Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 11(9): e0163318, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27685785

RESUMO

Bisphenol S (BPS) is increasingly used as a replacement plasticizer for bisphenol A (BPA) but its effects on human health have not been thoroughly examined. Recent evidence indicates that both BPA and BPS induce adipogenesis, although the mechanisms leading to this effect are unclear. In an effort to identify common and distinct mechanisms of action in inducing adipogenesis, transcriptional profiles of differentiating human preadipocytes exposed to BPA or BPS were compared. Human subcutaneous primary preadipocytes were differentiated in the presence of either 25 µM BPA or BPS for 2 and 4 days. Poly-A RNA-sequencing was used to identify differentially expressed genes (DEGs). Functional analysis of DEGs was undertaken in Ingenuity Pathway Analysis. BPA-treatment resulted in 472 and 176 DEGs on days 2 and 4, respectively, affecting pathways such as liver X receptor (LXR)/retinoid X receptor (RXR) activation, hepatic fibrosis and cholestasis. BPS-treatment resulted in 195 and 51 DEGs on days 2 and 4, respectively, revealing enrichment of genes associated with adipogenesis and lipid metabolism including the adipogenesis pathway and cholesterol biosynthesis. Interestingly, the transcription repressor N-CoR was identified as a negative upstream regulator in both BPA- and BPS-treated cells. This study presents the first comparison of BPA- and BPS-induced transcriptional profiles in human differentiating preadipocytes. While we previously showed that BPA and BPS both induce adipogenesis, the results from this study show that BPS affects adipose specific transcriptional changes earlier than BPA, and alters the expression of genes specifically related to adipogenesis and lipid metabolism. The findings provide insight into potential BPS and BPA-mediated mechanisms of action in inducing adipogenesis in human primary preadipocytes.

2.
Endocrinology ; 157(4): 1397-407, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27003841

RESUMO

Human exposure to bisphenol A has been associated with negative health outcomes in humans and its use is now regulated in a number of countries. Bisphenol S (BPS) is increasingly used as a replacement for bisphenol A; however, its effects on cellular metabolism and potential role as an endocrine disruptor have not been fully characterized. In the current study, we evaluated the effect of BPS on adipogenesis in primary human preadipocytes. The effect of BPS on the differentiation of human preadipocytes was determined after treatment with BPS at concentrations ranging from 0.1 nM to 25 µM by quantifying lipid accumulation and mRNA and protein levels of key adipogenic markers. Treatment of preadipocytes with 25 µM BPS induced lipid accumulation and increased the mRNA and protein levels of several adipogenic markers including lipoprotein lipase and adipocyte protein 2 (aP2). Cotreatment of cells with the estrogen receptor antagonist ICI-182,780 significantly inhibited BPS-induced lipid accumulation and affected aP2 but not lipoprotein lipase protein levels. Cotreatment of cells with the glucocorticoid receptor antagonist RU486 had no effect on BPS-induced lipid accumulation or protein levels. Furthermore, reporter gene assays using a synthetic promoter containing peroxisome proliferator-activated receptor-γ (PPARG)-response elements and a PPARG-responsive human aP2 promoter region showed that BPS was able to activate PPARG. To our knowledge, this study is the first to show that BPS induces lipid accumulation and differentiation of primary human preadipocytes, and this effect may be mediated through a PPARG pathway.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Fenóis/farmacologia , Sulfonas/farmacologia , Adipócitos/citologia , Adipócitos/metabolismo , Adulto , Western Blotting , Diferenciação Celular/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas do Receptor de Estrogênio/farmacologia , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Fulvestranto , Expressão Gênica/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Microscopia de Fluorescência , PPAR gama/genética , PPAR gama/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
4.
Environ Health Perspect ; 123(12): 1287-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26018136

RESUMO

BACKGROUND: Exposure to common environmental substances, such as bisphenol A (BPA), has been associated with a number of negative health outcomes. In vivo, BPA is rapidly converted to its predominant metabolite, BPA-glucuronide (BPA-G), which has long been believed to be biologically inactive because it lacks estrogenic activity. However, the effects of BPA-G on cellular metabolism have not been characterized. In the present study we examined the effect of BPA-G on adipogenesis. METHODS: The effect of BPA-G on the differentiation of human and 3T3L1 murine preadipocytes was evaluated in vitro by quantifying lipid accumulation and the expression of adipogenic markers. RESULTS: Treatment of 3T3L1 preadipocytes with 10 µM BPA-G induced a significant increase in lipid accumulation, mRNA expression of the adipogenic markers sterol regulatory element binding factor 1 (SREBF1) and lipoprotein lipase (LPL), and protein levels of LPL, aP2, and adipsin. Treatment of primary human preadipocytes with BPA-G also induced adipogenesis as determined by aP2 levels. Co-treatment of cells with the estrogen receptor (ER) antagonist fulvestrant (ICI) significantly inhibited the BPA-G-induced increase in LPL and aP2 levels, whereas treatment with ICI alone had no effect. Moreover, BPA-G did not display any significant estrogenic activity. CONCLUSIONS: To our knowledge, this study is the first to report that BPA-G induces adipocyte differentiation and is not simply an inactive metabolite. The fact that BPA-G induced adipogenesis and was inhibited by an ER antagonist yet showed no estrogenic activity suggests that it has no classical ER transcriptional activation function and acts through a pathway that remains to be determined.


Assuntos
Adipócitos/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Glucuronídeos/farmacologia , Fenóis/farmacologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas do Receptor de Estrogênio/farmacologia , Fulvestranto , Regulação da Expressão Gênica , Humanos , Metabolismo dos Lipídeos , Camundongos , RNA Mensageiro/metabolismo
5.
Adipocyte ; 3(3): 170-9, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25068083

RESUMO

Environmental pollutants, such as bisphenol A (BPA), have the potential to affect the differentiation processes and the biology of the adipose tissue. The 3T3-L1 model is one of the murine cell models used extensively for the investigation of the molecular events that govern the differentiation of adipocytes from a committed preadipocyte to a mature, lipid laden adipocyte. Most of the studies investigating the effects of BPA on preadipocyte differentiation have investigated the effects of this chemical in the presence of an optimal differentiation cocktail containing high concentrations of the synthetic glucocorticoid dexamethasone, conditions that result in 90% to 100% of differentiated adipocytes. Our studies employed the 3T3-L1 cell model in the absence of exogenous glucocorticoids. We show that BPA is able to increase the differentiation of the 3T3-L1 cells under these conditions. Furthermore, the effect of BPA was observed in the absence of the synthetic glucocorticoid (dexamethasone), a hormone known to be required for the differentiation of the 3T3-L1 cells. In addition, BPA upregulated the mRNA expression and protein levels of the terminal marker of adipogenesis the fatty acid binding protein (aP2) in these cells. Interestingly, the known modulators of adipogenesis such as the peroxisome proliferator-activated receptor (PPAR) γ or CCAAT enhancer binding protein (C/EBP) α were not elevated at the mRNA or protein level in response to BPA. Furthermore, BPA upregulated the expression levels of the marker of adipogenesis aP2, through an effect on the transcriptional activity of C/EBPδ and the glucocorticoid receptor (GR) at its promoter.

6.
Obesity (Silver Spring) ; 22(11): 2333-43, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25047013

RESUMO

OBJECTIVE: Exposure to the endocrine-disrupting chemical bisphenol A (BPA) is correlated with obesity and adipogenesis of human preadipocytes. However, the mechanism of action of BPA-induced human adipogenesis remains to be determined. METHODS: Primary human preadipocytes were differentiated in the presence of 50 µM BPA or 1 µM dexamethasone (DEX) for 48 hours. Potential mechanisms of BPA-induced adipogenesis were evaluated using gene expression microarray analysis. RESULTS: Microarray analysis revealed 373 differentially expressed genes following BPA treatment, including upregulation of sterol regulatory element binding factor 1 (SREBF1), a key regulator of lipid metabolism. For DEX-treated preadipocytes, 2167 genes were differentially expressed, including upregulation of the adipogenic marker lipoprotein lipase. Ingenuity Pathway Analysis was used to identify functional annotations of the gene expression changes associated with response to BPA and DEX. BPA exposure was associated with expression changes in the genes involved in triacylglycerol accumulation while DEX was linked to triacylglycerol and fatty acid metabolism. The analysis also revealed enrichment of genes following BPA exposure in the thyroid-receptor/retinoic X receptor (TR/RXR) and mammalian target of rapamycin (mTOR) signaling pathways. CONCLUSIONS: Our data suggest that potential mechanisms of action of BPA-induced adipogenesis involve SREBF1, the TR/RXR, and the mTOR pathways.


Assuntos
Adipócitos/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Fenóis/farmacologia , Adipócitos/fisiologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Diferenciação Celular/genética , Células Cultivadas , Dexametasona/farmacologia , Humanos , Metabolismo dos Lipídeos/genética , Lipase Lipoproteica/metabolismo , Análise em Microsséries , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Transdução de Sinais/genética , Triglicerídeos/metabolismo
7.
Viral Immunol ; 23(1): 17-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20121399

RESUMO

Studies from our laboratory and those of others have implicated lipopolysaccharide (LPS)-induced MAPK signaling as an important pathway in the regulation of cytokine expression. In this article, the regulation of IL-12 expression in two different human myeloid cell populations was evaluated. In primary monocytes, the inhibition of p38 enhanced IL-12 production, whereas it downregulated IL-12 production in THP-1 cells. The role of MAPK signaling in transcription factor binding to the IL-12p40 promoter was subsequently determined. In primary monocytes, ERK and p38 inhibition increased binding of AP-1 and Sp1, respectively, to the IL-12p40 promoter, while JNK inhibition increased NF-kappaB, AP-1, and Sp1 binding. In THP-1 cells, p38, ERK, and JNK inhibition increased NF-kappaB and Sp1 binding to the IL-12p40 promoter, while inhibiting AP-1 binding. In monocytes, mutations in the NF-kappaB, AP-1, Sp1, or Ets-2 binding sites resulted in complete inhibition of LPS-stimulated IL-12p40 promoter activity using a luciferase-based assay. In contrast, promoter activity was abrogated in THP-1 cells only when the Sp1 or Ets-2 binding sites were mutated. Transcription factor binding to the IL-12p40 promoter following in-vitro HIV infection demonstrated several differences between monocytes and THP-1 cells. Infection with HIV produced an increase in NF-kappaB, AP-1, and Sp1 binding in primary monocytes. In contrast, binding of Ets-2 was dramatically impaired following HIV infection of monocytes, but was unaffected in THP-1 cells. These data clearly show that although LPS induces IL-12p40 expression in primary monocytes and THP-1 cells, the signaling pathways involved and the effect of HIV infection differ and can have disparate effects in these two cell types.


Assuntos
Infecções por HIV/imunologia , Subunidade p40 da Interleucina-12/biossíntese , Lipopolissacarídeos/imunologia , Sistema de Sinalização das MAP Quinases , Células Mieloides/imunologia , Transdução de Sinais , Linhagem Celular , Células Cultivadas , DNA/metabolismo , Humanos , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Proto-Oncogênica c-ets-2/metabolismo , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição AP-1/metabolismo
8.
Biochem Cell Biol ; 85(6): 696-708, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18059528

RESUMO

The effect of lipoprotein electrostatic properties on the catalytic regulation of hepatic lipase (HL) was investigated. Enrichment of serum or very low density lipoprotein (VLDL) with oleic acid increased lipoprotein negative charge and stimulated lipid hydrolysis by HL. Similarly, enrichment of serum or isolated lipoproteins with the anionic phospholipids phosphatidylinositol (PI), phosphatidic acid, or phosphatidylserine also increased lipoprotein negative charge and stimulated hydrolysis by HL. Anionic lipids had a small effect on phospholipid hydrolysis, but significantly stimulated triacylglyceride (TG) hydrolysis. High density lipoprotein (HDL) charge appears to have a specific effect on lipolysis. Enrichment of HDL with PI significantly stimulated VLDL-TG hydrolysis by HL. To determine whether HDL charge affects the association of HL with HDL and VLDL, HL-lipoprotein interactions were probed immunochemically. Under normal circumstances, HL associates with HDL particles, and only small amounts bind to VLDL. PI enrichment of HDL blocked the binding of HL with HDL. These data indicate that increasing the negative charge of HDL stimulates VLDL-TG hydrolysis by reducing the association of HL with HDL. Therefore, HDL controls the hydrolysis of VLDL by affecting the interlipoprotein association of HL. Lipoprotein electrostatic properties regulate lipase association and are an important regulator of the binding and activity of lipolytic enzymes.


Assuntos
Lipase/metabolismo , Lipoproteínas/metabolismo , Jejum/sangue , Ácidos Graxos/metabolismo , Humanos , Hidrólise , Hiperlipidemias/sangue , Lipoproteínas HDL/sangue , Lipoproteínas VLDL/sangue , Modelos Biológicos , Fosfatidilinositóis/sangue , Período Pós-Prandial , Eletricidade Estática , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA