Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Data Brief ; 9: 635-641, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27766294

RESUMO

This article describes qPCR analysis for the Adig/Smaf1 gene in multiple in vitro adipocyte differentiation models including white and brown adipogenesis, cell lines and primary cultures. The article also contains qPCR data for transcript levels of Adig/Smaf1 in a wide panel of murine tissues. Expression of Adig/Smaf1 transcript in white and brown adipose tissue in fasted and refed mice is reported and also data for Adig/Smaf1 transcript expression in genetically obese ob/ob mice. Data on the effects of siRNA-mediated knockdown of Srebp1c on Adig/Smaf1 transcript levels in 3T3-L1 adipocytes are shown. Luciferase reporter assays provide data for regulation of an ~ 2 kb fragment of the 5' flanking region of Adig/Smaf1 gene by PPARγ/RXRα. This data is related to a research article describing Adig/Smaf1 protein expression, "Expression, regulation and functional assessment of the 80 amino acid Small Adipocyte Factor 1 (Smaf1) protein in adipocytes" (G. Ren, P. Eskandari, S. Wang, C.M. Smas, 2016) [1].

2.
Arch Biochem Biophys ; 590: 27-36, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26427354

RESUMO

The gene for Small Adipocyte Factor 1, Smaf1 (also known as adipogenin, ADIG), encodes a ∼600 base transcript that is highly upregulated during 3T3-L1 in vitro adipogenesis and markedly enriched in adipose tissues. Based on the lack of an obvious open reading frame in the Smaf1 transcript, it is not known if the Smaf1 gene is protein coding or non-coding RNA. Using a peptide from a putative open reading frame of Smaf1 as antigen, we generated antibodies for western analysis. Our studies prove that Smaf1 encodes an adipose-enriched protein which in western blot analysis migrates at ∼10 kDa. Rapid induction of Smaf1 protein occurs during in vitro adipogenesis and its expression in 3T3-L1 adipocytes is positively regulated by insulin and glucose. Moreover, siRNA studies reveal that expression of Smaf1 in adipocytes is wholly dependent on PPARγ. On the other hand, use of siRNA for Smaf1 to nearly abolish its protein expression in adipocytes revealed that Smaf1 does not have a major role in adipocyte triglyceride accumulation, lipolysis or insulin-stimulated pAkt induction. However, immunolocalization studies using HA-tagged Smaf1 reveal enrichment at adipocyte lipid droplets. Together our findings show that Smaf1 is a novel small protein endogenous to adipocytes and that Smaf1 expression is closely tied to PPARγ-mediated signals and the adipocyte phenotype.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Gotículas Lipídicas/metabolismo , Proteínas Nucleares/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Regulação da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peso Molecular , Proteínas Nucleares/química , PPAR gama/metabolismo
3.
J Clin Endocrinol Metab ; 100(10): E1299-307, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26204133

RESUMO

OBJECTIVE: Circulating ANGPTL8 has recently been used as a marker of insulin action. We studied expression and insulin regulation of ANGPTL8 and ANGPTL3 in vivo and in vitro. DESIGN AND METHODS: Expression of ANGPTL8 and ANGPTL3 was studied in 34 paired samples of human liver and adipose tissue. Effects of insulin on 1) plasma concentrations and adipose tissue expression of ANGPTL8 and ANGPTL3 (in vivo 6-h euglycemic hyperinsulinemia; n = 18), and 2) ANGPTL8 and ANGPTL3 gene and protein expression in immortalized human hepatocytes (IHH) and adipocytes were measured. Effect of ANGPTL3 on secretion of ANGPTL8 in cells stably overexpressing ANGPTL3, -8, or both was determined. RESULTS: ANGPTL3 was only expressed in the liver, whereas ANGPTL8 was expressed in both tissues. In vivo hyperinsulinemia significantly decreased both plasma ANGPTL8 and ANGPTL3 at 3 and 6 hours. Insulin increased ANGPTL8 expression in human adipose tissue 14- and 18-fold at 3 and 6 hours and ANGPTL8 was the most insulin-responsive transcript on microarray. Insulin also increased ANPGTL8 in cultured adipocytes and IHH but the protein mainly remained intracellular. In vitro in IHH, insulin decreased ANGPTL3 gene expression and secretion of ANGPTL3 into growth medium. Overexpression of ANGPTL8 in CHO cells did not result in its release into culture medium while abundant secretion occurred in cells co-expressing ANGPTL3 and -8. CONCLUSIONS: Insulin decreases plasma ANGPTL3 by decreasing ANGPTL3 expression in the liver. Insulin markedly increases ANGPTL8 in adipose tissue and the liver but not in plasma. These data show that measurement of plasma ANGPTL3 but not -8 reflects insulin action in target tissues.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Angiopoietinas/metabolismo , Hepatócitos/efeitos dos fármacos , Insulina/farmacologia , Fígado/efeitos dos fármacos , Hormônios Peptídicos/metabolismo , Tecido Adiposo/metabolismo , Proteína 3 Semelhante a Angiopoietina , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Angiopoietinas/genética , Animais , Células CHO , Linhagem Celular , Cricetulus , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Hormônios Peptídicos/genética
4.
J Biol Chem ; 289(17): 12029-12039, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24627478

RESUMO

In adipocytes, lipolysis is a highly regulated process involving hormonal signals, lipid droplet-associated proteins, and lipases. The discovery of new lipid droplet-associated proteins added complexity to the current model of lipolysis. In this study, we used cultured human adipocytes to demonstrate that fat-specific protein 27 (FSP27), an abundantly expressed protein in adipocytes, regulates both basal and stimulated lipolysis by interacting with adipose triglyceride lipase (ATGL, also called desnutrin or PNPLA2). We identified a core domain of FSP27, amino acids 120-220, that interacts with ATGL to inhibit its lipolytic function and promote triglyceride storage. We also defined the role of FSP27 in free fatty acid-induced insulin resistance in adipocytes. FSP27 depletion in human adipocytes increased lipolysis and inhibited insulin signaling by decreasing AKT phosphorylation. However, reducing lipolysis by either depletion of ATGL or expression of exogenous full-length FSP27 or amino acids 120-220 protected human adipocytes against the adverse effects of free fatty acids on insulin signaling. In embryonic fibroblasts derived from ATGL KO mice, exogenous free fatty acids did not affect insulin sensitivity. Our results demonstrate a crucial role for FSP27-ATGL interactions in regulating lipolysis, triglyceride accumulation, and insulin signaling in human adipocytes.


Assuntos
Adipócitos/enzimologia , Resistência à Insulina , Lipase/metabolismo , Lipólise/fisiologia , Proteínas/fisiologia , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Humanos , Insulina/metabolismo , Insulina/farmacologia , Camundongos , Camundongos Knockout , Fosforilação , Ligação Proteica , Proteínas/genética , Transdução de Sinais , Triglicerídeos/metabolismo
5.
PLoS One ; 8(6): e65531, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23799023

RESUMO

Differentiation of Embryonic Stem Cells 1 (Dies1) was recently identified as a novel type I immunoglobulin (IgG) domain-containing plasma membrane protein important for effective differentiation of a murine pluripotent embryonic stem cell line. In this setting, Dies1 enhances bone morphogenetic protein 4 (BMP4) signaling. Here we show Dies1 transcript expression is induced ∼225-fold during in vitro adipogenesis of 3T3-L1 murine preadipocytes. Immunocytochemical imaging using ectopic expression of Flag-tagged Dies1 in 3T3-L1 adipocytes revealed localization to the adipocyte plasma membrane. Modulation of adipocyte phenotype with with tumor necrosis factor-α (TNFα) treatment or by siRNA knockdown of the master pro-adipogenic transcription factor peroxisome proliferator activated receptor gamma (PPARγ) resulted in a 90% and 60% reduction of Dies1 transcript levels, respectively. Moreover, siRNA-mediated Dies1 knockdown in 3T3-L1 preadipocytes inhibited adipogenic conversion. Such cultures had a 35% decrease in lipid content and a 45%-65% reduction in expression of key adipocyte transcripts, including that for PPARγ. The standard protocol for full in vitro adipogenic conversion of committed preadipocytes, such as 3T3-L1, does not include BMP4 treatment. Thus we posit the positive role of Dies1 in adipogenesis, unlike that for Dies1 in differentiation of embryonic stem cells, does not include its pro-BMP4 effects. In support of this idea, 3T3-L1 adipocytes knocked down for Dies1 did not evidence decreased phospho-Smad1 levels upon BMP4 exposure. qPCR analysis of Dies1 transcript in multiple murine and human tissues reveals high enrichment in white adipose tissue (WAT). Interestingly, we observed a 10-fold induction of Dies1 transcript in WAT of fasted vs. fed mice, suggesting a role for Dies1 in nutritional response of mature fat cells in vivo. Together our data identify Dies1 as a new differentiation-dependent adipocyte plasma membrane protein whose expression is required for effective adipogenesis and that may also play a role in regard to nutritional status in WAT.


Assuntos
Adipócitos/citologia , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Imunoglobulinas/fisiologia , Proteínas de Membrana/fisiologia , Células 3T3-L1 , Animais , Sequência de Bases , Proteína Morfogenética Óssea 4/metabolismo , Linhagem Celular , Primers do DNA , Perfilação da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Interferente Pequeno , Transdução de Sinais
6.
Mol Endocrinol ; 27(1): 116-26, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23204327

RESUMO

Adipose triglyceride lipase (ATGL) is the key triacylglycerol hydrolase in adipocytes. The precise mechanisms by which ATGL action is regulated by lipid droplet (LD) coat proteins and responds to hormonal stimulation are incompletely defined. By combining usage of loss- and gain-of-function approaches, we sought to determine the respective roles of perilipin 1 and fat-specific protein 27 (FSP27) in the control of ATGL-mediated lipolysis in adipocytes. Knockdown of endogenous perilipin 1 expression resulted in elevated basal lipolysis that was less responsive to ß-adrenergic agonist isoproterenol. In comparison, depletion of FSP27 protein increased both basal and stimulated lipolysis with no significant impact on the overall response of cells to isoproterenol. In vitro assays showed that perilipin but not FSP27 was able to inhibit the triacylglycerol hydrolase activity of ATGL. Perilipin 1 also attenuated dose-dependent activation of ATGL by its Coactivator Comparative Gene identification-58. Accordingly, depletion of perilipin 1 and CGI-58 in adipocytes inversely affected basal lipolysis specifically mediated by overexpressed ATGL. Moreover, although depletion of perilipin 1 abolished the LD translocation of ATGL stimulated by isoproterenol, absence of FSP27 resulted in multilocularization of LDs along with increased LD presence of ATGL under both basal and stimulated conditions. Interestingly, knockdown of ATGL expression increased LD size and decreased LD number in FSP27-depeleted cells. Together, our results demonstrate that although FSP27 acts to constitutively limit the LD presence of ATGL, perilipin 1 plays an essential role in mediating the response of ATGL action to ß-adrenergic hormones.


Assuntos
Adipócitos/enzimologia , Proteínas de Transporte/genética , Lipase/metabolismo , Lipólise , Fosfoproteínas/genética , Proteínas/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferase/fisiologia , Células 3T3-L1 , Animais , Proteínas de Transporte/metabolismo , Ativação Enzimática , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Forma das Organelas , Organelas/enzimologia , Perilipina-1 , Fosfoproteínas/metabolismo , Transporte Proteico , Proteínas/metabolismo , RNA Interferente Pequeno/genética
7.
Am J Physiol Endocrinol Metab ; 303(3): E334-51, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22569073

RESUMO

To identify new genes that are important in fat metabolism, we utilized the Lexicon-Genentech knockout database of genes encoding transmembrane and secreted factors and whole murine genome transcriptional profiling data that we generated for 3T3-L1 in vitro adipogenesis. Cross-referencing null models evidencing metabolic phenotypes with genes induced in adipogenesis led to identification of a new gene, which we named RIFL (refeeding induced fat and liver). RIFL-null mice have serum triglyceride levels approximately one-third of wild type. RIFL transcript is induced >100-fold during 3T3-L1 adipogenesis and is also increased markedly during adipogenesis of murine and human primary preadipocytes. siRNA-mediated knockdown of RIFL during 3T3-L1 adipogenesis results in an ~35% decrease in adipocyte triglyceride content. Murine RIFL transcript is highly enriched in white and brown adipose tissue and liver. Fractionation of WAT reveals that RIFL transcript is exclusive to adipocytes with a lack of expression in stromal-vascular cells. Nutritional and hormonal studies are consistent with a prolipogenic function for RIFL. There is evidence of an approximately eightfold increase in RIFL transcript level in WAT in ob/ob mice compared with wild-type mice. RIFL transcript level in WAT and liver is increased ~80- and 12-fold, respectively, following refeeding of fasted mice. Treatment of 3T3-L1 adipocytes with insulin increases RIFL transcript ≤35-fold, whereas agents that stimulate lipolysis downregulate RIFL. Interestingly, the 198-amino acid RIFL protein is predicted to be secreted and shows ~30% overall conservation with the NH(2)-terminal half of angiopoietin-like 3, a liver-secreted protein that impacts lipid metabolism. In summary, our data suggest that RIFL is an important new regulator of lipid metabolism.


Assuntos
Adipócitos/efeitos dos fármacos , Insulina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Células 3T3-L1 , Adipócitos/metabolismo , Adipócitos/fisiologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Clonagem Molecular , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Genes/fisiologia , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , RNA Interferente Pequeno/farmacologia
8.
J Cell Biochem ; 111(1): 195-206, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20503194

RESUMO

The ubiquitously expressed basic helix-loop-helix (bHLH) transcription factors E12 and E47, products of alternative splicing of the E2A/TCF3 gene, regulate diverse biological processes including cell growth, differentiation and development. To search for novel protein interactions for E12, we utilized the bHLH domain of E12 as a bait in yeast two-hybrid screening. Yeast two-hybrid, mammalian two-hybrid, and co-immunoprecipitation analyses demonstrate specific interaction of E12 with RANBP17, a novel member of the importin-beta superfamily; this interaction maps to the CRM1 homology region of RANBP17. Ectopic expression of RANBP17 leads to a approximately 3-fold increase in E2A/MyoD mediated transactivation of an E-box regulated luciferase reporter gene. Interaction and transactivation studies also revealed similar functions for RANBP16/XPO7. Furthermore, ectopic expression of either RANBP16 or RANBP17 resulted in increased level of endogenous transcript for the cyclin-dependent kinase inhibitor, p21(Waf1/Cip1), a well-characterized E2A target gene. Together, these biochemical and functional data reveal RANBP16 and RANBP17 as novel regulators of E2A protein action.


Assuntos
Carioferinas/metabolismo , Fator 3 de Transcrição/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Animais , Células COS , Chlorocebus aethiops , Genes Reporter , Células HeLa , Humanos , Carioferinas/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Fator 3 de Transcrição/genética , Transcrição Gênica , Ativação Transcricional , Técnicas do Sistema de Duplo-Híbrido , Proteína ran de Ligação ao GTP/genética
9.
Diabetes ; 59(4): 775-81, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20068142

RESUMO

OBJECTIVE: In metazoans, target of rapamycin complex 1 (TORC1) plays the key role in nutrient- and hormone-dependent control of metabolism. However, the role of TORC1 in regulation of triglyceride storage and metabolism remains largely unknown. RESEARCH DESIGN AND METHODS: In this study, we analyzed the effect of activation and inhibition of the mammalian TORC1 (mTORC1) signaling pathway on the expression of adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), lipolysis, lipogenesis, and lipid storage in different mammalian cells. RESULTS: Activation of mTORC1 signaling in 3T3-L1 adipocytes by ectopic expression of Rheb inhibits expression of ATGL and HSL at the level of transcription, suppresses lipolysis, increases de novo lipogenesis, and promotes intracellular accumulation of triglycerides. Inhibition of mTORC1 signaling by rapamycin or by knockdown of raptor stimulates lipolysis primarily via activation of ATGL expression. Analogous results have been obtained in C2C12 myoblasts and mouse embryonic fibroblasts with genetic ablation of tuberous sclerosis 2 (TSC2) gene. Overexpression of ATGL in these cells antagonized the lipogenic effect of TSC2 knockout. CONCLUSIONS: Our findings demonstrate that mTORC1 promotes fat storage in mammalian cells by suppression of lipolysis and stimulation of de novo lipogenesis.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo/anatomia & histologia , Lipólise/fisiologia , Fatores de Transcrição/fisiologia , Células 3T3 , Adipócitos/citologia , Animais , Compostos Azo , Sequência de Bases , Diferenciação Celular , Células Cultivadas/enzimologia , Células Cultivadas/fisiologia , Genes Reporter , Lipase/genética , Camundongos , Camundongos Knockout , Consumo de Oxigênio , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , RNA Interferente Pequeno/genética , Fatores de Transcrição/genética , Transfecção , Triglicerídeos/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
10.
Mech Dev ; 127(3-4): 183-202, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20043993

RESUMO

Obesity is a public health crisis in the United States. Targeting preadipocyte to adipocyte conversion may be an effective approach to regulate adipose mass. Using differential screening we identified Fstl1, a secreted glycoprotein with roles in immunomodulation, cell growth, cardioprotection, and vascularization, as a "preadipokine". Fstl1 is highly expressed in 3T3-L1 preadipocytes and dramatically downregulated early in their differentiation to adipocytes. Northern blot analysis of murine tissues reveals white adipose tissue (WAT), lung and heart as primary sites of Fstl1 transcript expression. In WAT, Fstl1 transcript is restricted to the preadipocyte-containing stromal-vascular cell population. Time course studies in multiple adipogenesis models reveal downregulation of Fstl1 is a hallmark of white and brown adipocyte conversion. By Western blot, we show culture media of 3T3-L1 preadipocytes contains high levels of Fstl1 protein that rapidly decline in adipocyte conversion. Moreover, we observe a correlation between preadipocyte phenotype and Fstl1 expression in that TNFalpha-mediated de-differentiation of 3T3-L1 adipocytes is accompanied by re-expression of Fstl1 transcript and protein. Treatment of 3T3-L1 preadipocytes with a panel of 18 hormones and other agents revealed the demethylating agent 5-aza-cytidine decreases Fstl1 transcript and protein levels by approximately 90%. Furthermore, of 10 additional preadipocyte-expressed genes analyzed we find Pref-1, Col1A1, Sca-1/Ly6a, Lox and Thbs2, are also downregulated by 5-aza-cytidine. Using luciferase reporter constructs containing 791 or 3922 bp of the Fstl1 5' flanking region, we determine negative transcriptional regulation by Kruppel-like factor 15. Together, our data suggest downregulation of Fstl1 expression may be an important feature of preadipocyte to adipocyte conversion.


Assuntos
Adipócitos/citologia , Diferenciação Celular/fisiologia , Regulação para Baixo , Proteínas Relacionadas à Folistatina/genética , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Northern Blotting , Western Blotting , Proteínas Relacionadas à Folistatina/metabolismo , Proteínas Relacionadas à Folistatina/fisiologia , Camundongos , RNA Mensageiro/genética
11.
Am J Physiol Endocrinol Metab ; 297(6): E1395-413, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19843876

RESUMO

The adipocyte-specific protein FSP27, also known as CIDEC, is one of three cell death-inducing DFF45-like effector (CIDE) proteins. The first known function for CIDEs was promotion of apoptosis upon ectopic expression in mammalian cells. Recent studies in endogenous settings demonstrated key roles for CIDEs in energy metabolism. FSP27 is a lipid droplet-associated protein whose heterologous expression enhances formation of enlarged lipid droplets and is required for unilocular lipid droplets typical of white adipocytes in vivo. Here, we delineate relationships between apoptotic function and lipid droplet localization of FSP27. We demonstrate that ectopic expression of FSP27 induces enlarged lipid droplets in multiple human cell lines, which is indicative that its mechanism involves ubiquitously present, rather than adipocyte-specific, cellular machinery. Furthermore, promotion of lipid droplet formation in HeLa cells via culture in exogenous oleic acid offsets FSP27-mediated apoptosis. Using transient cotransfections and analysis of lipid droplets in HeLa cells stably expressing FSP27, we show that FSP27 does not protect lipid droplets from action of ATGL lipase. Domain mapping with eGFP-FSP27 deletion constructs indicates that lipid droplet localization of FSP27 requires amino acids 174-192 of its CIDE C domain. The apoptotic mechanism of FSP27, which we show involves caspase-9 and mitochondrial cytochrome c, also requires this 19-amino acid region. Interaction assays determine the FSP27 CIDE C domain complexes with CIDEA, and Western blot reveals that FSP27 protein levels are reduced by coexpression of CIDEA. Overall, our findings demonstrate the function of the FSP27 CIDE C domain and/or regions thereof for apoptosis, lipid droplet localization, and CIDEA interaction.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Caspases/metabolismo , Metabolismo dos Lipídeos/fisiologia , Animais , Western Blotting , Células COS , Linhagem Celular , Chlorocebus aethiops , Citocromos c/metabolismo , Fragmentação do DNA , Dimerização , Metabolismo Energético , Células HeLa , Humanos , Imuno-Histoquímica , Lipase/biossíntese , Lipase/metabolismo , Relação Estrutura-Atividade , Técnicas do Sistema de Duplo-Híbrido
12.
BMC Res Notes ; 1: 85, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18803820

RESUMO

BACKGROUND: White adipose tissue is not only an energy storage organ; it also functions as an endocrine organ. The coordination and integration of numerous gene expression events is required to establish and maintain the adipocyte phenotype. FINDINGS: We previously observed a 45-fold upregulation for a transcript encoding a novel predicted transmembrane protein, Tmem182, upon brown preadipocyte to adipocyte conversion. Here we use real-time PCR analysis to further characterize Tmem182 transcript expression in the adipocyte lineage. Analysis across a panel of 10 murine tissues revealed highest Tmem182 transcript expression in white adipose tissues (WAT), with 10-fold to 20-fold higher levels than in brown adipose tissue (BAT). Tmem182 transcript expression is ~3-fold upregulated in BAT of genetically obese (ob/ob) mice vs. wild type C57BL/6. Analysis of three in vitro models of white adipogenesis indicates markedly enriched expression of Tmem182 transcript in adipocytes vs. preadipocytes. Compared to 3T3-L1 preadipocytes, a 157-fold higher level of Tmem182 transcript is detected at 3 day post-induction of adipogenesis and an ~2500-fold higher level in mature 3T3-L1 adipocytes. TNFalpha treatment of 3T3-L1 adipocytes resulted in a ~90% decrease in Tmem182 transcript level. As skeletal muscle and heart were also found to express Tmem182 transcript, we assessed expression in C2C12 myogenesis and observed a ~770-fold upregulation upon conversion of myoblasts to myocytes. CONCLUSION: WAT is the most prominent site of Tmem182 transcript expression and levels of transcript for Tmem182 are altered in adipose tissues of ob/ob mice and upon exposure of 3T3-L1 adipocytes to the proinflammatory cytokine TNFalpha. The dramatic upregulation of Tmem182 transcript during in vitro adipogenesis and myogenesis suggests Tmem182 may function in intracellular pathways important in these two cell types.

13.
BMC Genomics ; 9: 397, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18721461

RESUMO

BACKGROUND: The co-morbidities of obesity are tied to location of excess fat in the intra-abdominal as compared to subcutaneous white adipose tissue (WAT) depot. Genes distinctly expressed in WAT depots may impart depot-dependent physiological functions. To identify such genes, we prepared subtractive cDNA libraries from murine subcutaneous (SC) or intra-abdominal epididymal (EP) white adipocytes. RESULTS: Differential screening and qPCR validation identified 7 transcripts with 2.5-fold or greater enrichment in EP vs. SC adipocytes. Boc, a component of the hedgehog signaling pathway demonstrated highest enrichment (approximately 12-fold) in EP adipocytes. We also identified a dramatic enrichment in SC adipocytes vs. EP adipocytes and in SC WAT vs. EP WAT for transcript(s) for the major urinary proteins (Mups), small secreted proteins with pheromone functions that are members of the lipocalin family. Expression of Boc and Mup transcript was further assessed in murine tissues, adipogenesis models, and obesity. qPCR analysis reveals that EP WAT is a major site of expression of Boc transcript. Furthermore, Boc transcript expression decreased in obese EP WAT with a concomitant upregulation of Boc transcript in the obese SC WAT depot. Assessment of the Boc binding partner Cdon in adipose tissue and cell fractions thereof, revealed transcript expression similar to Boc; suggestive of a role for the Boc-Cdon axis in WAT depot function. Mup transcripts were predominantly expressed in liver and in the SC and RP WAT depots and increased several thousand-fold during differentiation of primary murine preadipocytes to adipocytes. Mup transcripts were also markedly reduced in SC WAT and liver of ob/ob genetically obese mice compared to wild type. CONCLUSION: Further assessment of WAT depot-enriched transcripts may uncover distinctions in WAT depot gene expression that illuminate the physiological impact of regional adiposity.


Assuntos
Perfilação da Expressão Gênica , Gordura Intra-Abdominal/metabolismo , Obesidade/genética , Gordura Subcutânea/metabolismo , Células 3T3-L1 , Animais , Moléculas de Adesão Celular/genética , Epididimo/citologia , Epididimo/metabolismo , Expressão Gênica , Biblioteca Gênica , Imunoglobulina G/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Hibridização de Ácido Nucleico , Proteínas/genética , RNA/genética , Receptores de Superfície Celular/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Am J Physiol Endocrinol Metab ; 295(1): E205-15, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18492766

RESUMO

White adipose tissue functions in energy storage and as an endocrine organ. DNA microarray analysis led us to identify Wdnm1-like, a distant member of the whey acidic protein/four-disulfide core (WAP/4-DSC) family, as a differentiation-dependent gene in white and brown adipogenesis. Wdnm1-like is a novel 6.8-kDa protein, and Western blot analysis reveals secretion into culture media. Wdnm1-like transcript is selectively expressed in adipose tissue and liver and is enriched approximately 500-fold in white adipose depots vs. brown. Cellular fractionation of WAT demonstrates Wdnm1-like transcript expression is restricted to the adipocyte population. Studies in 3T3-L1 preadipocytes, an in vitro model of white adipogenesis, indicate Wdnm1-like transcript increases within 6 h of adipogenic induction with an approximately 17,000-fold increase by day 7. Dramatic upregulation of Wdnm1-like also accompanies white adipogenesis of ScAP-23 preadipocytes and primary preadipocytes. TNF-alpha treatment of 3T3-L1 adipocytes increased Wdnm1-like transcript level 2.4-fold and was attenuated by pretreatment with the p38 MAP kinase inhibitor SB203580. A number of WAP/4-DSC family proteins function as protease inhibitors. This, taken with the role of extracellular remodeling in adipogenesis, led us to address effects of Wdnm1-like on matrix metalloproteinase (MMP) activity. Gelatin zymography of HT1080 fibrosarcoma cells transfected with a Wdnm1-like expression construct revealed markedly increased levels of active MMP-2. Our findings identify a new member of the adipocyte "secretome" that functions to enhance MMP-2 activity. We postulate that Wdnm1-like may play roles in remodeling of the extracellular milieu in adipogenesis, as well as in tumor microenvironments where adipocytes are key stromal components.


Assuntos
Adipocinas/metabolismo , Tecido Adiposo Branco/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Células 3T3-L1 , Adipócitos Marrons/enzimologia , Adipócitos Marrons/metabolismo , Adipogenia/fisiologia , Adipocinas/biossíntese , Adipocinas/genética , Tecido Adiposo Branco/enzimologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Ativação Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Transcrição Gênica , Transfecção
15.
Am J Physiol Endocrinol Metab ; 294(4): E654-67, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18198355

RESUMO

Fat-specific protein 27 (FSP27)/CIDEC was initially identified by its upregulation in TA1 adipogenesis and is one of three cell death-inducing DFF45-like effector (CIDE) family proapoptotic proteins. Ectopic expression of CIDEs promotes apoptosis of mammalian cells. On the other hand, FSP27 has very recently been illustrated to regulate lipid droplet size and promote lipid storage in adipocytes. Regulation of endogenous FSP27 expression is unknown. We assessed the FSP27 transcript level in the well-characterized 3T3-L1 in vitro adipocyte differentiation model and found its emergence parallels the adipocyte-enriched transcript adipocyte fatty acid binding protein and stearoyl Co-A desaturase 1. Furthermore, FSP27 is a differentiation-dependent transcript in adipogenesis of primary rodent and human preadipocytes and in brown adipogenesis. The FSP27 transcript is inversely regulated by TNF-alpha and insulin, consistent with an antilipolytic function. It is nearly abolished with a 4-h exposure of 3T3-L1 adipocytes to 10 ng/ml TNF-alpha, while treatment with 100 nM insulin increased the FSP27 transcript eightfold. In the latter case LY-294002 blocked this response, indicating involvement of phosphatidylinositol 3-kinase signals. Northern blot analysis of murine tissues indicated exclusive expression of FSP27 in white and brown adipose tissue; however, a dramatic upregulation occurred in the liver of ob/ob mice. Ectopic expression of murine FSP27 in 293T cells and in 3T3-L1 preadipocytes led to the appearance of key apoptotic hallmarks and cell death. However, despite the upregulation for FSP27 in adipogenesis, we failed to detect DNA laddering indicative of apoptosis in 3T3-L1 adipocytes. This suggests that adipogenesis is accompanied by decreased susceptibility to the proapoptotic effects of FSP27. Overall, our findings support roles for FSP27 in cell death and in adipocyte function.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Apoptose/fisiologia , Metabolismo Energético/fisiologia , Proteínas/metabolismo , Células 3T3-L1 , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Insulina/farmacologia , Rim/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteínas dos Microfilamentos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
16.
Physiol Genomics ; 31(2): 328-42, 2007 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-17609412

RESUMO

The 3T3-L1 model of in vitro adipogenesis has provided key insights into the molecular nature of this process. However, given that 3T3-L1 are of an embryonic origin, it is not clear to what extent they represent adipogenesis as it occurs in white adipose tissue (WAT). With the goal of better defining preadipocytes and adipogenesis in WAT, we have generated a new cell culture model from adipocyte precursors present in C57BL/6 mouse subcutaneous WAT. ScAP-23 preadipocytes show fibroblastic morphology, and on treatment with dexamethasone, 3-methylisobutylxanthine, insulin, and indomethacin, convert to nearly 100% adipocyte morphology. ScAP-23 adipocytes contain abundant lipid droplets and express transcripts for PPAR gamma, C/EBP family, and SREBP-1c transcription factors, SCD1, aFABP, ATGL, GLUT4, FAS, LDL, and GPDH, and are insulin responsive. Differential screening of 1,176 genes using nylon DNA arrays identified 10 transcripts enriched in ScAP-23 adipocytes vs. preadipocytes and 26 transcripts enriched in ScAP-23 preadipocytes vs. adipocytes. Semiquantitative or real-time PCR analyses identified a common cohort of 14 transcripts markedly downregulated in both ScAP-23 and 3T3-L1 adipogenesis. These included catenin-beta1, chemokine ligand-2, serine or cysteine peptidase inhibitor f1, aurora kinase B, thrombospondin2, and solute carrier-7a5. Five of these transcripts (Ccl2, Serpinf1, Aurkb, Thbs2, and Slc7a5) demonstrated at least a twofold increase in WAT from obese (ob/ob) mice compared with that of wild-type mice. This suggests that comparative gene expression studies of ScAP-23 and 3T3-L1 adipogenesis may be particularly fruitful in identifying preadipocyte-expressed genes that play a role in adipose tissue physiology and/or pathophysiology.


Assuntos
Adipócitos/citologia , Adipogenia/genética , Linhagem Celular , Perfilação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Gordura Subcutânea/citologia , 1-Metil-3-Isobutilxantina/farmacologia , Células 3T3-L1/metabolismo , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Dexametasona/farmacologia , Indometacina/farmacologia , Insulina/farmacologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/genética , Obesidade/metabolismo , Receptor de Insulina/análise , Transcrição Gênica
17.
Arch Biochem Biophys ; 464(1): 100-11, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17559793

RESUMO

We have determined that adipocytes are a major site of expression of the transcript for the novel alcohol dehydrogenase (ADH), Adhfe1. Adhfe1 is unique in that the sequence of its encoded protein places it among the iron-activated ADHs. Western blot analysis reveals Adhfe1 encodes a 50 kDa protein and immunocytochemical staining indicates mitochondrial localization. Adhfe1 transcript exhibits differentiation-dependent expression during in vitro brown and white adipogenesis. Unlike many adipocyte-enriched genes, however, Adhfe1 transcript expression in adipocytes is refractory to TNFalpha-mediated downregulation. However, use of pharmacological inhibitors reveals PI 3-kinase-mediated signals maintain the basal level of Adhfe1 transcript in 3T3-L1 adipocytes. Tissue profiling studies show Adhfe1 transcript is restricted to white and brown adipose tissues, liver, and kidney. In comparison to C57BL/6 mice, Adhfe1 transcript is downregulated 40% in white adipose tissue of ob/ob obese mice. Further characterization of Adhfe1 should yield new insights into adipocyte function and energy metabolism.


Assuntos
Álcool Desidrogenase/fisiologia , Oxirredutases do Álcool/biossíntese , Oxirredutases do Álcool/fisiologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Álcool Desidrogenase/metabolismo , Oxirredutases do Álcool/genética , Sequência de Aminoácidos , Animais , Células COS , Diferenciação Celular , Chlorocebus aethiops , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteínas Mitocondriais , Modelos Biológicos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Fator de Necrose Tumoral alfa/metabolismo
18.
Am J Physiol Endocrinol Metab ; 293(1): E110-20, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17356011

RESUMO

A marked degree of macrophage infiltration of white adipose tissue (WAT) occurs in obesity and may link excess adiposity with the chronic inflammatory state underlying metabolic syndrome and other comorbidities of obesity. Excess deposition of fat in the intra-abdominal vs. subcutaneous WAT depots is a key component of metabolic syndrome. Through construction and differential screening of a murine ob/ob WAT cDNA library, we identified Slc37a2, a novel sugar transporter of the major facilitator superfamily, to be twofold enriched in intra-abdominal vs. subcutaneous fat. We find Slc37a2 is a macrophage-enriched transcript. In murine tissues, Slc37a2 transcript is restricted to spleen, thymus, and obese WAT. It is also readily detected in the RAW264.7 macrophage cell line and increases 46-fold during macrophage differentiation of THP-1 human monocytes. Compared with wild-type mice, Slc37a2 transcript is increased epididymal ninefold in ob/ob WAT and assessment of expression of the macrophage marker emr1 indicated upregulation of Slc37a2 transcript in macrophages populating ob/ob WAT. Studies with PNGase F and tunicamycin reveal the Slc37a2 protein is posttranslationally modified by addition of N-linked glycans. Slc37a2 protein migrates as heterogeneous species of approximately 50-75 kDa and its ectopic expression in mammalian cells results in the appearance of large intracellular vacuoles. We postulate that the function of this macrophage-specific putative sugar transporter is central to the metabolism of the macrophage population specifically present in obese WAT.


Assuntos
Tecido Adiposo Branco/metabolismo , Antiporters/genética , Antiporters/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Obesidade/genética , Sequência de Aminoácidos , Animais , Células COS , Quimiotaxia de Leucócito , Chlorocebus aethiops , Células HeLa , Humanos , Gordura Intra-Abdominal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Dados de Sequência Molecular , Obesidade/metabolismo , Especificidade de Órgãos , Transfecção
19.
Am J Physiol Endocrinol Metab ; 291(1): E115-27, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16705060

RESUMO

The minimal adipose phenotype of hormone-sensitive lipase (HSL)-null mice suggested that other hormonally responsive lipase(s) were present in adipocytes. Recent studies have characterized a new adipose tissue triglyceride lipase, ATGL/PNPLA2/destnutrin/iPLA2zeta/TTS2.2 (ATGL). We had previously cloned a novel adipose-enriched transcript by differential screening and recently determined its identity with murine ATGL. We report here on the regulation of ATGL by TNF-alpha and insulin in 3T3-L1 adipocytes and identify ATGL as a target for transcriptional activation by the key adipogenic transcription factor PPARgamma. Insulin at 100 nM resulted in a marked decrease in ATGL transcript that was effectively blocked by inhibitors for PI 3-kinase and p70 ribosomal protein S6 kinase. TNF-alpha treatment decreased ATGL transcript in a time-dependent manner that paralleled TNF-alpha downregulation of PPARgamma with a maximal decrease noted by 6 h. TNF-alpha effects on ATGL were attenuated by pretreatment with PD-98059, LY-294002, or rapamycin, suggesting involvement of the p44/42 MAP kinase, PI 3-kinase, and p70 ribosomal protein S6 kinase signals. To study transcriptional regulation of ATGL, we cloned 2,979 bp of the murine ATGL 5'-flanking region. Compared with promoterless pGL2-Basic, the -2979/+21 ATGL luciferase construct demonstrated 120- and 40-fold increases in activity in white and brown adipocytes, respectively. Luciferase reporter activities for a series of eight ATGL promoter deletions revealed that the -928/+21, -1738/+21, -1979/+21, and -2979/+21 constructs were transactivated by PPARgamma. Our findings identify the novel lipase ATGL to be a target gene for TNF-alpha and insulin action in adipocytes and reveal that it is subject to transcriptional control by PPARgamma-mediated signals.


Assuntos
Tecido Adiposo/enzimologia , Insulina/farmacologia , Lipase/metabolismo , PPAR gama/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Células 3T3-L1 , Adipócitos/enzimologia , Adipócitos/fisiologia , Tecido Adiposo/fisiologia , Animais , Northern Blotting , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Lipase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Obesos , Microscopia de Fluorescência , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
20.
Biochem Biophys Res Commun ; 326(1): 36-44, 2005 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-15567149

RESUMO

With the aim of identifying novel molecular pathways in the adipocyte, we conducted differential screening of DNA filter arrays with probes from 3T3-L1 preadipocytes and adipocytes, and discovered a novel 0.7kb transcript we term small adipocyte factor 1 (SMAF1). SMAF1 encodes a wholly novel 10kDa protein. Transfection and localization studies of a SMAF1-EGFP fusion construct indicate nuclear localization, suggestive of a possible regulatory role. Northern blot analysis of various murine tissues indicates adipose tissue-restricted expression, and fractionation of adipose tissue reveals that SMAF1 is expressed soley in adipocytes and not in the stromal-vascular cell population. Northern blot analysis of brown and white adipogenic conversion reveals that expression of SMAF1 closely parallels emergence of an adipocyte phenotype and that TNFalpha-mediated dedifferentiation of 3T3-L1 adipocytes results in a rapid decline of SMAF1 transcript. These data indicate that SMAF1 is closely tied to the adipocyte phenotype and predict a novel and possibly regulatory role for this gene in adipocyte function.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Diferenciação Celular/efeitos dos fármacos , Clonagem Molecular , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Proteínas Nucleares/genética , Especificidade de Órgãos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...