Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Stem Cells ; 38(3): 369-381, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31778245

RESUMO

Thyroid hormones are regarded as the major controllers of metabolic rate and oxygen consumption in mammals. Although it has been demonstrated that thyroid hormone supplementation improves bovine embryo development in vitro, the cellular mechanisms underlying these effects are so far unknown. In this study, we investigated the role of thyroid hormone in development of human preimplantation embryos. Embryos were cultured in the presence or absence of 10-7 M triiodothyronine (T3) till blastocyst stage. Inner cell mass (ICM) and trophectoderm (TE) were separated mechanically and subjected to RNAseq or quantification of mitochondrial DNA copy number. Analyses were performed using DESeq (v1.16.0 on R v3.1.3), MeV4.9 and MitoMiner 4.0v2018 JUN platforms. We found that the exposure of human preimplantation embryos to T3 had a profound impact on nuclear gene transcription only in the cells of ICM (1178 regulated genes-10.5% of 11 196 expressed genes) and almost no effect on cells of TE (38 regulated genes-0.3% of expressed genes). The analyses suggest that T3 induces in ICM a shift in ribosome and oxidative phosphorylation activity, as the upregulated genes are contributing to the composition and organization of the respiratory chain and associated cofactors involved in mitoribosome assembly and stability. Furthermore, a number of genes affecting the citric acid cycle energy production have reduced expression. Our findings might explain why thyroid disorders in women have been associated with reduced fertility and adverse pregnancy outcome. Our data also raise a possibility that supplementation of culture media with T3 may improve outcomes for women undergoing in vitro fertilization.


Assuntos
Blastocisto/metabolismo , Mitocôndrias/metabolismo , Hormônios Tireóideos/metabolismo , Feminino , Humanos , Fosforilação Oxidativa , Gravidez
2.
Stem Cells ; 33(2): 416-28, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25330987

RESUMO

Biological processes require close cooperation of multiple transcription factors that integrate different signals. Thyroid hormone receptors (TRs) induce Krüppel-like factor 9 (KLF9) to regulate neurogenesis. Here, we show that triiodothyronine (T3) also works through TR to induce KLF9 in HepG2 liver cells, mouse liver, and mouse and human primary hepatocytes and sought to understand TR/KLF9 network function in the hepatocyte lineage and stem cells. Knockdown experiments reveal that KLF9 regulates hundreds of HepG2 target genes and modulates T3 response. Together, T3 and KLF9 target genes influence pathways implicated in stem cell self-renewal and differentiation, including Notch signaling, and we verify that T3 and KLF9 cooperate to regulate key Notch pathway genes and work independently to regulate others. T3 also induces KLF9 in human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSC) and this effect persists during differentiation to definitive endoderm and hiPSC-derived hepatocytes. Microarray analysis reveals that T3 regulates hundreds of hESC and hiPSC target genes that cluster into many of the same pathways implicated in TR and KLF9 regulation in HepG2 cells. KLF9 knockdown confirms that TR and KLF9 cooperate to regulate Notch pathway genes in hESC and hiPSC, albeit in a partly cell-specific manner. Broader analysis of T3 responsive hESC/hiPSC genes suggests that TRs regulate multiple early steps in ESC differentiation. We propose that TRs cooperate with KLF9 to regulate hepatocyte proliferation and differentiation and early stages of organogenesis and that TRs exert widespread and important influences on ESC biology.


Assuntos
Diferenciação Celular/fisiologia , Hepatócitos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Pluripotentes/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Transdução de Sinais/fisiologia , Animais , Feminino , Células Hep G2 , Hepatócitos/citologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Células-Tronco Pluripotentes/citologia , Receptores Notch/genética , Receptores Notch/metabolismo , Receptores dos Hormônios Tireóideos/genética , Tri-Iodotironina/genética , Tri-Iodotironina/metabolismo
3.
Biochem Biophys Res Commun ; 464(3): 718-23, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26168725

RESUMO

Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor gamma (PPARγ) agonists that improve insulin resistance but trigger side effects such as weight gain, edema, congestive heart failure and bone loss. GQ-16 is a PPARγ partial agonist that improves glucose tolerance and insulin sensitivity in mouse models of obesity and diabetes without inducing weight gain or edema. It is not clear whether GQ-16 acts as a partial agonist at all PPARγ target genes, or whether it displays gene-selective actions. To determine how GQ-16 influences PPARγ activity on a gene by gene basis, we compared effects of rosiglitazone (Rosi) and GQ-16 in mature 3T3-L1 adipocytes using microarray and qRT-PCR. Rosi changed expression of 1156 genes in 3T3-L1, but GQ-16 only changed 89 genes. GQ-16 generally showed weak effects upon Rosi induced genes, consistent with partial agonist actions, but a subset of modestly Rosi induced and strongly repressed genes displayed disproportionately strong GQ-16 responses. PPARγ partial agonists MLR24 and SR1664 also exhibit disproportionately strong effects on transcriptional repression. We conclude that GQ-16 displays a continuum of weak partial agonist effects but efficiently represses some negatively regulated PPARγ responsive genes. Strong repressive effects could contribute to physiologic actions of GQ-16.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Resistência à Insulina/genética , Camundongos , Rosiglitazona
4.
J Immunol ; 186(7): 4354-60, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21357268

RESUMO

Glucocorticoids exert potent anti-inflammatory effects by repressing proinflammatory genes. We previously demonstrated that estrogens repress numerous proinflammatory genes in U2OS cells. The objective of this study was to determine if cross talk occurs between the glucocorticoid receptor (GR) and estrogen receptor (ER)α. The effects of dexamethasone (Dex) and estradiol on 23 proinflammatory genes were examined in human U2OS cells stably transfected with ERα or GR. Three classes of genes were regulated by ERα and/or GR. Thirteen genes were repressed by both estradiol and Dex (ER/GR-repressed genes). Five genes were repressed by ER (ER-only repressed genes), and another five genes were repressed by GR (GR-only repressed genes). To examine if cross talk occurs between ER and GR at ER/GR-repressed genes, U2OS-GR cells were infected with an adenovirus that expresses ERα. The ER antagonist, ICI 182780 (ICI), blocked Dex repression of ER/GR-repressed genes. ICI did not have any effect on the GR-only repressed genes or genes activated by Dex. These results demonstrate that ICI acts on subset of proinflammatory genes in the presence of ERα but not on GR-activated genes. ICI recruited ERα to the IL-8 promoter but did not prevent Dex recruitment of GR. ICI antagonized Dex repression of the TNF response element by blocking the recruitment of nuclear coactivator 2. These findings indicate that the ICI-ERα complex blocks Dex-mediated repression by interfering with nuclear coactivator 2 recruitment to GR. Our results suggest that it might be possible to exploit ER and GR cross talk for glucocorticoid therapies using drugs that interact with ERs.


Assuntos
Mediadores da Inflamação/fisiologia , Receptor Cross-Talk/imunologia , Receptores de Estrogênio/fisiologia , Receptores de Glucocorticoides/fisiologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Linhagem Celular Tumoral , Dexametasona/farmacologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Estradiol/fisiologia , Antagonistas de Estrogênios/farmacologia , Fulvestranto , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/antagonistas & inibidores , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Receptor Cross-Talk/efeitos dos fármacos , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Glucocorticoides/antagonistas & inibidores
5.
R Soc Open Sci ; 10(5): 230109, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37153362

RESUMO

We previously demonstrated that hypothyroidism increases peroxisomal biogenesis in rat brown adipose tissue (BAT). We also showed heterogeneity in peroxisomal origin and their unique structural association with mitochondria and/or lipid bodies to carry out ß-oxidation, contributing thus to BAT thermogenesis. Distinctive heterogeneity creates structural compartmentalization within peroxisomal population, raising the question of whether it is followed by their functional compartmentalization regarding localization/colocalization of two main acyl-CoA oxidase (ACOX) isoforms, ACOX1 and ACOX3. ACOX is the first and rate-limiting enzyme of peroxisomal ß-oxidation, and, to date, their protein expression patterns in BAT have not been fully defined. Therefore, we used methimazole-induced hypothyroidism to study ACOX1 and ACOX3 protein expression and their tissue immunolocalization. Additionally, we analysed their specific peroxisomal localization and colocalization in parallel with peroxisomal structural compartmentalization in brown adipocytes. Hypothyroidism caused a linear increase in ACOX1 expression, while a temporary decrease in ACOX3 levels is only recovered to the control level at day 21. Peroxisomal ACOX1 and ACOX3 localization and colocalization patterns entirely mirrored heterogeneous peroxisomal biogenesis pathways and structural compartmentalization, e.g. associations with mitochondria and/or lipid bodies. Hence, different ACOX isoforms localization/colocalization creates distinct functional heterogeneity of peroxisomes and drives their functional compartmentalization in rat brown adipocytes.

6.
FEBS Lett ; 2023 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-38140817

RESUMO

Redox and metabolic processes are tightly coupled in both physiological and pathological conditions. In cancer, their integration occurs at multiple levels and is characterized by synchronized reprogramming both in the tumor tissue and its specific but heterogeneous microenvironment. In breast cancer, the principal microenvironment is the cancer-associated adipose tissue (CAAT). Understanding how the redox-metabolic reprogramming becomes coordinated in human breast cancer is imperative both for cancer prevention and for the establishment of new therapeutic approaches. This review aims to provide an overview of the current knowledge of the redox profiles and regulation of intermediary metabolism in breast cancer while considering the tumor and CAAT of breast cancer as a unique Warburg's pseudo-organ. As cancer is now recognized as a systemic metabolic disease, we have paid particular attention to the cell-specific redox-metabolic reprogramming and the roles of estrogen receptors and circadian rhythms, as well as their crosstalk in the development, growth, progression, and prognosis of breast cancer.

7.
Mol Pharmacol ; 81(6): 788-99, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22391103

RESUMO

The peroxisome proliferator-activated receptor γ (PPARγ) is a target for treatment of type II diabetes and other conditions. PPARγ full agonists, such as thiazolidinediones (TZDs), are effective insulin sensitizers and anti-inflammatory agents, but their use is limited by adverse side effects. Luteolin is a flavonoid with anti-inflammatory actions that binds PPARγ but, unlike TZDs, does not promote adipocyte differentiation. However, previous reports suggested variously that luteolin is a PPARγ agonist or an antagonist. We show that luteolin exhibits weak partial agonist/antagonist activity in transfections, inhibits several PPARγ target genes in 3T3-L1 cells (LPL, ORL1, and CEBPα) and PPARγ-dependent adipogenesis, but activates GLUT4 to a similar degree as rosiglitazone, implying gene-specific partial agonism. The crystal structure of the PPARγ ligand-binding domain (LBD) reveals that luteolin occupies a buried ligand-binding pocket (LBP) but binds an inactive PPARγ LBD conformer and occupies a space near the ß-sheet region far from the activation helix (H12), consistent with partial agonist/antagonist actions. A single myristic acid molecule simultaneously binds the LBP, suggesting that luteolin may cooperate with other ligands to bind PPARγ, and molecular dynamics simulations show that luteolin and myristic acid cooperate to stabilize the Ω-loop among H2', H3, and the ß-sheet region. It is noteworthy that luteolin strongly suppresses hypertonicity-induced release of the pro-inflammatory interleukin-8 from human corneal epithelial cells and reverses reductions in transepithelial electrical resistance. This effect is PPARγ-dependent. We propose that activities of luteolin are related to its singular binding mode, that anti-inflammatory activity does not require H12 stabilization, and that our structure can be useful in developing safe selective PPARγ modulators.


Assuntos
Luteolina/farmacologia , PPAR gama/agonistas , Células 3T3-L1 , Animais , Sequência de Bases , Primers do DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Luteolina/química , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Ácido Mirístico/química , PPAR gama/química , PPAR gama/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosiglitazona , Tiazolidinedionas/antagonistas & inibidores , Tiazolidinedionas/farmacologia
8.
J Biol Chem ; 286(35): 30723-30731, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21719705

RESUMO

Peroxisome proliferator-activated receptor γ (PPARγ) activation induces adipogenesis and also enhances lipogenesis, mitochondrial activity, and insulin sensitivity in adipocytes. Whereas some studies implicate PPARγ coactivator 1α (PGC-1α) in the mitochondrial effect, the mechanisms involved in PPARγ regulation of adipocyte mitochondrial function are not resolved. PPARγ-activating ligands (thiazolidinediones (TZDs)) are important insulin sensitizers and were recently shown to indirectly induce PGC-1ß transcription in osteoclasts. Here, we asked whether similar effects occur in adipocytes and show that TZDs also strongly induce PGC-1ß in cultured 3T3-L1 cells. This effect, however, differs from the indirect effect proposed for bone and is rapid and direct and involves PPARγ interactions with an intronic PPARγ response element cluster in the PGC-1ß locus. TZD treatment of cultured adipocytes results in up-regulation of mitochondrial marker genes, and increased mitochondrial activity and use of short interfering RNA confirms that these effects require PGC-1ß. PGC-1ß did not participate in PPARγ effects on adipogenesis or lipogenesis, and PGC-1ß knockdown did not alter insulin-responsive glucose uptake into 3T3-L1 cells. Similar effects on PGC-1ß and mitochondrial gene expression are seen in vivo; fractionation of obese mouse adipose tissue reveals that PPARγ and PGC-1ß, but not PGC-1α, are coordinately up-regulated in adipocytes relative to preadipocytes and that TZD treatment induces PGC-1ß and mitochondrial marker genes in adipose tissue of obese mice. We propose that PPARγ directly induces PGC-1ß expression in adipocytes and that this effect regulates adipocyte mitochondrial activity.


Assuntos
Adipócitos/citologia , PPAR gama/metabolismo , Transativadores/metabolismo , Células 3T3-L1 , Tecido Adiposo/metabolismo , Animais , Ácidos Graxos/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Obesos , Mitocôndrias/metabolismo , Modelos Biológicos , Consumo de Oxigênio , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Tiazolidinedionas/farmacologia , Fatores de Transcrição
9.
Breast Cancer Res Treat ; 129(3): 777-84, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21120602

RESUMO

The role of estrogen receptor beta (ERß) in breast cancer is unclear. ERß is considered to have a protective role in breast cancer development based on findings demonstrating that ERß expression inhibits ERα-mediated proliferation of breast cancer cells. We previously demonstrated that ERß causes a ligand independent G2 cell cycle arrest in MCF-7 cells. To study the mechanisms of the ERß-mediated G2 cell cycle arrest, we investigated its effects on the regulatory pathways responsible for the G2/M phase transition. We found that ERß inhibits CDK1 activity, which is the critical determinant of the G2/M progression. CDK1 activity is modulated by both stimulatory and inhibitory factors. Cyclin B1 is the major activator of CDK1. ERß inhibited the cell cycle-dependent stimulation of cyclin B1 mRNA and protein. GADD45A and BTG2 are two major inhibitors of CDK1, which have been implicated in breast tumor formation. Based on these findings, we explored if the expression pattern of GADD45A and BTG2 is affected by ERß. We found that ERß stimulates GADD45A and BTG2 mRNA levels. The induction of these two genes is caused by ERß binding directly to these genes and recruiting c-jun and NCOA2. Our findings demonstrated that unliganded ERß causes a G2 cell cycle arrest by inactivating CDK1 through the repression of cyclin B1 and stimulation of GADD45A and BTG2 expression. These results provide evidence that drugs that stimulate the production of unliganded ERß may be effective new therapies to prevent breast cancer.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Receptor beta de Estrogênio/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteína Quinase CDC2/genética , Proteínas de Ciclo Celular/genética , Divisão Celular , Linhagem Celular Tumoral , Ciclina B1/genética , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular , Regulação da Expressão Gênica , Genes jun , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Nucleares/genética , Coativador 2 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/metabolismo , RNA Mensageiro , Proteínas Supressoras de Tumor/genética
10.
Cell Death Dis ; 10(4): 277, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30894508

RESUMO

Variability among donors, non-standardized methods for isolation, and characterization contribute to mesenchymal stem/stromal cell (MSC) heterogeneity. Induced pluripotent stem cell (iPSCs)-derived MSCs would circumvent many of current issues and enable large-scale production of standardized cellular therapy. To explore differences between native MSCs (nMSCs) and iPSC-derived MSCs (iMSCs), we developed isogeneic lines from Wharton's jelly (WJ) from the umbilical cords of two donors (#12 and #13) under xeno-free conditions. Next, we reprogrammed them into iPSCs (iPSC12 and iPSC13) and subsequently differentiated them back into iMSCs (iMSC12 and iMSC13) using two different protocols, which we named ARG and TEX. We assessed their differentiation capability, transcriptome, immunomodulatory potential, and interferon-γ (IFNG)-induced changes in metabolome. Our data demonstrated that although both differentiation protocols yield iMSCs similar to their parental nMSCs, there are substantial differences. The ARG protocol resulted in iMSCs with a strong immunomodulatory potential and lower plasticity and proliferation rate, whereas the TEX protocol raised iMSCs with a higher proliferation rate, better differentiation potential, though weak immunomodulatory response. Our data suggest that, following a careful selection and screening of donors, nMSCs from umbilical's cord WJ can be easily reprogrammed into iPSCs, providing an unlimited source of material for differentiation into iMSCs. However, the differentiation protocol should be chosen depending on their clinical use.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferon gama/farmacologia , Células-Tronco Mesenquimais/metabolismo , Metaboloma/efeitos dos fármacos , Cordão Umbilical/citologia , Plasticidade Celular , Proliferação de Células , Células Cultivadas , Técnicas de Reprogramação Celular/métodos , Feminino , Humanos , Transcriptoma/efeitos dos fármacos
11.
Artigo em Inglês | MEDLINE | ID: mdl-30671024

RESUMO

Thyroid hormone receptors (TRs) are responsible for mediating thyroid hormone (T3 and T4) actions at a cellular level. They belong to the nuclear receptor (NR) superfamily and execute their main functions inside the cell nuclei as hormone-regulated transcription factors. These receptors also exhibit so-called "non-classic" actions, for which other cellular proteins, apart from coregulators inside nuclei, regulate their activity. Aiming to find alternative pathways of TR modulation, we searched for interacting proteins and found that PDIA1 interacts with TRß in a yeast two-hybrid screening assay. The functional implications of PDIA1-TR interactions are still unclear; however, our co-immunoprecipitation (co-IP) and fluorescence assay results showed that PDI was able to bind both TR isoforms in vitro. Moreover, T3 appears to have no important role in these interactions in cellular assays, where PDIA1 was able to regulate transcription of TRα and TRß-mediated genes in different ways depending on the promoter region and on the TR isoform involved. Although PDIA1 appears to act as a coregulator, it binds to a TR surface that does not interfere with coactivator binding. However, the TR:PDIA1 complex affinity and activation are different depending on the TR isoform. Such differences may reflect the structural organization of the PDIA1:TR complex, as shown by models depicting an interaction interface with exposed cysteines from both proteins, suggesting that PDIA1 might modulate TR by its thiol reductase/isomerase activity.

12.
Endocrinology ; 148(2): 538-47, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17095596

RESUMO

Novel estrogenic therapies are needed that ameliorate menopausal symptoms and have the bone-sparing effects of endogenous estrogens but do not promote breast or uterine cancer. Recent evidence suggests that selective activation of the estrogen receptor (ER)-beta subtype inhibits breast cancer cell proliferation. To establish whether ERbeta-selective ligands represent a viable approach to improve hormone therapy, we investigated whether the estrogenic activities present in an herbal extract, MF101, used to treat hot flashes, are ERbeta selective. MF101 promoted ERbeta, but not ERalpha, activation of an estrogen response element upstream of the luciferase reporter gene. MF101 also selectively regulates transcription of endogenous genes through ERbeta. The ERbeta selectivity was not due to differential binding because MF101 binds equally to ERalpha and ERbeta. Fluorescence resonance energy transfer and protease digestion studies showed that MF101 produces a different conformation in ERalpha from ERbeta when compared with the conformations produced by estradiol. The specific conformational change induced by MF101 allows ERbeta to bind to an estrogen response element and recruit coregulatory proteins that are required for gene activation. MF101 did not activate the ERalpha-regulated proliferative genes, c-myc and cyclin D1, or stimulate MCF-7 breast cancer cell proliferation or tumor formation in a mouse xenograft model. Our results demonstrate that herbal ERbeta-selective estrogens may be a safer alternative for hormone therapy than estrogens that nonselectively activate both ER subtypes.


Assuntos
Anemarrhena/química , Receptor beta de Estrogênio/genética , Extratos Vegetais/farmacologia , Ativação Transcricional/efeitos dos fármacos , Animais , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Carcinógenos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Dietilestilbestrol , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Conformação Molecular , Transplante de Neoplasias , Tamanho do Órgão/efeitos dos fármacos , Extratos Vegetais/metabolismo , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transplante Heterólogo , Útero/efeitos dos fármacos , Útero/patologia
13.
Mol Biol Cell ; 15(3): 1262-72, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14699072

RESUMO

Estrogens and selective estrogen receptor modulators (SERMs) interact with estrogen receptor (ER) alpha and beta to activate or repress gene transcription. To understand how estrogens and SERMs exert tissue-specific effects, we performed microarray analysis to determine whether ERalpha or ERbeta regulate different target genes in response to estrogens and SERMs. We prepared human U2OS osteosarcoma cells that are stably transfected with a tetracycline-inducible vector to express ERalpha or ERbeta. Western blotting, immunohistochemistry, and immunoprecipitation studies confirmed that U2OS-ERalpha cells synthesized only ERalpha and that U2OS-ERbeta cells expressed exclusively ERbeta. U2OS-ERalpha and U2OS-ERbeta cells were treated either with 17beta-estradiol (E2), raloxifene, and tamoxifen for 18 h. Labeled cRNAs were hybridized with U95Av2 GeneChips (Affymetrix). A total of 228, 190, and 236 genes were significantly activated or repressed at least 1.74-fold in U2OS-ERalpha and U2OS-ERbeta cells by E2, raloxifene, and tamoxifen, respectively. Most genes regulated in ERalpha cells in response to E2, raloxifene, and tamoxifen were distinct from those regulated in ERbeta cells. Only 38 of the 228 (17%) genes were regulated by E2 in both U2OS-ERalpha and U2OS-ERbeta cells. Raloxifene and tamoxifen regulated only 27% of the same genes in both the ERalpha and ERbeta cells. A subset of genes involved in bone-related activities regulated by E2, raloxifene, and tamoxifen were also distinct. Our results demonstrate that most genes regulated by ERalpha are distinct from those regulated by ERbeta in response to E2 and SERMs. These results indicate that estrogens and SERMs exert tissue-specific effects by regulating unique sets of targets genes through ERalpha and ERbeta


Assuntos
Estradiol/metabolismo , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Cloridrato de Raloxifeno/farmacologia , Tamoxifeno/farmacologia , Células Tumorais Cultivadas
14.
Sci Rep ; 7(1): 17595, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29242640

RESUMO

We investigated whether maternal metabolic environment affects mesenchymal stromal/stem cells (MSCs) from umbilical cord's Wharton's Jelly (WJ) on a molecular level, and potentially render them unsuitable for clinical use in multiple recipients. In this pilot study on umbilical cords post partum from healthy non-obese (BMI = 19-25; n = 7) and obese (BMI ≥ 30; n = 7) donors undergoing elective Cesarean section, we found that WJ MSC from obese donors showed slower population doubling and a stronger immunosuppressive activity. Genome-wide DNA methylation of triple positive (CD73+CD90+CD105+) WJ MSCs found 67 genes with at least one CpG site where the methylation difference was ≥0.2 in four or more obese donors. Only one gene, PNPLA7, demonstrated significant difference on methylome, transcriptome and protein level. Although the number of analysed donors is limited, our data suggest that the altered metabolic environment related to excessive body weight might bear consequences on the WJ MSCs.


Assuntos
Células-Tronco Mesenquimais/patologia , Mães , Obesidade/patologia , Geleia de Wharton/patologia , Adulto , Antígeno CD56/metabolismo , Estudos de Casos e Controles , Diferenciação Celular , Metilação de DNA , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imunomodulação , Lipase/genética , Lisofosfolipase , Células-Tronco Mesenquimais/metabolismo , Obesidade/genética , Obesidade/imunologia , Obesidade/metabolismo , Projetos Piloto , Gravidez
15.
PLoS One ; 11(10): e0164407, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27732649

RESUMO

Thyroid hormone (TH) receptors (TRs α and ß) are homologous ligand-dependent transcription factors (TFs). While the TRs display distinct actions in development, metabolic regulation and other processes, comparisons of TRα and TRß dependent gene regulation mostly reveal similar mechanisms of action and few TR subtype specific genes. Here, we show that TRα predominates in multipotent human adipose derived stem cells (hADSC) whereas TRß is expressed at lower levels and is upregulated during hADSC differentiation. The TRs display several unusual properties in parental hADSC. First, TRs display predominantly cytoplasmic intracellular distribution and major TRα variants TRα1 and TRα2 colocalize with mitochondria. Second, knockdown experiments reveal that endogenous TRs influence hADSC cell morphology and expression of hundreds of genes in the absence of hormone, but do not respond to exogenous TH. Third, TRα and TRß affect hADSC in completely distinct ways; TRα regulates cell cycle associated processes while TRß may repress aspects of differentiation. TRα splice variant specific knockdown reveals that TRα1 and TRα2 both contribute to TRα-dependent gene expression in a gene specific manner. We propose that TRs work in a non-canonical and hormone independent manner in hADSC and that prominent subtype-specific activities emerge in the context of these unusual actions.


Assuntos
Tecido Adiposo/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco/citologia , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Inativação Gênica , Humanos , Células-Tronco/metabolismo , Receptores alfa dos Hormônios Tireóideos/análise , Receptores alfa dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/análise , Receptores beta dos Hormônios Tireóideos/genética , Tri-Iodotironina/análise , Tri-Iodotironina/genética , Tri-Iodotironina/metabolismo
16.
Nucl Recept Signal ; 13: e004, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26445566

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) display anti-inflammatory, antipyretic and analgesic properties by inhibiting cyclooxygenases and blocking prostaglandin production. Previous studies, however, suggested that some NSAIDs also modulate peroxisome proliferator activated receptors (PPARs), raising the possibility that such off target effects contribute to the spectrum of clinically relevant NSAID actions. In this study, we set out to understand how peroxisome proliferator activated receptor-γ (PPARγ/PPARG) interacts with NSAIDs using X-ray crystallography and to relate ligand binding modes to effects on receptor activity. We find that several NSAIDs (sulindac sulfide, diclofenac, indomethacin and ibuprofen) bind PPARγ and modulate PPARγ activity at pharmacologically relevant concentrations. Diclofenac acts as a partial agonist and binds to the PPARγ ligand binding pocket (LBP) in typical partial agonist mode, near the ß-sheets and helix 3. By contrast, two copies of indomethacin and sulindac sulfide bind the LBP and, in aggregate, these ligands engage in LBP contacts that resemble agonists. Accordingly, both compounds, and ibuprofen, act as strong partial agonists. Assessment of NSAID activities in PPARγ-dependent 3T3-L1 cells reveals that NSAIDs display adipogenic activities and exclusively regulate PPARγ-dependent target genes in a manner that is consistent with their observed binding modes. Further, PPARγ knockdown eliminates indomethacin activities at selected endogenous genes, confirming receptor-dependence of observed effects. We propose that it is important to consider how individual NSAIDs interact with PPARγ to understand their activities, and that it will be interesting to determine whether high dose NSAID therapies result in PPAR activation.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , PPAR gama/metabolismo , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Animais , Agonismo Parcial de Drogas , Células HeLa , Humanos , Camundongos , Modelos Moleculares , PPAR gama/agonistas , PPAR gama/química , Conformação Proteica
17.
Int J Biochem Cell Biol ; 34(3): 279-85, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11849995

RESUMO

Glucocorticoid hormone receptor exists in the cytoplasm of target cells in the form of dynamic multiprotein heterocomplexes with heat shock proteins Hsp90 and Hsp70, and additional components of the molecular chaperone machinery. Whole body hyperthermic stress was previously shown to induce alterations in protein composition of these complexes increasing the share of Hsp70, but participation of individual Hsp70 family members was not investigated. In the present study the association of glucocorticoid receptor with constitutive and inducible forms of Hsp70 in the liver cytosol of rats exposed to 41 degrees C whole body hyperthermic stress was examined. Immunoprecipitation of glucocorticoid receptor heterocomplexes by monoclonal anti-receptor antibody (BuGR2) followed by quantitative immunoblotting revealed the presence of both nucleocytoplasmic Hsp70 family members, constitutive--Hsc70 and inducible--Hsp72, within the complexes. Immediately after the stress only Hsc70 was found in association with glucocorticoid receptor. However, after the induction of Hsp72 by stress, its appearance within the glucocorticoid receptor heterocomplexes was also recorded and the presence of both Hsp70 forms within the heterocomplexes was evident by the end of examined 24h period after the stress. This study confirms that heat stress affects protein composition of rat liver glucocorticoid receptor heterocomplexes increasing the share of Hsp70 and shows that this increase could be equally ascribed to constitutive and inducible forms of Hsp70.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Transtornos de Estresse por Calor/metabolismo , Fígado/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Fracionamento Celular , Hipertermia Induzida , Substâncias Macromoleculares , Masculino , Isoformas de Proteínas , Ratos , Ratos Wistar
18.
J Clin Endocrinol Metab ; 99(1): 151-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24217905

RESUMO

CONTEXT: Brown adipose tissue (BAT) has the unique ability of generating heat due to the expression of mitochondrial uncoupling protein 1 (UCP1). A recent discovery regarding functional BAT in adult humans has increased interest in the molecular pathways of BAT development and functionality. An important role for estrogen in white adipose tissue was shown, but the possible role of estrogen in human fetal BAT (fBAT) is unclear. OBJECTIVE: The objective of this study was to determine whether human fBAT expresses estrogen receptor α (ERα) and ERß. In addition, we examined their localization as well as their correlation with crucial proteins involved in BAT differentiation, proliferation, mitochondriogenesis and thermogenesis including peroxisome proliferator-activated receptor γ (PPARγ), proliferating cell nuclear antigen (PCNA), PPARγ-coactivator-1α (PGC-1α), and UCP1. DESIGN: The fBAT was obtained from 4 human male fetuses aged 15, 17, 20, and 23 weeks gestation. ERα and ERß expression was assessed using Western blotting, immunohistochemistry, and immunocytochemistry. Possible correlations with PPARγ, PCNA, PGC-1α, and UCP1 were examined by double immunofluorescence. RESULTS: Both ERα and ERß were expressed in human fBAT, with ERα being dominant. Unlike ERß, which was present only in mature brown adipocytes, we detected ERα in mature adipocytes, preadipocytes, mesenchymal and endothelial cells. In addition, double immunofluorescence supported the notion that differentiation in fBAT probably involves ERα. Immunocytochemical analysis revealed mitochondrial localization of both receptors. CONCLUSION: The expression of both ERα and ERß in human fBAT suggests a role for estrogen in its development, primarily via ERα. In addition, our results indicate that fBAT mitochondria could be targeted by estrogens and pointed out the possible role of both ERs in mitochondriogenesis.


Assuntos
Tecido Adiposo Marrom/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feto/metabolismo , Tecido Adiposo Marrom/embriologia , Tecido Adiposo Marrom/crescimento & desenvolvimento , Idade Gestacional , Humanos , Imuno-Histoquímica , Canais Iônicos/metabolismo , Masculino , Proteínas Mitocondriais/metabolismo , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Distribuição Tecidual , Fatores de Transcrição/metabolismo , Proteína Desacopladora 1
19.
Stem Cell Reports ; 2(5): 675-89, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24936454

RESUMO

Cornification and epidermal barrier defects are associated with a number of clinically diverse skin disorders. However, a suitable in vitro model for studying normal barrier function and barrier defects is still lacking. Here, we demonstrate the generation of human epidermal equivalents (HEEs) from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). HEEs are structurally similar to native epidermis, with a functional permeability barrier. We exposed a pure population of hESC/iPSC-derived keratinocytes, whose transcriptome corresponds to the gene signature of normal primary human keratinocytes (NHKs), to a sequential high-to-low humidity environment in an air/liquid interface culture. The resulting HEEs had all of the cellular strata of the human epidermis, with skin barrier properties similar to those of normal skin. Such HEEs generated from disease-specific iPSCs will be an invaluable tool not only for dissecting molecular mechanisms that lead to epidermal barrier defects but also for drug development and screening.


Assuntos
Células-Tronco Embrionárias/metabolismo , Epiderme/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Transição Epitelial-Mesenquimal , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Queratina-14/genética , Queratina-14/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Permeabilidade , Análise de Componente Principal , Teratoma/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
20.
PLoS One ; 8(7): e70097, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922917

RESUMO

Sirtuin 1 (SIRT1) NAD(+)-dependent deacetylase regulates energy metabolism by modulating expression of genes involved in gluconeogenesis and other liver fasting responses. While many effects of SIRT1 on gene expression are mediated by deacetylation and activation of peroxisome proliferator activated receptor coactivator α (PGC-1α), SIRT1 also binds directly to DNA bound transcription factors, including nuclear receptors (NRs), to modulate their activity. Since thyroid hormone receptor ß1 (TRß1) regulates several SIRT1 target genes in liver and interacts with PGC-1α, we hypothesized that SIRT1 may influence TRß1. Here, we confirm that SIRT1 cooperates with PGC-1α to enhance response to triiodothyronine, T3. We also find, however, that SIRT1 stimulates TRß1 activity in a manner that is independent of PGC-1α but requires SIRT1 deacetylase activity. SIRT1 interacts with TRß1 in vitro, promotes TRß1 deacetylation in the presence of T3 and enhances ubiquitin-dependent TRß1 turnover; a common response of NRs to activating ligands. More surprisingly, SIRT1 knockdown only strongly inhibits T3 response of a subset of TRß1 target genes, including glucose 6 phosphatase (G-6-Pc), and this is associated with blockade of TRß1 binding to the G-6-Pc promoter. Drugs that target the SIRT1 pathway, resveratrol and nicotinamide, modulate T3 response at dual TRß1/SIRT1 target genes. We propose that SIRT1 is a gene-specific TRß1 co-regulator and TRß1/SIRT1 interactions could play important roles in regulation of liver metabolic response. Our results open possibilities for modulation of subsets of TR target genes with drugs that influence the SIRT1 pathway.


Assuntos
Sirtuína 1/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Fatores de Transcrição/metabolismo , Acetilação , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Mapas de Interação de Proteínas , Sirtuína 1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA