Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
Endocrinology ; 142(12): 5220-5, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713218

ABSTRACT

White adipose tissue is known to contain the components of the renin-angiotensin system, which gives rise to angiotensin II from angiotensinogen (AGT). Recent evidence obtained in vitro and ex vivo is in favor of angiotensin II acting as a trophic factor of adipose tissue development. To determine whether AGT plays a role in vivo in this process, comparative studies were performed in AGT-deficient (agt(-/-)) mice and control wild-type mice. The results showed that agt(-/-) mice gain less weight than wild-type mice in response to a chow or high fat diet. Adipose tissue mass from weaning to adulthood appeared altered rather specifically, as both the size and the weight of other organs were almost unchanged. Food intake was similar for both genotypes, suggesting a decreased metabolic efficiency in agt(-/-) mice. Consistent with this hypothesis, cellularity measurement indicated hypotrophy of adipocytes in agt(-/-) mice with a parallel decrease in the fatty acid synthase activity. Moreover, AGT-deficient mice exhibited a significantly increased locomotor activity, whereas metabolic rate and mRNA levels of uncoupling proteins remained similar in both genotypes. Thus, AGT appears to be involved in the regulation of fat mass through a combination of decreased lipogenesis and increased locomotor activity that may be centrally mediated.


Subject(s)
Adipose Tissue/growth & development , Angiotensinogen/deficiency , Diet , Motor Activity/physiology , Weight Gain , Adipose Tissue/pathology , Adipose Tissue, Brown/growth & development , Adipose Tissue, Brown/pathology , Angiotensinogen/genetics , Animals , Mice , Mice, Inbred ICR , Mice, Knockout/genetics , Reference Values , Thermogenesis
2.
FASEB J ; 15(14): 2727-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11606482

ABSTRACT

White adipose tissue and liver are important angiotensinogen (AGT) production sites. Until now, plasma AGT was considered to be a reflection of hepatic production. Because plasma AGT concentration has been reported to correlate with blood pressure, and to be associated with body mass index, we investigated whether adipose AGT is released locally and into the blood stream. For this purpose, we have generated transgenic mice either in which adipose AGT is overexpressed or in which AGT expression is restricted to adipose tissue. This was achieved by the use of the aP2 adipocyte-specific promoter driving the expression of rat agt cDNA in both wild-type and hypotensive AGT-deficient mice. Our results show that in both genotypes, targeted expression of AGT in adipose tissue increases fat mass. Mice whose AGT expression is restricted to adipose tissue have AGT circulating in the blood stream, are normotensive, and exhibit restored renal function compared with AGT-deficient mice. Moreover, mice that overexpress adipose AGT have increased levels of circulating AGT, compared with wild-type mice, and are hypertensive. These animal models demonstrate that AGT produced by adipose tissue plays a role in both local adipose tissue development and in the endocrine system, which supports a role of adipose AGT in hypertensive obese patients.


Subject(s)
Adipose Tissue/growth & development , Angiotensinogen/physiology , Blood Pressure/physiology , Adipocytes/pathology , Adipose Tissue/cytology , Angiotensinogen/blood , Angiotensinogen/genetics , Animals , Drinking , Gene Expression Regulation , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renin/genetics , Renin/metabolism , Urination
5.
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
6.
Int J Obes Relat Metab Disord ; 24 Suppl 4: S33-5, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11126238

ABSTRACT

Adipose tissue is an important source of angiotensinogen (AGT). Recent evidence shows that a local renin-angiotensinogen system (RAS) is present in human adipose tissue and may act as a distinct system from plasma RAS. In obese patients, the involvement of angiotensin II (angII) as a consequence of increased plasma AGT secreted from adipose tissue has been proposed in the development of hypertension. Another role of AGT via angII in the development of adipose tissue is supported by the following: (i) in vitro, angII stimulates the production and release of prostacyclin from adipocytes, which in turn promotes the differentiation of precursor cells into adipocytes; (ii) ex vivo and in vivo, both angII and (carba)prostacyclin promote the formation of new fat cells; and (iii) AGT -/- mice exhibit a slowing down of adipose tissue development, as compared to wild-type mice. Altogether the data are consistent with an autocrine/paracrine mechanism implicating AGT, angII and prostacyclin in adipose tissue development.


Subject(s)
Adipocytes/cytology , Adipose Tissue/growth & development , Angiotensin II/genetics , Angiotensinogen/genetics , Obesity/physiopathology , Adipocytes/metabolism , Angiotensin II/metabolism , Angiotensinogen/metabolism , Epoprostenol/genetics , Epoprostenol/physiology , Gene Expression Regulation , Humans
7.
Hypertension ; 35(6): 1270-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10856276

ABSTRACT

The renin-angiotensin system has long been recognized as an important regulator of systemic blood pressure and renal electrolyte homeostasis, and local renin-angiotensin systems have also been implicated in pathological changes of organ structure and function by modulation of gene expression, growth, fibrosis, and inflammatory response. Recently, substantial data have been accumulated in support of the notion that adipose tissue, besides other endocrine functions, also hosts a local renin-angiotensin system. In the first part of this review, we describe the components of the adipose tissue renin-angiotensin system in human and rodent animal models with respect to regulation of angiotensinogen expression and secretion, formation of angiotensin peptides, and the existence of angiotensin II receptors. In the second part, we describe the role of the adipose tissue renin-angiotensin system in the process of adipogenic differentiation and in the regulation of body weight. We also detail the differential regulation of the adipose tissue renin-angiotensin system in obesity and hypertension and thereby also speculate on its possible role in the development of obesity-associated hypertension. Although some findings on the adipose tissue renin-angiotensin system appear to be confusing, its involvement in the physiology and pathophysiology of adipose tissue has been confirmed by several functional studies. Nevertheless, future studies with more carefully described phenotypes are necessary to conclude whether obesity (by stimulation of adipogenic differentiation) and hypertension are associated with changes of renin-angiotensin system activity in adipose tissue. If so, the physiological relevance of this system in animal models and humans may warrant further interest.


Subject(s)
Adipose Tissue/physiology , Adipose Tissue/physiopathology , Hypertension/physiopathology , Obesity/physiopathology , Renin-Angiotensin System/physiology , Humans , Reference Values
8.
Mol Cell Endocrinol ; 160(1-2): 149-56, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10715548

ABSTRACT

Prostacyclin (PGI(2)) and its stable analogue carbacyclin (cPGI(2)) are known to trigger the protein kinase A pathway after binding to the cell surface IP receptor and to promote or enhance terminal differentiation of adipose precursor cells to adipose cells. The early expression of C/EBPbeta and C/EBPdelta is known to be critical for adipocyte differentiation in vitro as well as in vivo. We report herein that in Ob1771 and 3T3-F442A preadipose cells, activation of the IP receptor by specific agonists (PGI(2), cPGI(2) and BMY 45778) is sufficient to up-regulate rapidly the expression of C/EBPbeta and C/EBPdelta. Cyclic AMP-elevating agents are able to substitute for IP receptor agonists, in agreement with the coupling of IP receptor to adenylate cyclase. Consistent with the fact that PGI(2) is released from preadipose cells and behaves as a paracrine/autocrine effector of adipose cell differentiation, the present results favor a key role of prostacyclin by means of the IP receptor and its intracellular signaling pathway in eliciting the critical early expression of both transcription factors.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/genetics , Neoplasm Proteins , Nerve Tissue Proteins , Nuclear Proteins/genetics , Receptors, Prostaglandin/metabolism , Thiazolidinediones , 3T3 Cells , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Proteins , Carrier Proteins/genetics , Cell Differentiation , Clone Cells , Colforsin/administration & dosage , Cyclic AMP/metabolism , Epoprostenol/metabolism , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Kinetics , Mice , Myelin P2 Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Epoprostenol , Receptors, Prostaglandin/agonists , Rosiglitazone , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Thiazoles/administration & dosage , Transcription Factors/metabolism , Up-Regulation
9.
Prostaglandins Other Lipid Mediat ; 57(5-6): 305-17, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10480485

ABSTRACT

To clarify the molecular basis for the prostaglandin (PG) mediated effects in adipose cells at various stages of their development, expression of mRNAs encoding receptors specific for prostaglandin E2, F2alpha and I2 (i.e. EP, FP, and IP receptors) was investigated in differentiating clonal Ob1771 pre-adipocytes, as well as in mouse primary adipose precursor cells and mature adipocytes. We have further characterized the differential expression of mRNAs encoding three subtypes of the EP receptor, i.e. EP1, EP3, and EP4, and examined the expression of mRNAs encoding the three isoforms (alpha, beta, and gamma) of the EP3 receptor. Altogether the results show that the expression of IP, FP, EP1, and EP4 receptor mRNAs was considerably more pronounced in pre-adipose cells than in adipose cells, mRNAs encoding the alpha, beta, and gamma isoforms of the EP3 receptor were all exclusively expressed in freshly isolated mature adipocytes. These data may indicate that PGI2, PGF2alpha, and PGE2 may interact directly with specific receptors in pre-adipose cells, whose transduction mechanisms are known to affect maturation related changes. In mature adipocytes, however, the equipment of mRNAs encoding the EP3 receptor isoforms is in agreement with the well known effect of PGE2 on adenylate cyclase and lipolysis in mature adipocytes.


Subject(s)
Adipocytes/metabolism , Cell Differentiation , RNA, Messenger/genetics , Receptors, Prostaglandin/genetics , Adipocytes/cytology , Animals , Base Sequence , DNA Primers , Female , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Article in English | MEDLINE | ID: mdl-10471126

ABSTRACT

Adipose cell differentiation from adipoblasts to preadipose and to adipose cells is a multistep process. Terminal differentiation of preadipose cells expressing early markers to adipose cells expressing late and very late markers and accumulating triacylglycerol requires a combination of circulating and locally-produced hormones. Prostacyclin (PGI2), one of the major metabolites of arachidonic acid in adipose tissue, has been shown to exert autocrine and paracrine adipogenic effects in vitro. As discussed herein, multiple arguments support the proposition that PGI2 is a key prostanoid involved in adipogenesis.


Subject(s)
Adipose Tissue, Brown/metabolism , Epoprostenol/physiology , Animals , Humans , Prostaglandins/physiology
11.
Diabetes ; 48(4): 890-5, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10102708

ABSTRACT

Plasminogen activator inhibitor 1 (PAI-1) is likely to play a role in vascular disease, primarily in subjects with android obesity. It has been demonstrated that PAI-1 is overexpressed in adipose tissue from obese subjects and that visceral adipose tissue produced more PAI-1 than subcutaneous fat. In the present study, the effect of insulin and glucocorticoids, which are key mediators of adipose tissue metabolism, was examined in relation to PAI-1 synthesis by human adipose tissue explants (HAT), collagenase isolated human adipocytes (IHA), cultured human stromal cells (cSC), and differentiated adipocytes from the murine clonal cell line 3T3-F442A. A significant increase in PAI-1 antigen release (1.5-fold) from HAT was detectable after 16 h of treatment with insulin concentrations of at least 10(-8) mol/l. This was associated with a PAI-1 mRNA increase. Concomitant addition of insulin (10(-8) mol/l) to forskolin (5 x 10(-5) mol/l) reversed the decrease in PAI-1 antigen caused by forskolin alone. No effect on PAI-1 antigen was observed when insulin was incubated with IHA or cSC. 3T3 F442A cells were sensitive to insulin with a four- and twofold increase in PAI-1 antigen and mRNA levels, respectively, after 16 h of stimulation with 10(-8) mol/l. Dexamethasone (DXM) significantly enhanced PAI-1 antigen and mRNA expression by HAT (1.5- and 2.5-fold increase, respectively) at concentrations of at least 10(-8) mol/l. A higher stimulation was observed with IHA (sevenfold increase) and with the differentiated 3T3 F442 cell line. Cortisol was found to be less potent than DXM. No effect was observed when glucocorticoids were incubated with cSC. Coincubation of HAT with insulin (10(-7) mol/l) and DXM (10(-7) mol/l) led to an additive effect on PAI-1 synthesis. These results support the hypothesis that PAI-1 expression in human adipose tissue is controlled by insulin and glucocorticoids and may help to explain the increase in plasma PAI-1 levels observed in patients with android obesity.


Subject(s)
Adipose Tissue/metabolism , Glucocorticoids/pharmacology , Insulin/pharmacology , Plasminogen Activator Inhibitor 1/biosynthesis , 3T3 Cells , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adult , Animals , Culture Techniques , Dexamethasone/pharmacology , Drug Combinations , Female , Humans , Mice , Middle Aged , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/metabolism
12.
Biochem Biophys Res Commun ; 250(1): 77-82, 1998 Sep 08.
Article in English | MEDLINE | ID: mdl-9735335

ABSTRACT

Adipose tissue is an important source of angiotensinogen (AT) after liver. Since an association exists between body mass index, hypertension, and insulin-resistance, the role of insulin on the regulation of AT gene expression and AT secretion was examined in cultured Ob1771 and 3T3-F442A adipose cells. Within a physiological range of concentrations (1-17 nM), insulin exerted a negative effect on the abundance of AT mRNA and the secretion of AT. Alterations of insulin-resistance by treatment of adipose cells with TNF-alpha or the thiazolidinedione BRL49653 led respectively to a decrease or an increase in the potency of insulin to down-regulate AT gene expression, whereas maximal inhibition by insulin increased from 30% in TNFalpha-treated cells to 60% in BRL49653-treated cells. These results suggest that a potential link between insulin resistance and high blood pressure may exist by means of increased AT secretion from adipose tissue, especially in obese subjects.


Subject(s)
Adipose Tissue/drug effects , Angiotensinogen/genetics , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Insulin/pharmacology , Thiazolidinediones , 3T3 Cells , Adipose Tissue/metabolism , Angiotensinogen/metabolism , Animals , Cells, Cultured , Hypoglycemic Agents/pharmacology , Mice , RNA, Messenger/genetics , Rosiglitazone , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
13.
Biochem Biophys Res Commun ; 238(2): 606-11, 1997 Sep 18.
Article in English | MEDLINE | ID: mdl-9299560

ABSTRACT

UCP-2 is a member of the emerging family of UCP homologues. Upon high-fat feeding, UCP-2 mRNA levels are increased in epididymal fat pads of A/J mice, suggesting that the flux of fatty acids entering adipose tissue may regulate UCP-2 gene expression. Since fatty acids act as positive transcriptional regulators of lipid-related genes by means of peroxisome proliferator-activated receptors (PPARs), the regulation of UCP-2 gene expression by PPAR agonists (carbacyclin, alpha-bromopalmitate, BRL49653) has been examined in mouse preadipose and adipose cells in primary cultures or from clonal lines (Ob1771, 3T3-F442A, 1B8). In preadipose cells, carbacyclin and alpha-bromopalmitate are active and BRL49653 shows no effect, whereas all these ligands are active in adipose cells. The stimulatory effect of PPAR agonists is potentiated by RXR agonists in adipose cells. In contrast to the UCP-1 gene, norepinephrine as a cAMP-elevating agent does not enhance the expression of UCP-2 gene. Altogether, the data favor a predominant role of PPARdelta in preadipose cells and the involvement of PPARgamma2 in adipose cells in up-regulating UCP-2 gene expression. Thus, a potential link between fatty acid metabolism and thermogenesis may exist in PPAR-expressing tissues.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Gene Expression Regulation/drug effects , Mitochondria/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Thiazolidinediones , Transcription Factors/agonists , Uncoupling Agents , Animals , Cell Differentiation/genetics , Cell Line , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Mice , Mitochondria/genetics , Palmitates/pharmacology , Rosiglitazone , Thiazoles/pharmacology
14.
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
15.
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
16.
Biochem J ; 322 ( Pt 1): 235-9, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9078267

ABSTRACT

Adipocytes represent an important source of angiotensiongen (AT). Angiotensin II (A-II) stimulates in vitro and in vivo the formation and release of prostacyclin which acts as a potent adipogenic signal in triggering the terminal differentiation of preadipocytes into adipocytes [Darimont, Vassaux, Gaillard. Ailhaud and Négrel (1994) Int. J. Obes. 18, 783-788]. Since fatty acids have been reported to activate in preadipose cells the expression of various differentiation-dependent genes, the role of fatty acids in the regulation of AT gene expression was investigated. Long-chain natural and non-metabolized fatty acids as well as peroxisome proliferators behave as activators of AT gene expression. Accumulation of AT mRNA parallels that of the adipocyte fatty acid-binding protein gene and is primarily due to transcriptional activation of the AT gene. AT mRNA decreases after fatty acid removal (half-life approx. 8 h). Secretion of AT is also observed but appears mainly as a late differentiation-dependent phenomenon. Thus the AT gene appears to be a fatty acid-responsive gene; this regulation provides a potential link between the flux of fatty acids and the potential of adipose tissue to produce AT and possibly A-II.


Subject(s)
Adipocytes/metabolism , Angiotensinogen/genetics , Fatty Acids/physiology , Gene Expression Regulation/drug effects , Neoplasm Proteins , Nerve Tissue Proteins , Stem Cells/metabolism , Adipocytes/cytology , Angiotensinogen/biosynthesis , Angiotensinogen/metabolism , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/drug effects , Carrier Proteins/genetics , Cell Differentiation , Cell Line , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Half-Life , Intracellular Signaling Peptides and Proteins , Mice , Myelin P2 Protein/biosynthesis , Myelin P2 Protein/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Transcription, Genetic
17.
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
18.
FEBS Lett ; 397(1): 117-21, 1996 Nov 11.
Article in English | MEDLINE | ID: mdl-8941726

ABSTRACT

Prostacyclin, one of the major prostanoids generated in adipose tissue, has been previously described as an autocrine/paracrine adipogenic effector, acting, in preadipose cells, by means of cAMP and free Ca2+ as cell surface receptor-mediated messengers. The present study presents evidence for the first time that its stable analogue, carbaprostacyclin, is unique among prostanoids in regulating the expression of two differentiation-dependent genes in preadipose and adipose cells in a way distinct from that elicited by its cell surface receptor. This regulation is likely mediated by some member(s) of the peroxisome proliferator-activated receptor family and suggests that prostacyclin behaves as an intracrine effector of adipose cell differentiation.


Subject(s)
Adipocytes/metabolism , Angiotensinogen/genetics , Carrier Proteins/genetics , Epoprostenol/analogs & derivatives , Gene Expression Regulation/drug effects , Myelin P2 Protein/genetics , Neoplasm Proteins , Nerve Tissue Proteins , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Angiotensinogen/biosynthesis , Animals , Carrier Proteins/biosynthesis , Cell Differentiation , Cell Line , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Epoprostenol/pharmacology , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Hormone Antagonists/pharmacology , Mice , Mifepristone/pharmacology , Myelin P2 Protein/biosynthesis , Prostaglandins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Epoprostenol , Receptors, Prostaglandin/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
...