Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 292(4): E1101-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17164440

RESUMEN

Recent evidence has been presented that expression of lipogenic genes is downregulated in adipose tissue of ob/ob mice as well as in human obesity, suggesting a functionally lipoatrophic state. Using (2)H(2)O labeling, we measured three adipose tissue biosynthetic processes concurrently: triglyceride (TG) synthesis, palmitate de novo lipogenesis (DNL), and cell proliferation (adipogenesis). To determine the effect of the ob/ob mutation (leptin deficiency) on these parameters, adipose dynamics were compared in ob/ob, leptin-treated ob/ob, food-restricted ob/ob, and lean control mice. Adipose tissue fluxes for TG synthesis, de novo lipogenesis (DNL), and adipogenesis were dramatically increased in ob/ob mice compared with lean controls. Low-dose leptin treatment (2 microg/day) via miniosmotic pump suppressed all fluxes to control levels or below. Food restriction in ob/ob mice only modestly reduced DNL, with no change in TG synthesis or adipogenesis. Measurement of mRNA levels in age-matched ob/ob mice showed generally normal expression levels for most of the selected lipid anabolic genes, and leptin treatment had, with few exceptions, only modest effects on their expression. We conclude that leptin deficiency per se results in marked elevations in flux through diverse lipid anabolic pathways in adipose tissue (DNL, TG synthesis, and cell proliferation), independent of food intake, but that gene expression fails to reflect these changes in flux.


Asunto(s)
Tejido Adiposo/metabolismo , Expresión Génica , Lipogénesis/genética , Ratones Obesos , Obesidad/fisiopatología , Adipogénesis , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Peso Corporal , Ingestión de Alimentos , Femenino , Privación de Alimentos , Insulina/sangre , Leptina/sangre , Leptina/deficiencia , Leptina/farmacología , Lipólisis , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Concentración Osmolar , Palmitatos/metabolismo , Triglicéridos/biosíntesis
2.
Leukemia ; 19(8): 1404-10, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15959531

RESUMEN

Dlk1 (Pref-1) is a transmembrane and secreted protein, which is a member of the epidermal growth factor-like family, homologous to Notch/Delta/Serrate. We have found by real-time RT-PCR that Dlk1 mRNA levels were high in CD34(+) cells in 10 of 12 MDS samples compared with CD34(+) cells from 11 normals. Also, Dlk1 mRNA was elevated in mononuclear, low density bone marrow cells from 11/38 MDS patients, 5/11 AML M6 and 2/4 AML M7 samples. Furthermore, 5/6 erythroleukemia and 2/2 megakaryocytic leukemia cell lines highly expressed Dlk1 mRNA. Levels of Dlk1 mRNA markedly increased during megakaryocytic differentiation of both CMK megakaryoblasts as well as normal CD34(+) hematopoietic stem cells. High serum levels of Dlk1 occurred in RA (4/10) and essential thrombocythemia (2/10) patients. Functional studies showed that forced expression of Dlk1 enhanced proliferation of K562 cells growing in 1% fetal bovine serum. Analysis of hematopoiesis of Dlk1 knockout mice suggested that Dlk1 contributed to granulocyte, megakaryocyte and B-cell clonogenic growth and was needed for generation of splenic B-cells. In summary, Dlk1 is overexpressed in selected samples of MDS (especially RA and RAEB) and AML (particularly M6, M7), and it appears to be associated with normal development of megakaryocytes and B cells.


Asunto(s)
Glicoproteínas/genética , Enfermedades Hematológicas/patología , Hematopoyesis/genética , Animales , Antígenos CD34 , Estudios de Casos y Controles , Diferenciación Celular , Proliferación Celular , Células Clonales/patología , Regulación de la Expresión Génica , Glicoproteínas/sangre , Glicoproteínas/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Humanos , Leucemia/genética , Leucemia/patología , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Horm Metab Res ; 34(11-12): 664-70, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12660879

RESUMEN

Adipose tissue is the source of a wide array of factors of great biological significance that are involved in many aspects of organism physiology, including appetite control and peripheral metabolism. Here, we describe two secreted factors from adipose tissue that inhibit adipogenesis. Pref-1 is a preadipocyte secreted factor synthesized as a transmembrane protein that undergoes proteolitic cleavage to generate two distinct soluble forms. In vitro assays have demonstrated that only the large soluble form of Pref-1 is biologically active and inhibits adipocyte differentiation. In vivo, mice lacking Pref-1 expression show accelerated fat deposition, perinatal mortality and growth retardation as well as distinct skeletal malformations, highlighting the importance of Pref-1 during mouse development in addition to its role in adipose tissue development. ADSF/resistin is secreted by adipocytes and inhibits adipose cells differentiation in vitro. Its function is still unclear, but its expression and high circulating levels have been associated with an impairment of insulin action. The findings show that Pref-1 and possibly ADSF/resistin secretion control fat cell differentiation and adipose tissue development.


Asunto(s)
Adipocitos/citología , Adipocitos/fisiología , Hormonas Ectópicas/fisiología , Proteínas de la Membrana/fisiología , Proteínas Represoras/fisiología , Tejido Adiposo/crecimiento & desarrollo , Aldosterona/fisiología , Animales , Proteínas de Unión al Calcio , Diferenciación Celular/fisiología , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular , Ratones , Factor de Crecimiento Nervioso , Proteínas/fisiología , Resistina
4.
J Biol Chem ; 276(14): 11252-6, 2001 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-11278254

RESUMEN

A 12.5-kDa cysteine-rich adipose tissue-specific secretory factor (ADSF/resistin) is a novel secreted protein rich in serine and cysteine residues with a unique cysteine repeat motif of CX(12)CX(8)CXCX(3)CX(10)CXCXCX(9)CC. A single 0.8-kilobase mRNA coding for this protein was found in various murine white adipose tissues including inguinal and epididymal fats and also in brown adipose tissue but not in any other tissues examined. Two species of mRNAs with sizes of 1.4 and 0.8 kilobases were found in rat adipose tissue. Sequence analysis indicates that this is because of two polyadenylation signals, the proximal one with the sequence AATACA with a single base mismatch from murine AATAAA and the distal consensus sequence AATAAA. The mRNA level was markedly increased during 3T3-L1 and primary preadipocyte differentiation into adipocytes. Its expression in adipose tissue is under tight nutritional and hormonal regulation; the mRNA level was very low during fasting and increased 25-fold when fasted mice were refed a high carbohydrate diet. It was also very low in adipose tissue of streptozotocin-diabetes and increased 23-fold upon insulin administration. Upon treatment with the conditioned medium from COS cells transfected with the expression vector, conversion of 3T3-L1 cells to adipocytes was inhibited by 80%. The regulated expression pattern suggesting this factor as an adipose sensor for the nutritional state of the animals and the inhibitory effect on adipocyte differentiation implicate its function as a feedback regulator of adipogenesis.


Asunto(s)
Adipocitos/fisiología , Hormonas Ectópicas/fisiología , Proteínas , Adipocitos/patología , Secuencia de Aminoácidos , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Péptidos y Proteínas de Señalización Intercelular , Ratones , Datos de Secuencia Molecular , Factor de Crecimiento Nervioso , ARN Mensajero/análisis , Ratas , Resistina , Alineación de Secuencia , Estreptozocina
5.
Int J Obes Relat Metab Disord ; 24 Suppl 4: S15-9, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11126233

RESUMEN

During conversion of preadipocytes to adipocytes, growth arrest and subsequent activation of adipocyte genes by the transcription factors, C/EBPalpha and PPARgamma, lead to adipogenesis. During differentiation, these cells not only start expressing those genes necessary for adipocyte function, but also undergo changes in morphology to become rounded lipid filled adipocytes. Various factors in cell-cell communication or cell-matrix interaction may govern whether preadipocytes are kept in an undifferentiated state or undergo differentiation. In an attempt to identify molecules that play critical roles in the conversion of preadipocytes to adipocytes, we cloned by differential screening several regulatory molecules, including pref-1. Pref-1 is an inhibitor of adipocyte differentiation and is synthesized as a plasma membrane protein containing 6 EGF-repeats in the extracellular domain. Pref-1 is highly expressed in 3T3-L1 preadipocytes, but is not detectable in mature fat cells. Dexamethasone, a component of standard differentiation agents, inhibits pref-1 transcription and thereby promotes adipogenesis. Downregulation of pref-1 is required for adipose conversion and constitutive expression of pref-1 inhibits adipogenesis. Conversely, decreasing pref-1 levels by antisense pref-1 transfection greatly enhances adipogenesis. The ectodomain of pref-1 is cleaved to generate a biologically active 50kDa soluble form. There are four major forms of membrane pref-1 resulting from alternate splicing. Two of these forms which have a deletion that includes the putative processing site proximal to the membrane do not produce a biologically active soluble form. This indicates that alternate splicing may determine the range of action, juxtacrine or paracrine, of pref-1.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/crecimiento & desarrollo , Diferenciación Celular , Proteínas de la Membrana/fisiología , Proteínas Represoras/fisiología , Proteínas de Unión al Calcio , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas Represoras/química , Proteínas Represoras/genética
6.
Proc Natl Acad Sci U S A ; 97(19): 10619-24, 2000 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-10962028

RESUMEN

The transcription of fatty acid synthase (FAS), a central enzyme in de novo lipogenesis, is dramatically induced by fasting/refeeding and insulin. We reported that upstream stimulatory factor binding to the -65 E-box is required for induction of the FAS transcription by insulin in 3T3-L1 adipocytes. On the other hand, we recently found that two upstream 5' regions are required for induction in vivo by fasting/refeeding and insulin; one at -278 to -131 albeit at a low level, and the other at -444 to -278 with an E-box at -332 where upstream stimulatory factor functions for maximal induction. Here, we generated double transgenic mice carrying the chloramphenicol acetyltransferase reporter driven by the various 5' deletions of the FAS promoter region and a truncated active form of the sterol regulatory element (SRE) binding protein (SREBP)-1a. We found that SREBP participates in the nutritional regulation of the FAS promoter and that the region between -278 and -131 bp is required for SREBP function. We demonstrate that SREBP binds the -150 canonical SRE present between -278 and -131, and SREBP can function through the -150 SRE in cultured cells. These in vivo and in vitro results indicate that SREBP is involved in the nutritional induction of the FAS promoter via the -278/-131 region and that the -150 SRE is the target sequence.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Proteínas de Unión al ADN/fisiología , Ácido Graso Sintasas/genética , Alimentos , Regulación Enzimológica de la Expresión Génica , Proteínas Nucleares/fisiología , Factores de Transcripción , Células 3T3 , Animales , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/genética , Cartilla de ADN , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Inanición , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Activación Transcripcional
7.
J Biol Chem ; 275(22): 16845-50, 2000 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-10828068

RESUMEN

In an attempt to study molecules that play a regulatory role early in adipocyte differentiation, we identified by differential display ENC-1, a Drosophila kelch-related protein. ENC-1 colocalizes with actin filaments. ENC-1 is expressed in adipose tissue, specifically in the adipose-derived stroma-vascular fraction. ENC-1 mRNA levels are transiently increased 8-12-fold early in in vitro adipocyte differentiation of primary cells of the adipose-derived stroma-vascular fraction and of 3T3-L1 cells. Treatment with the adipogenic inducers dexamethasone and methylisobutylxanthine causes an increase in ENC-1 mRNA levels specifically in preadipocytes, and methylisobutylxanthine is the main effector of ENC-1 expression. The induction of ENC-1 precedes expression of the transcription factors, peroxisome proliferator-activated receptor (PPARgamma) and CCAAT/enhancer-binding protein (C/EBPalpha), and other adipocyte markers including adipocyte fatty acid-binding protein. The ENC-1 induction correlates with the subsequent differentiation of primary stroma-vascular cells into adipocytes. Furthermore, decreasing the endogenous ENC-1 levels by stable antisense transfection, thereby preventing the transient induction, effectively inhibits 3T3-L1 adipocyte differentiation. Overall, these studies indicate that ENC-1, an actin-binding protein, plays a regulatory role early in adipocyte differentiation when cytoskeletal reorganization and cell shape change from fibroblastic preadipocytes to spherical adipocytes occur.


Asunto(s)
Actinas/metabolismo , Adipocitos/citología , Diferenciación Celular/fisiología , Proteínas de Microfilamentos/genética , Neuropéptidos , Proteínas Nucleares , 1-Metil-3-Isobutilxantina/farmacología , Células 3T3 , Adipocitos/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Dexametasona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/fisiología
8.
J Biol Chem ; 275(14): 10121-7, 2000 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-10744693

RESUMEN

We previously reported that 2.1 kilobase pairs of the 5'-flanking sequence are sufficient for tissue-specific and hormonal/metabolic regulation of the fatty-acid synthase (FAS) gene in transgenic mice. We also demonstrated that the -65 E-box is required for insulin regulation of the FAS promoter using 3T3-L1 adipocytes in culture. To further define sequences required for FAS gene expression, we generated transgenic mice carrying from -644, -444, -278, and -131 to +67 base pairs of the rat FAS 5'-flanking sequence fused to the chloramphenicol acetyltransferase (CAT) reporter gene. Similar to the expression observed with -2100-FAS-CAT transgenic mice, transgenic mice harboring -644-FAS-CAT and -444-FAS-CAT expressed high levels of CAT mRNA only in lipogenic tissues (liver and adipose tissue) in a manner identical to the endogenous FAS mRNA. In contrast, -278-FAS-CAT and -131-FAS-CAT transgenic mice did not show appreciable CAT expression in any of the tissues examined. When previously fasted mice were refed a high carbohydrate, fat-free diet, CAT mRNA expression in transgenic mice harboring -644-FAS-CAT and -444-FAS-CAT was induced dramatically in liver and adipose tissue. The induction was virtually identical to that observed in -2100-FAS-CAT transgenic mice and to the endogenous FAS mRNA. In contrast, -278-FAS-CAT transgenic mice showed induction by feeding, but at a much lower magnitude in both liver and adipose tissue. The -131-FAS-CAT transgenic mice did not show any CAT expression either when fasted or refed a high carbohydrate diet. To study further the effect of insulin, we made these transgenic mice insulin-deficient by streptozotocin treatment. Insulin administration to the streptozotocin-diabetic mice increased CAT mRNA levels driven by the -644 FAS and -444 FAS promoters in liver and adipose tissue, paralleling the endogenous FAS mRNA levels. In the case of -278-FAS-CAT, the induction observed was at a much lower magnitude, and deletion to -131 base pairs did not show any increase in CAT expression by insulin. This study demonstrates that the sequence requirement for FAS gene regulation employing an in vitro culture system does not reflect the in vivo situation and that two 5'-flanking regions are required for proper nutritional and insulin regulation of the FAS gene. Cotransfection of the upstream stimulatory factor and various FAS promoter-luciferase constructs as well as in vitro binding studies suggest a function for the upstream stimulatory factor at both the -65 and -332 E-box sequences.


Asunto(s)
Regiones no Traducidas 5'/genética , Ácido Graso Sintasas/genética , Regulación Enzimológica de la Expresión Génica , Regiones Promotoras Genéticas , Tejido Adiposo/enzimología , Animales , Cloranfenicol O-Acetiltransferasa/genética , Ácido Graso Sintasas/biosíntesis , Hígado/enzimología , Ratones , Ratones Transgénicos , ARN Mensajero/genética , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Mapeo Restrictivo
9.
J Nutr ; 130(2S Suppl): 315S-320S, 2000 02.
Artículo en Inglés | MEDLINE | ID: mdl-10721895

RESUMEN

Expression of critical enzymes in fatty acid and fat biosynthesis is tightly controlled by nutritional and hormonal stimuli. The expression of fatty acid synthase, which catalyzes all reactions for synthesis of palmitate from acetyl-CoA and malonyl-CoA, and of mitochondrial glycerol-3-phosphate acyltransferase, which catalyzes the first acylation step in glycerophospholipid synthesis, is decreased to an undetectable level during fasting. Food intake, especially a high carbohydrate, fat-free diet after fasting, causes a dramatic increase in the transcription of these genes. Insulin secretion is increased during feeding and has a positive effect on expression. By using adipocytes in culture and transgenic mice that express the reporter gene driven by the fatty acid synthase promoter, the cis-acting sequence that mediates insulin regulation of the fatty acid synthase promoter was defined. Upstream stimulatory factors (USF) that bind to the -65 E-box are required for insulin-mediated transcriptional activation of the fatty acid symthase gene. Sterol regulatory element binding protein (SREBP)-1 may be also involved in induction of these genes during feeding. Using specific inhibitors and expressing various signaling molecules, we found that insulin regulation of the fatty acid synthase promoter is mediated by the phosphatidylinositol (PI)3-kinase signaling pathway and that protein kinase B/akt is a downstream effector.


Asunto(s)
Ácido Graso Sintasas/fisiología , Ácidos Grasos/biosíntesis , Glicerol-3-Fosfato O-Aciltransferasa/biosíntesis , Insulina/fisiología , Hígado/enzimología , Mitocondrias/metabolismo , Animales , Ayuno/metabolismo , Ácido Graso Sintasas/genética , Regulación Enzimológica de la Expresión Génica , Hígado/metabolismo , Mitocondrias/enzimología , Transducción de Señal/fisiología
10.
J Biol Chem ; 274(49): 34728-34, 1999 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-10574940

RESUMEN

Glycerol-3-phosphate acyltransferase (GPAT) catalyzes the initial and committed step in glycerolipid biosynthesis. We previously cloned the cDNA sequence to murine mitochondrial GPAT (Yet, S-F., Lee, S., Hahm, Y. T., and Sul, H.S. (1993) Biochemistry 32, 9486-9491). We expressed the protein in insect cells which was targeted to mitochondria, purified, and reconstituted mitochondrial GPAT activity using phospholipids (Yet, S.-F., Moon, Y., and Sul, H. S. (1995) Biochemistry 34, 7303-7310). Deletion of the seven amino acids from mitochondrial GPAT, (312)IFLEGTR(318), which is highly conserved among acyltransferases in glycerolipid biosynthesis, drastically reduced mitochondrial GPAT activity. Treatment of mitochondrial GPAT with arginine-modifying agents, phenylglyoxal and cyclohexanedione, inactivated the enzyme. Two highly conserved arginine residues, Arg-318, in the seven amino stretch, and Arg-278, were identified. Substitution of Arg-318 with either alanine, histidine, or lysine reduced the mitochondrial GPAT activity by over 90%. On the other hand, although substitution of Arg-278 with alanine and histidine decreased mitochondrial GPAT activity by 90%, replacement with lysine reduced activity by only 25%. A substitution of the nonconserved Arg-279 with either alanine, histidine, or lysine did not alter mitochondrial GPAT activity. Moreover, R278K mitochondrial GPAT still showed sensitivity to arginine-modifying agents, as in the case of wild-type mitochondrial GPAT. These results suggest that Arg-318 may be critical for mitochondrial GPAT activity, whereas Arg-278 can be replaced by a basic amino acid. Examination of the other conserved residues in the seven amino acid stretch revealed that Phe-313 and Glu-315 are also important, but conservative substitutions can partially maintain activity; substitution with alanine reduced activity by 83 and 72%, respectively, whereas substituting Phe-313 with tyrosine and Glu-315 with glutamine had even lesser effect. In addition, there was no change in fatty acyl-CoA selectivity. Kinetic analysis of the R318K and R318A mitochondrial GPAT showed an 89 and 95%, respectively, decrease in catalytic efficiency but no major change in substrate binding as indicated by the K(m) values for palmitoyl-CoA and glycerol 3-phosphate. These studies indicate importance of the conserved seven amino acid stretch for mitochondrial GPAT activity and the significance of Arg-318 for catalysis.


Asunto(s)
Glicerol-3-Fosfato O-Aciltransferasa/química , Glicerol-3-Fosfato O-Aciltransferasa/metabolismo , Mitocondrias/enzimología , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Secuencia de Bases , Northern Blotting , Western Blotting , Células COS , Catálisis , Secuencia Conservada , Ciclohexanonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Glicerol-3-Fosfato O-Aciltransferasa/antagonistas & inhibidores , Glicerol-3-Fosfato O-Aciltransferasa/genética , Cinética , Ratones , Datos de Secuencia Molecular , Fenilglioxal/farmacología , Plásmidos/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Transfección , Tripsina/farmacología
11.
J Biol Chem ; 274(18): 12632-41, 1999 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-10212243

RESUMEN

Pref-1 is an epidermal growth factor-like domain-containing transmembrane protein that is cleaved to generate a soluble factor. It is abundant in 3T3-L1 preadipocytes but absent in mature adipocytes. Constitutive expression of pref-1 or the addition of its ectodomain inhibits adipogenesis. We find that the pref-1 gene is an early target of dexamethasone, a component of the dexamethasone/methylisobutylxanthine differentiation mixture used routinely for adipoconversion. The time course of the decrease in pref-1 mRNA by dexamethasone reflected the pref-1 mRNA half-life determined by actinomycin D treatment. Nuclear run-on assays showed that dexamethasone attenuates pref-1 transcription. We demonstrate a correlation between pref-1 down-regulation and adipoconversion by varying the time period and concentration of dexamethasone. Increasing the dexamethasone treatment from 2 to 4 days resulted in a time-dependent pref-1 down-regulation and increased differentiation as measured by adipocyte marker mRNAs. The dexamethasone concentration between 1 and 10 nM showed a dose-dependent decrease in pref-1 mRNA and an enhancement of adipogenesis. To test the hypothesis that dexamethasone initiation of adipoconversion may be via down-regulation of pref-1, we lowered endogenous pref-1 mRNA levels by stably transfecting 3T3-L1 preadipocytes with antisense pref-1. At 1 microM, antisense cells had enhanced adipose conversion; a similar degree of differentiation occurred with 2 nM dexamethasone, a concentration that does not support differentiation of control 3T3-L1 cells. We conclude that dexamethasone-mediated repression of pref-1 contributes to the mechanisms whereby glucocorticoids promote adipogenesis.


Asunto(s)
Adipocitos/efectos de los fármacos , Diferenciación Celular/genética , Dexametasona/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas Represoras/genética , 1-Metil-3-Isobutilxantina/farmacología , Células 3T3 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteínas de Unión al Calcio , Péptidos y Proteínas de Señalización Intercelular , Cinética , Ratones , Oligonucleótidos Antisentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección
13.
J Biol Chem ; 273(48): 31751-8, 1998 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-9822638

RESUMEN

Preadipocyte factor-1 (Pref-1) is a transmembrane epidermal growth factor-like domain-containing protein highly expressed in 3T3-L1 preadipocytes, but is undetectable in mature fat cells; this down-regulation is required for adipocyte differentiation. We show here that pref-1 transcription is markedly suppressed during adipose conversion and results in decreased Pref-1 RNA levels. Using 3T3-L1 cells stably transfected with Pref-1 5'-deletion constructs truncated at -6000, -2100, -1300, -692, -300, -235, -193, -183, -170, -93, and -45 base pairs, we determined that the -183 to -170 region is responsible for the suppression of the pref-1 gene during adipogenesis. This is distinct from the -93 to -45 sequence important for pref-1 promoter activity in preadipocytes. The placement of a 40-base pair -193 to -154 pref-1 sequence containing the putative SAD (suppression in adipocyte differentiation) element upstream of the SV40 promoter decreased promoter activity by 85% upon adipocyte differentiation, compared with 40% observed with the SV40 promoter alone. The SAD element is therefore sufficient for adipocyte differentiation-dependent down-regulation of a heterologous promoter. A DNA-protein complex was observed when the -193 to -174 sequence was used with 3T3-L1 nuclear extracts in gel mobility shift assays. Competition with oligonucleotides harboring base substitution mutations identified a core sequence of -183AAAGA-179 as crucial for DNA-protein complex formation. UV cross-linking predicts that an approximately 63-kDa protein specifically binds the SAD element.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas Represoras/genética , Transcripción Genética , Células 3T3 , Animales , Secuencia de Bases , Sitios de Unión , Proteínas de Unión al Calcio , Diferenciación Celular , Péptidos y Proteínas de Señalización Intercelular , Luciferasas , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/metabolismo , Ratones , Mutagénesis , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/biosíntesis , Proteínas Represoras/metabolismo , Eliminación de Secuencia , Supresión Genética , Transfección
14.
J Biol Chem ; 273(39): 25420-6, 1998 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-9738010

RESUMEN

Fatty acid synthase (FAS) is a critical enzyme in de novo lipogenesis. It catalyzes the seven steps in the conversion of malonyl-CoA and acetyl-CoA to palmitate. We have shown that the rate of FAS transcription is induced dramatically when fasted animals are refed with a high carbohydrate, fat-free diet or when streptozotocin-diabetic mice are given insulin. The FAS promoter was up-regulated by insulin through the proximal insulin response sequence containing an E-box motif at the -65-base pair position. Binding of upstream stimulatory factors to the -65 E-box is functionally required for insulin regulation of the FAS promoter. In the present study, we characterized signaling pathways in the insulin stimulation of FAS transcription using specific inhibitors for various signaling molecules and transfecting engineered phosphatidylinositol (PI) 3-kinase subunits and protein kinase B (PKB)/Akt. PD98059 and rapamycin, which inhibit MAP kinase and P70 S6 kinase, respectively, had little effect on the insulin-stimulated FAS promoter activity in 3T3-L1 adipocytes. On the other hand, wortmannin and LY294002, which specifically inactivate PI 3-kinase, strongly inhibited the insulin-stimulated FAS promoter activity. As shown in RNase protection assays, LY294002 also inhibited insulin stimulation of the endogenous FAS mRNA levels in 3T3-L1 adipocytes. Cotransfection of expression vectors for the constitutively active P110 subunit of PI 3-kinase resulted in an elevated FAS promoter activity in the absence of insulin and a loss of further insulin stimulation. Transfecting a dominant negative P85 subunit of PI 3-kinase decreased FAS promoter activity and blocked insulin stimulation. Furthermore, cotransfected wild-type PKB/Akt increased FAS promoter activity in the absence of insulin and a loss of insulin responsiveness of the FAS promoter. On the other hand, kinase-dead PKB/Akt acted in a dominant negative manner to decrease the FAS promoter activity and abolished its insulin responsiveness. These results demonstrate that insulin stimulation of fatty acid synthase promoter is mediated by the PI 3-kinase pathway and that PKB/Akt is involved as a downstream effector.


Asunto(s)
Ácido Graso Sintasas/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Insulina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Ratones , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo
15.
Annu Rev Nutr ; 18: 331-51, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9706228

RESUMEN

The activities of critical enzymes in fatty acid and triacylglycerol biosynthesis are tightly controlled by different nutritional, hormonal, and developmental conditions. Feeding previously fasted animals high-carbohydrate, low-fat diets causes a dramatic induction of enzymes-such as fatty acid synthase (FAS) and mitochondrial glycerol-3-phosphate acyltransferase (GPAT)-involved in fatty acid and triacylglycerol synthesis. During fasting and refeeding, transcription of these two enzymes is coordinately regulated by nutrients and hormones, such as glucose, insulin, glucagon, glucocorticoids, and thyroid hormone. Insulin stimulates transcription of the FAS and mitochondrial GPAT genes, and glucagon antagonizes the insulin effect through the cis-acting elements within the promoters and their bound trans-acting factors. This review discusses advances made in the understanding of the transcriptional regulation of FAS and mitochondrial GPAT genes, with emphasis on elucidation of the mechanisms by which multiple nutrients and hormones achieve their effects.


Asunto(s)
Ácido Graso Sintasas/genética , Glicerol-3-Fosfato O-Aciltransferasa/genética , Hormonas , Fenómenos Fisiológicos de la Nutrición , Transcripción Genética , Animales , Regulación de la Expresión Génica , Humanos , Mitocondrias/enzimología
16.
Physiol Rev ; 78(3): 783-809, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9674695

RESUMEN

The adipocyte plays a critical role in energy balance. Adipose tissue growth involves an increase in adipocyte size and the formation of new adipocytes from precursor cells. For the last 20 years, the cellular and molecular mechanisms of adipocyte differentiation have been extensively studied using preadipocyte culture systems. Committed preadipocytes undergo growth arrest and subsequent terminal differentiation into adipocytes. This is accompanied by a dramatic increase in expression of adipocyte genes including adipocyte fatty acid binding protein and lipid-metabolizing enzymes. Characterization of regulatory regions of adipose-specific genes has led to the identification of the transcription factors peroxisome proliferator-activated receptor-gamma (PPAR-gamma) and CCAAT/enhancer binding protein (C/EBP), which play a key role in the complex transcriptional cascade during adipocyte differentiation. Growth and differentiation of preadipocytes is controlled by communication between individual cells or between cells and the extracellular environment. Various hormones and growth factors that affect adipocyte differentiation in a positive or negative manner have been identified. In addition, components involved in cell-cell or cell-matrix interactions such as preadipocyte factor-1 and extracellular matrix proteins are also pivotal in regulating the differentiation process. Identification of these molecules has yielded clues to the biochemical pathways that ultimately result in transcriptional activation via PPAR-gamma and C/EBP. Studies on the regulation of the these transcription factors and the mode of action of various agents that influence adipocyte differentiation will reveal the physiological and pathophysiological mechanisms underlying adipose tissue development.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/citología , Tejido Adiposo/crecimiento & desarrollo , Animales , Diferenciación Celular/fisiología , Humanos
17.
Artículo en Inglés | MEDLINE | ID: mdl-9594578

RESUMEN

Adipocytes have highly specialized function of accumulating fat as stored energy that can be used during periods of food deprivation. The process of fat synthesis and development of adipose tissue are under hormonal and nutritional control. This review first describes transcription of the two critical enzymes involved in fat synthesis, fatty acid synthase and mitochondrial glycerol-3-phosphate acyltransferase, is decreased to an undetectable level during fasting. Food intake, especially a high carbohydrate, fat-free diet, subsequent to fasting causes dramatic increase in transcription of these genes. Insulin secretion is increased during feeding, having a positive effect, whereas cAMP, which mediates the effect of glucagon which increases during fasting, has a negative effect on transcription of these genes. Using adipocytes in culture and in transgenic mice that express liciferase driven by the fatty acid synthase promoter, cis-acting and trans-acting factors that may mediate the transcriptional regulation were examined. Upstream stimulatory factors (USFs) that bind to -65 E-box are required for insulin-mediated transcriptional activation of the fatty acid synthase gene. This review next describes how pref-1 is a novel inhibitor of adipose differentiation and is a plasma membrane protein containing six EGF-repeats in the extracellular domain. Pref-1 is highly expressed in 3T3-L1 preadipocytes, but is not detectable in mature fat cells. Down regulation of pref-1 is required for adipose differentiation, and constitutive expression of pref-1 inhibits adipogenesis. Moreover, the ectodomain of pref-1 is cleaved to generate a biologically active 50 kDa soluble form. There are four major forms of membrane pref-1 resulting from alternate splicing, but two of the forms with a larger deletion do not produce biologically active soluble form, indicating that alternate splicing determines the range of action, juxtacrine or paracrine, of the pref-1.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/metabolismo , Lípidos/biosíntesis , Tejido Adiposo/citología , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio , Diferenciación Celular/efectos de los fármacos , Ácido Graso Sintasas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/farmacología , Ratones , Datos de Secuencia Molecular , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/farmacología , Homología de Secuencia de Aminoácido , Activación Transcripcional
18.
Biochim Biophys Acta ; 1348(1-2): 17-26, 1997 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-9370312

RESUMEN

Glycerol-3-phosphate acyltransferase (GPAT) is the first committed, and presumed to be a rate-limiting, step in glycerophospholipid biosynthesis. There are two isoforms of GPAT, a mitochondrial and a microsomal form. Mitochondrial GPAT has recently been purified and its gene has been cloned and expressed in baculovirus-infected cells. The GPAT activity was reconstituted using the purified enzyme and various phospholipids. Mitochondrial GPAT prefers saturated fatty acyl-CoA as a substrate. This preference may contribute to the observed asymmetric distribution of saturated and unsaturated fatty acids at the sn-1 and sn-2 positions of cellular glycerophospholipids. A region of homology to various acyltransferases that may be important for catalysis or fatty acyl-CoA binding is present in mitochondrial GPAT. Mitochondrial GPAT is upregulated at the transcriptional level by refeeding a high carbohydrate, fat-free diet to previously fasted mice and by insulin administration to diabetic animals, whereas microsomal GPAT activity is largely unaffected by these treatments.


Asunto(s)
Glicerol-3-Fosfato O-Aciltransferasa/metabolismo , Mitocondrias/enzimología , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Animales , Clonación Molecular , Glicerol-3-Fosfato O-Aciltransferasa/química , Glicerol-3-Fosfato O-Aciltransferasa/genética , Ratones , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
19.
J Biol Chem ; 272(42): 26367-74, 1997 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-9334210

RESUMEN

Fatty acid synthase (FAS) plays a central role in de novo lipogenesis in mammals. We have shown that FAS transcription rate is induced dramatically when fasted animals are refed with a high carbohydrate diet or when streptozotocin-diabetic mice are given insulin. We also reported that FAS gene transcription was up-regulated by insulin through the proximal promoter region from -71 to -50 and that upstream stimulatory factors (USFs), including USF1 and USF2, interact with this region in vitro. In the present study, by using site-directed mutagenesis of the -71/-50 region and correlating functional assays of the mutated promoter with USF binding activities, we demonstrate that the -65/-60 E-box motif (5'-CATGTG-3') is functionally required for insulin regulation and that USFs are in vivo components of the insulin response complex. Mutation of the -65/-60 E-box sequence abolished insulin response in both transiently and stably transfected 3T3-L1 adipocytes in the -2. 1 kb promoter context, which contains all the necessary regulatory elements of the promoter based on our previous transgenic mice studies, and in the minimal -67 promoter context. Gel mobility shift assays demonstrated that USFs can no longer bind to the -71/-50 promoter region when the E-box is mutated. Cotransfection of USF1 and USF2 expression vectors with the FAS promoter-luciferase reporter constructs increased insulin-stimulated FAS promoter activity. Moreover, cotransfection of dominant negative USF1 and USF2 mutants lacking the DNA binding domain inhibited the insulin stimulation of the FAS promoter activity. On the other hand, site-directed mutagenesis of the -65/-60 E-box surrounding sequences within the overlapped tandem copies of sterol regulatory element-binding protein (SREBP) binding sites prevented SREBP from binding to -71/-50 promoter region in vitro but had no effect on insulin regulation of the FAS promoter in vivo. When rat liver nuclear extracts were used in gel mobility shift assays, only USF-containing protein-DNA complexes that can be supershifted by specific USF antibodies were observed. These results demonstrate that upstream stimulatory factor binding to the E-box at -65 is required for insulin regulation of the fatty acid synthase promoter.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Ácido Graso Sintasas/genética , Insulina/fisiología , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Ratones , Proteínas Nucleares/metabolismo , Unión Proteica , Ratas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Factores Estimuladores hacia 5'
20.
Mol Cell Biol ; 17(2): 977-88, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9001251

RESUMEN

pref-1 is an epidermal growth factor-like repeat protein present on the surface of preadipocytes that functions in the maintenance of the preadipose state. pref-1 expression is completely abolished during 3T3-L1 adipocyte differentiation. Bypassing this downregulation by constitutive expression of full-length transmembrane pref-1 in preadipocytes drastically inhibits differentiation. For the first time, we show processing of cell-associated pref-1 to generate both a soluble pref-1 protein of approximately 50 kDa that corresponds to the ectodomain and also smaller products of 24 to 25 kDa and 31 kDa. Furthermore, while all four of the alternately spliced forms of pref-1 produce cell-associated protein, only the two largest of the four alternately spliced isoforms undergo cleavage in the juxtamembrane region to release the soluble 50-kDa ectodomain. We demonstrate that addition of Escherichia coli-expressed pref-1 ectodomain to 3T3-L1 preadipocytes blocks differentiation, thus overriding the adipogenic actions of dexamethasone and methylisobutylxanthine. The inhibitory effects of the pref-1 ectodomain are blocked by preincubation of the protein with pref-1 antibody. That the ectodomain alone is sufficient for inhibition demonstrates that transmembrane pref-1 can be processed to generate an inhibitory soluble form, thereby greatly extending its range of action. Furthermore, we present evidence that alternate splicing is the mechanism that governs the production of transmembrane versus soluble pref-1, thereby determining the mode of action, juxtacrine or paracrine, of the pref-1 protein.


Asunto(s)
Adipocitos/citología , Proteínas de la Membrana/metabolismo , Proteínas Represoras/metabolismo , Células 3T3 , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Células COS , Proteínas de Unión al Calcio , Diferenciación Celular , Membrana Celular , ADN Complementario , Escherichia coli/genética , Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/farmacología , Ratones , Datos de Secuencia Molecular , Peso Molecular , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/farmacología , Solubilidad , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...