 treatment and interfering RNA against PPARgamma(2) are able to abrogate RTZ-induced differentiation without a significant change of PPARgamma(1) gene expression. These results are consistent with previous results obtained in animal models and suggest that in humans PPARgamma(2) may also be the key isoform involved in fat storage.


Subject(s)
Adipocytes/drug effects , Anilides/pharmacology , Cell Differentiation/drug effects , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Thiazolidinediones/pharmacology , Adipocytes/cytology , Adult , Cell Line , Cells, Cultured , Female , Gene Expression/drug effects , Humans , Obesity/metabolism , PPAR gamma/genetics , RNA, Messenger/metabolism , Rosiglitazone
3.
J Endocrinol ; 197(2): 297-307, 2008 May.
Article in English | MEDLINE | ID: mdl-18434359

ABSTRACT

Glucocorticoid excess increases fat mass, preferentially within omental depots; yet circulating cortisol concentrations are normal in most patients with metabolic syndrome (MS). At a pre-receptor level, 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) activates cortisol from cortisone locally within adipose tissue, and inhibition of 11beta-HSD1 in liver and adipose tissue has been proposed as a novel therapy to treat MS by reducing hepatic glucose output and adiposity. Using a transformed human subcutaneous preadipocyte cell line (Chub-S7) and human primary preadipocytes, we have defined the role of glucocorticoids and 11beta-HSD1 in regulating adipose tissue differentiation. Human cells were differentiated with 1.0 microM cortisol (F), or cortisone (E) with or without 100 nM of a highly selective 11beta-HSD1 inhibitor PF-877423. 11beta-HSD1 mRNA expression increased across adipocyte differentiation (P<0.001, n=4), which was paralleled by an increase in 11beta-HSD1 oxo-reductase activity (from nil on day 0 to 5.9+/-1.9 pmol/mg per h on day 16, P<0.01, n=7). Cortisone enhanced adipocyte differentiation; fatty acid-binding protein 4 expression increased 312-fold (P<0.001) and glycerol-3-phosphate dehydrogenase 47-fold (P<0.001) versus controls. This was abolished by co-incubation with PF-877423. In addition, cellular lipid content decreased significantly. These findings were confirmed in the primary cultures of human subcutaneous preadipocytes. The increase in 11beta-HSD1 mRNA expression and activity is essential for the induction of human adipogenesis. Blocking adipogenesis with a novel and specific 11beta-HSD1 inhibitor may represent a novel approach to treat obesity in patients with MS.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/physiology , Adipogenesis/drug effects , Enzyme Inhibitors/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Cell Differentiation , Cells, Cultured , Fatty Acid-Binding Proteins/genetics , Glucocorticoids/metabolism , Humans , Metabolic Syndrome/etiology , PPAR gamma/genetics , RNA, Messenger/analysis
4.
Int J Obes (Lond) ; 32(4): 648-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18087263

ABSTRACT

OBJECTIVE: To evaluate the ontogeny of adipose tissue dynamics in obese and lean Zucker rat pups, from suckling to puberty. METHODS: The trial had a two-group parallel design. Sixty-two male Zucker rat pups shared within 15 litters received deuterated water for 5 days, prior killing at different age. Adipose tissues were collected for (2)H-enrichment analyses using mass spectrometry to determine fat cell proliferation and lipid synthesis rates. Rats were assigned to obese and lean rat groups by genotyping. RESULTS: The time course (from days 13 to 55) of all adipose tissue growth showed that the highest fractional rates of fat cell proliferation, triacylglycerol (TG) synthesis and de novo lipogenesis (DNL) took place during early suckling in all rat pups. The appearance of excessive fat mass growth in the obese rats, as compared with lean rats, was first shown through a significant increase in DNL at the end of suckling (P<0.05). The TG synthesis rate was enhanced (P<0.05) from the end of suckling and early postweaning until day 55 (from 122+/-10 to 498+/-78 in obese pups and from 25+/-6 to 75+/-26 mg new TG per day in lean pups (median+/-s.e.m., P<0.01)). In contrast, only by day 55 did the fractional proliferation rate of fat cells in retroperitoneal and epididymal depots in the obese rats supersede that of the lean rats (P<0.05). CONCLUSION: The early suckling period constitutes the most active period for adipose tissue development in normal rats. In the obese Zucker rat model, adipose hypertrophy primarily contributes to the early onset of obesity, while hyperplasia increases after puberty. Early onset of adipose tissue growth may play a determinant role in the development of obesity later in life.


Subject(s)
Adipogenesis , Obesity/physiopathology , Thinness/physiopathology , Adipose Tissue/pathology , Animals , Biomarkers/blood , Body Weight , Cell Proliferation , Lactation/physiology , Lipogenesis , Liver/pathology , Male , Obesity/metabolism , Obesity/pathology , Organ Size , Rats , Rats, Zucker , Thinness/metabolism , Thinness/pathology , Triglycerides/biosynthesis
5.
Int J Obes (Lond) ; 32(1): 112-20, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17637700

ABSTRACT

OBJECTIVE: The objective of this study was to determine the contribution of mesothelial cells, present in human omental adipose tissue (OAT) but not in the subcutaneous depot (SAT), on the expression of inflammation-related factors. DESIGN: Comparison of the expression profiles of inflammation-related genes in mesothelial cells with those in the adipocyte-enriched (AEF) and stromal vascular fractions (SVF) and localization of interleukin-18 (IL-18) expression in adipose depots. SUBJECTS: Eleven obese Caucasian female subjects undergoing gastric bypass surgery (body mass index: 43.6+/-1.3 kg/m(2); age: 41.6+/-2.3 years). MEASUREMENTS: The expression profiles of cytokine and chemokine-related genes in mesothelial cells and in cell fractions prepared from OAT were assessed by the microarray technique. The differential expression of IL-18 was confirmed by real-time PCR and the protein was localized in adipose depots by immunohistochemistry. RESULTS: Microarray data analysis demonstrated that, of the 16 cytokine and chemokine-related genes that were upregulated in mesothelial cells compared with the AEF, IL-18 was the cytokine with the highest differential expression. IL-18 expression was similar in mesothelial cells and the SVF. In both SAT and OAT, IL-18 was immunolocalized in neutrophils and mast cells, but not in macrophages nor adipocytes. This cytokine was also detected in mesothelial cells in OAT. This additional source of expression may explain the higher IL-18 expression levels in OAT than SAT (+5.9-fold). CONCLUSION: By their capacity to express inflammatory-related factors, and in particular the proinflammatory cytokine IL-18 in OAT, mesothelial cells appear as a new player in the process of low-grade inflammation associated with obesity.


Subject(s)
Adipose Tissue/metabolism , Epithelial Cells/metabolism , Interleukin-18/metabolism , Obesity/metabolism , Omentum/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adipose Tissue/cytology , Adult , Epithelium/metabolism , Female , Humans , Immunohistochemistry , Inflammation/genetics , Interleukin-18/genetics , Microarray Analysis , Obesity/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Rapid Commun Mass Spectrom ; 18(7): 795-8, 2004.
Article in English | MEDLINE | ID: mdl-15052562

ABSTRACT

This paper presents a high-throughput method for the simultaneous determination of deuterium and oxygen-18 (18O) enrichment of water samples isolated from blood. This analytical method enables rapid and simple determination of these enrichments of microgram quantities of water. Water is converted into hydrogen and carbon monoxide gases by the use of a high-temperature conversion elemental analyzer (TC-EA), that are then transferred on-line into the isotope ratio mass spectrometer. Accuracy determined with the standard light Antartic precipitation (SLAP) and Greenland ice sheet precipitation (GISP) is reliable for deuterium and 18O enrichments. The range of linearity is from 0 up to 0.09 atom percent excess (APE, i.e. -78 up to 5725 delta per mil (dpm)) for deuterium enrichment and from 0 up to 0.17 APE (-11 up to 890 dpm) for 18O enrichment. Memory effects do exist but can be avoided by analyzing the biological samples in quintuplet. This method allows the determination of 1440 samples per week, i.e. 288 biological samples per week.


Subject(s)
Blood , Deuterium/blood , Hot Temperature , Mass Spectrometry/methods , Oxygen/blood , Water/chemistry , Administration, Oral , Animals , Calibration , Deuterium/administration & dosage , Deuterium/pharmacokinetics , Male , Oxygen/administration & dosage , Oxygen/pharmacokinetics , Oxygen Isotopes , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Water/administration & dosage
7.
Cell Death Differ ; 10(9): 1025-31, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12934077

ABSTRACT

Overexpression of SV40 T-antigen (SV40 T-Ag) has been widely used to overcome replicative senescence of human primary cells and to promote cell immortalization. However, in the case of certain cell types, such as preadipocytes, the differentiation process of immortalized cells is blocked by SV40 T-Ag expression. In this study, human telomerase reverse transcriptase (hTERT) and papillomavirus E7 oncoprotein (HPV-E7) genes were coexpressed in human preadipocytes to test whether this combination could maintain cell differentiation capacity after immortalization. We demonstrated that the HPV-E7/hTERT expressing preadipocytes displayed an indefinite life span. Interestingly, immortalized cells were diploid and presented no chromosomic alterations. These immortalized cells were able to accumulate and hydrolyze intracellular triglycerides and to express adipocyte markers. These data demonstrate, for the first time, that coexpression of hTERT and HPV-E7 in human preadipocytes allows cells not only to display an indefinite life span but also to retain their capacity to differentiate.


Subject(s)
Adipocytes/cytology , Cell Transformation, Viral , Oncogene Proteins, Viral/metabolism , Stem Cells/cytology , Telomerase/metabolism , Adipocytes/metabolism , Adult , Cell Differentiation , Cell Line, Transformed , Cells, Cultured , DNA-Binding Proteins , Female , Gene Expression , Humans , Karyotyping , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Stem Cells/enzymology , Stem Cells/virology
8.
Endocrinology ; 142(10): 4522-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564718

ABSTRACT

GLP-1 (glucagon-like peptide-1) is a potent insulin secretagogue released from L cells in the intestine. The regulation of GLP-1 secretion has been described both in vivo and in vitro in several animal species, but data from human cellular models are lacking. For this purpose, factors and cell-signaling pathways regulating GLP-1 secretion were investigated in the NCI-H716 human intestinal cell line. After differentiation, these cells homogeneously produced 16.8 pmol GLP-1/mg protein with a basal release of 4.2% during a 2-h incubation period. Nutrients, such as palmitic acid, oleic acid, and meat hydrolysate, stimulated GLP-1 secretion in a dose-dependent manner, as did the cholinergic agonist carbachol and the neuromediator gastrin-releasing peptide. Along with stimulating GLP-1 release, gastrin-releasing peptide, like ionomycin, increased intracellular calcium levels. Activators of PKA and PKC were able to increase GLP-1 secretion in NCI-H716 cells. However, neither PKA activators nor meat hydrolysate increased proglucagon mRNA levels. These findings indicate that the NCI-H716 cell line constitutes a unique model to study the cellular mechanism of GLP-1 secretion in humans and suggest potential interspecies divergence in the regulation of proglucagon gene expression in enteroendocrine cells.


Subject(s)
Glucagon/metabolism , Intestines/physiology , Peptide Fragments/metabolism , Protein Precursors/metabolism , Animals , Cell Line , Diet , Dose-Response Relationship, Drug , Glucagon-Like Peptide 1 , Humans , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , Protein Hydrolysates/pharmacology
9.
FASEB J ; 15(11): 2033-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11511517

ABSTRACT

The skeletal muscle mitochondrial uncoupling protein-3 (UCP3) promotes substrate oxidation, but direct evidence for its metabolic role is lacking. Here, we show that UCP3 overexpression in cultured human muscle cells decreased mitochondrial membrane potential (DYm). Despite this, the ATP content was not significantly decreased compared with control cells, whereas ADP content was reduced and thus the ATP/ADP ratio raised. This finding was contrasts with the effect caused by the chemical protonophoric uncoupler, CCCP, which lowered DYm, ATP, and the ATP/ADP ratio. UCP3-overexpression enhanced oxidation of oleate, regardless of the presence of glucose, whereas etomoxir, which blocks fatty acid entry to mitochondria, suppressed the UCP3 effect. Glucose oxidation was stimulated in UCP3-overexpressing cells, but this effect was inhibited by oleate. UCP3 caused weak increase of both 2-Deoxyglucose uptake and glycolytic rate, which differed from the marked stimulation by CCCP. We concluded that UCP3 promoted nutrient oxidation by lowering DYm and enhanced fatty acid-dependent inhibition of glucose oxidation. Unlike the uncoupler CCCP, however, UCP3 raised the ATP/ADP ratio and modestly increased glucose uptake and glycolysis. We propose that this differential effect provides a biological significance to UCP3, which is up-regulated in metabolic stress situations where it could be involved in nutrient partitioning.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Carrier Proteins/physiology , Fatty Acids/metabolism , Glucose/metabolism , Intracellular Membranes/physiology , Mitochondria, Muscle/physiology , Carrier Proteins/genetics , Cells, Cultured , Gene Expression , Glycolysis , Humans , Ion Channels , Membrane Potentials/physiology , Mitochondrial Proteins , Muscles/cytology , Muscles/metabolism , Muscles/physiology , Oxidation-Reduction , Uncoupling Agents , Uncoupling Protein 3
11.
Endocrinology ; 142(1): 487-92, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145613

ABSTRACT

White adipose tissue is known to contain the components of the renin-angiotensin system giving rise to angiotensin II (AngII). In vitro, prostacyclin is synthesized from arachidonic acid through the activity of cyclooxygenases 1 and 2 and is released from AngII-stimulated adipocytes. Prostacyclin, in turn, is able to favor adipocyte formation. Based upon in vivo and ex vivo experiments combined to immunocytochemical staining of glycerol-3-phosphate dehydrogenase (GPDH), an indicator of adipocyte formation, it is reported herein that AngII favors the appearance of GPDH-positive cells. In the presence of a cyclooxygenase inhibitor, this adipogenic effect is abolished, whereas that of (carba)prostacyclin, a stable analog of prostacyclin that bypasses this inhibition, appears unaltered. Taken together, these results are in favor of AngII acting as a trophic factor implicated locally in adipose tissue development. It is proposed that AngII enhances the formation of GPDH-expressing cells from preadipocytes in response to prostacyclin released from adipocytes.


Subject(s)
Adipocytes/cytology , Adipose Tissue/physiology , Angiotensin II/pharmacology , Glycerolphosphate Dehydrogenase/metabolism , Adipocytes/drug effects , Adipose Tissue/cytology , Adipose Tissue/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Epoprostenol/pharmacology , Glycerolphosphate Dehydrogenase/deficiency , Glycerolphosphate Dehydrogenase/genetics , Homozygote , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Rats , Rats, Wistar
12.
J Lipid Res ; 41(1): 84-92, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10627505

ABSTRACT

Intestinal fatty acid-binding protein (I-FABP) is a cytosolic protein expressed at high levels (up to 2% of cytosolic proteins) in the small intestine epithelium. Despite cell transfection studies, its function is still unclear. Indeed, different effects on fatty acid metabolism depending on the cell type and the amount of I-FABP expressed have been reported. Furthermore, a decrease in fatty acid incorporation has been unexpectedly obtained when I-FABP reached 0. 72% of cytosolic proteins in fibroblasts (Prows et al. 1997. Arch. Biochem. Biophys. 340: 135). In the present study, the effect of a high level of I-FABP similar to amounts present in the small intestine was investigated in the human colon adenocarcinoma cell line, Caco-2. After transfection with human I-FABP cDNA, a clone expressing 1.5% I-FABP and unchanged level of liver FABP was selected. These cells, which had a lower rate of proliferation as compared with mock-transfected cells, developed the typical morphological characteristics of differentiated enterocytes. Incubation of differentiated cells with [(14)C]palmitate showed a 34% reduction (P < 0.01) of fatty acid incorporation, whereas the relative distribution of radiolabel into triglycerides was not affected. A nonsignificant 21% reduction of fatty acid incorporation was observed with another clone expressing 10-fold less I-FABP. In conclusion, a high level of I-FABP expressed in a differentiated enterocyte model inhibited fatty acid incorporation, by a mechanism which remains to be defined.


Subject(s)
Carrier Proteins/genetics , Intestinal Mucosa/metabolism , Myelin P2 Protein/genetics , Neoplasm Proteins , Tumor Suppressor Proteins , Caco-2 Cells , Carrier Proteins/metabolism , Cell Differentiation , Cell Division , DNA, Complementary , Esterification , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Humans , Intestines/cytology , Intestines/ultrastructure , Microscopy, Electron , Myelin P2 Protein/metabolism , Palmitic Acid/metabolism , Transfection
13.
Am J Physiol ; 276(3): G606-12, 1999 03.
Article in English | MEDLINE | ID: mdl-10070036

ABSTRACT

Epidermal growth factor (EGF) has been reported to stimulate carbohydrate, amino acid, and electrolyte transport in the small intestine, but its effects on lipid transport are poorly documented. This study aimed to investigate EGF effects on fatty acid uptake and esterification in a human enterocyte cell line (Caco-2). EGF inhibited cell uptake of [14C]palmitate and markedly reduced its incorporation into triglycerides. In contrast, the incorporation in phospholipids was enhanced. To elucidate the mechanisms involved, key steps of lipid synthesis were investigated. The amount of intestinal fatty acid-binding protein (I-FABP), which is thought to be important for fatty acid absorption, and the activity of diacylglycerol acyltransferase (DGAT), an enzyme at the branch point of diacylglycerol utilization, were reduced. EGF effects on DGAT and on palmitate esterification occurred at 2-10 ng/ml, whereas effects on I-FABP and palmitate uptake occurred only at 10 ng/ml. This suggests that EGF inhibited palmitate uptake by reducing the I-FABP level and shifted its utilization from triglycerides to phospholipids by inhibiting DGAT. This increase in phospholipid synthesis might play a role in the restoration of enterocyte absorption function after intestinal mucosa injury.


Subject(s)
Epidermal Growth Factor/physiology , Fatty Acids/metabolism , Intestinal Mucosa/metabolism , Neoplasm Proteins , Repressor Proteins , Saccharomyces cerevisiae Proteins , Tumor Suppressor Proteins , Acyltransferases/metabolism , Caco-2 Cells , Carrier Proteins/metabolism , Coenzyme A Ligases/metabolism , Diacylglycerol O-Acyltransferase , Epidermal Growth Factor/pharmacology , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Humans , Intestines/pathology , Myelin P2 Protein/metabolism , Palmitic Acid/metabolism , Palmitic Acid/pharmacokinetics
14.
Exp Cell Res ; 244(2): 441-7, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9806794

ABSTRACT

Fatty acid-binding proteins (FABP) are small cytosolic proteins which are thought to play a key role in fatty acid metabolism. The intestine contains the intestinal (I-FABP) and the liver (L-FABP) isoforms, but their regulation is still poorly documented. In order to find suitable conditions for studying the regulation of the two FABP isoforms in Caco-2 cells, we investigated the effects of the presence of collagen during cell proliferation or differentiation. When collagen was present only during cell proliferation on culture dishes, I-FABP expression was enhanced, whereas sucrase-isomaltase was unaffected and L-FABP expression was merely accelerated. In contrast, when collagen was present during cell differentiation on filter inserts, both I-FABP and sucrase-isomaltase were strongly reduced, but L-FABP was not affected. Under the former conditions (the more suitable for studying FABP regulation), the peroxysome proliferator-activated receptor (PPAR) activators, clofibrate and alpha-bromopalmitate, enhanced the two isoforms. This study, which is the first one providing a quantitative protein analysis of I-FABP and L-FABP in Caco-2 cells, demonstrates different time courses of expression of these proteins during cell differentiation. It also shows that I-FABP is specifically regulated by collagen and that, under conditions optimal for their expression, both isoforms are modulated by metabolic factors.


Subject(s)
Carrier Proteins/metabolism , Collagen/physiology , Intestinal Mucosa/metabolism , Liver/metabolism , Myelin P2 Protein/metabolism , Neoplasm Proteins , Tumor Suppressor Proteins , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Antibodies/isolation & purification , Caco-2 Cells , Carrier Proteins/biosynthesis , Carrier Proteins/immunology , Clofibrate/pharmacology , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Humans , Intestines/drug effects , Liver/drug effects , Myelin P2 Protein/biosynthesis , Myelin P2 Protein/immunology , Palmitates/pharmacology , Protein Isoforms/biosynthesis , Protein Isoforms/immunology , Protein Isoforms/metabolism , Sucrase-Isomaltase Complex/metabolism
15.
Mol Cell Endocrinol ; 131(1): 67-77, 1997 Jul 04.
Article in English | MEDLINE | ID: mdl-9256365

ABSTRACT

Expression of mRNAs encoding the two prostaglandin endoperoxide synthase (PGHS) isoenzymes (PGHS-1 and -2) was investigated in differentiating clonal Ob1771 mouse preadipocytes and in mouse adipose tissues. Northern analysis revealed that the expression level of PGHS-1 mRNA was reduced by 98+/-0.2% (P <0.01) during differentiation of Ob1771 cells, whereas PGHS-2 mRNA was not detected. By reverse transcriptase-polymerase chain reaction analysis, however, both PGHS-1 and -2 mRNA was detected in Ob1771 preadipose cells. In addition. mRNAs encoding both isoforms were markedly expressed in primary adipose precursor cells with considerably lower expression levels in mature adipocytes (56 75% reduction, P<0.01). Furthermore, exposure to dexamethasone (10 nM) for both 24 h (explants of adipose tissue) and 48 h (Ob1771 adipose cells) resulted in enhanced expression of PGHS-1 mRNA. whereas expression of PGHS-2 mRNA in explants of adipose tissue (24 h incubation) was reduced by 83 +/- 9% (P<0.05). In contrast, exposure to angiotensin II (100 nM) enhanced expression of PGHS-1 mRNA both in mature adipocytes (4 h incubation) and explants of adipose tissue (24 h incubation), and elevated PGHS-2 mRNA expression in mature adipocytes (4 h incubation). In conclusion, this report suggests a differential expression of PGHS mRNAs during adipose cell differentiation, and further suggests that the machinery for prostaglandin synthesis in mature adipocytes may be induced by various hormones.


Subject(s)
Adipose Tissue/enzymology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Adipose Tissue/cytology , Adipose Tissue/drug effects , Angiotensin II/pharmacology , Animals , Cell Differentiation/genetics , Cell Line , Cyclooxygenase 1 , Cyclooxygenase 2 , Dexamethasone/pharmacology , Isoenzymes/drug effects , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Obese , Prostaglandin-Endoperoxide Synthases/drug effects , RNA, Messenger/biosynthesis , Stem Cells/enzymology
16.
J Cell Sci ; 110 ( Pt 11): 1279-85, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9202388

ABSTRACT

Embryonic stem cells, derived from the inner cell mass of murine blastocysts, can be maintained in a totipotent state in vitro. In appropriate conditions embryonic stem cells have been shown to differentiate in vitro into various derivatives of all three primary germ layers. We describe in this paper conditions to induce differentiation of embryonic stem cells reliably and at high efficiency into adipocytes. A prerequisite is to treat early developing embryonic stem cell-derived embryoid bodies with retinoic acid for a precise period of time. Retinoic acid could not be substituted by adipogenic hormones nor by potent activators of peroxisome proliferator-activated receptors. Treatment with retinoic acid resulted in the subsequent appearance of large clusters of mature adipocytes in embryoid body outgrowths. Lipogenic and lipolytic activities as well as high level expression of adipocyte specific genes could be detected in these cultures. Analysis of expression of potential adipogenic genes, such as peroxisome proliferator-activated receptors gamma and delta and CCAAT/enhancer binding protein beta, during differentiation of retinoic acid-treated embryoid bodies has been performed. The temporal pattern of expression of genes encoding these nuclear factors resembled that found during mouse embryogenesis. The differentiation of embryonic stem cells into adipocytes will provide an invaluable model for the characterisation of the role of genes expressed during the adipocyte development programme and for the identification of new adipogenic regulatory genes.


Subject(s)
Adipocytes/cytology , Adipose Tissue/embryology , Stem Cells/cytology , Animals , Biomarkers , CCAAT-Enhancer-Binding Proteins , Cell Differentiation/drug effects , Cells, Cultured , DNA-Binding Proteins/metabolism , Dimethyl Sulfoxide/pharmacology , Gene Expression , Mice , Nuclear Proteins/metabolism , Photomicrography , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology
17.
Endocrinology ; 138(3): 1092-6, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9048614

ABSTRACT

Ovarian hormones have been shown to modulate the metabolism of adipose cells obtained from adipose tissue of different animals. The aim of this study was to better understand the short- and long-term influences of estrogens on the in vivo lipolytic response of rat parametrial fat pads, determined by measurement of extracellular glycerol concentrations using in situ microdialysis. Possible direct effects of estrogens on lipolysis were studied by perfusion of a potent estrogenic analogue such as moxestrol. Moxestrol (10(-6) M) failed to increase glycerol concentrations in estrus, diestrus, or 8-day ovariectomized animals. However, the basal glycerol concentrations and the lipolytic responses stimulated by 10(-6) M isoproterenol were decreased in parametrial fat pads of diestrus, compared with estrus, rats. Greater decreases in basal and stimulated glycerol concentrations were observed in rats that had been ovariectomized for 8, 15, or 30 days. In ovariectomized rats, isoproterenol-induced lipolysis was restored to the levels observed in diestrus animals by a daily injection of 17 beta-estradiol for a period of 7 days. These results implicate estrogens as long-term modulators of in vivo basal and stimulated lipolytic responses of rat parametrial fat pad.


Subject(s)
Adipose Tissue/metabolism , Estrogens/metabolism , Lipolysis , Uterus , Adipose Tissue/drug effects , Animals , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Ethinyl Estradiol/analogs & derivatives , Ethinyl Estradiol/pharmacology , Female , Isoproterenol/pharmacology , Lipolysis/drug effects , Microdialysis , Ovariectomy , Rats , Rats, Wistar , Time Factors
18.
Biochem J ; 328 ( Pt 2): 701-6, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9371734

ABSTRACT

Adipose cells are an important source of angiotensinogen (AT). Its activation product, angiotensin II, stimulates in vitro and in vivo the production and release of prostacyclin which acts as a potent adipogenic signal in promoting the terminal differentiation of preadipocytes to adipocytes. Since glucocorticoids are known to promote adipose cell differentiation in vitro as well as in vivo, their role in the regulation of AT gene expression and secretion has been investigated in cultured Ob1771 mouse adipose cells. In contrast with liver cells, which are the major source of AT and the target of several hormones for the regulation of its expression, adipose cells are only responsive to glucocorticoids, which are able to up-regulate AT gene expression and AT secretion rapidly and dose-dependently. On exposure to glucocorticoids, accumulation of AT mRNA appears primarily to be due to transcriptional activation of the gene and is parallelled by secretion of the protein. Similar results on AT mRNA expression and AT secretion were obtained using explants of rat adipose tissue ex vivo demonstrating a major if not exclusive mechanism of regulation of AT production by glucocorticoids in mature adipose cells. Together these results provide a potential link between glucocorticoids, AT, the growth of adipose tissue and increased blood pressure.


Subject(s)
Adipocytes/metabolism , Angiotensinogen/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation , Glucocorticoids/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Angiotensin II/pharmacology , Angiotensinogen/genetics , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Colforsin/pharmacology , Estradiol/pharmacology , Growth Hormone/pharmacology , Hormone Antagonists/pharmacology , Male , Mice , Mifepristone/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar , Testis/cytology , Testis/metabolism
19.
Am J Physiol ; 271(4 Pt 1): E631-5, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8897849

ABSTRACT

Changes in blood flow induced by the beta-adrenoceptor agonist isoproterenol (Iso) and a stable analogue of the major metabolite of arachidonic acid in adipose tissue, carbaprostacyclin (cPGI2), have been studied in rat periepididymal fat pad with in situ microdialysis measuring the distribution ratio of 0.2% ethanol in the dialysate (outflow) to that in the perfusate (inflow) (O/I ratio). Local perfusions of 1 microM cPGI2 or 1 microM Iso led to reversible decreases of the O/I ratio that were similar to the decrease induced by the vasodilating reference drug hydralazine (Hydra, 630 microM). Interestingly, a continuous perfusion of Hydra at a submaximal vasodilating concentration (63 microM) was sufficient to prevent further vasodilatation induced by Iso or cPGI2. To take advantage of this observation, experiments were designed to evaluate the influence of the vasodilating effect of Iso or cPGI2 on the ability of either to induce lipolysis in vivo. The results showed that the vasodilating effect of Iso could contribute to glycerol removal from the extracellular fluid and demonstrate that cPGI2 was devoid of lipolytic activity.


Subject(s)
Adipose Tissue/blood supply , Adipose Tissue/metabolism , Epoprostenol/analogs & derivatives , Glycerol/metabolism , Animals , Epididymis , Epoprostenol/pharmacology , Hydralazine/pharmacology , Isoproterenol/pharmacology , Lipolysis/drug effects , Male , Microdialysis , Rats , Rats, Wistar , Regional Blood Flow , Vasodilation/drug effects , Vasodilator Agents/pharmacology
20.
Mol Cell Endocrinol ; 117(1): 17-25, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8734470

ABSTRACT

Two adenosine receptors A1 and A2 are associated with either stimulation (A2) or inhibition (A1) of adenylate cyclase. Using the clonal cell line Ob1771, we have studied the expression of the two receptors during the process of adipose conversion accelerated by exposure to dexamethasone and 3-isobutyl-l-methylxanthine (IBMX) during the first 3 days post-confluence. The effects mediated by the two receptors on preadipocyte differentiation and adipocyte metabolism were also investigated. The two adenosine agonists NECA and PIA were used as preferential agonists of the A2- and A1-receptor, respectively. In preadipose cells (just confluent), both of the mouse clonal line and human primary culture, NECA dose-dependently stimulated cAMP production with a significant higher potency (P < 0.01) than did PIA. In adipose cells (16-day post-confluent) NECA was found to exert a biphasic effect on forskolin-stimulated cAMP production: i.e., NECA was clearly inhibitory in the femto- to picomolar concentration range whereas this effect gradually diminished at higher concentrations. The effect of PIA in 16-day post-confluent adipose cells however, was purely inhibitory on both cAMP production (IC50: 33.52 +/- 0.44 fM) and lipolysis (64% +/- 7%; P < 0.01). These findings were corroborated by Northern blot analysis which revealed A1-receptor mRNA to be exclusively expressed in the mature adipocytes, whereas A2-receptor mRNA gradually declined during the differentiation process except in 16-day post-confluent cells. In addition, NECA significantly enhanced the effect of corticosterone-induced differentiation by 46.8% (P < 0.05) but failed to have any adipogenic potency acting either alone or in concert with carbaprostacyclin (cPGI2). Thus, endogenous adenosine may have a bimodal action on adipose tissue metabolism mediated through stimulatory A2- and inhibitory A1-receptors, respectively, as a function of adipose conversion.


Subject(s)
Adipose Tissue/metabolism , Receptors, Purinergic P1/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine-5'-(N-ethylcarboxamide) , Adipose Tissue/cytology , Adipose Tissue/drug effects , Animals , Blotting, Northern , Cell Differentiation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Dexamethasone/pharmacology , Humans , Lipolysis , Mice , Phenylisopropyladenosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...