Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-30778332

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive human brain tumors due to their high capacity to migrate and invade normal brain tissue. Epidemiological data report that GBM occur in a greater proportion in men than in women (3:2), suggesting the participation of sex hormones in the development of these tumors. It has been reported an increase in testosterone (T) levels in patients with GBM. In addition, androgen receptor (AR) is overexpressed in human GBM, and genetic silencing of AR, and its pharmacological inhibition, induce GBM cell death in vivo and in vitro. However, the role of T in proliferation, migration and invasion in human GBM cell lines has not been evaluated. We observed that T increased the number of U87, U251, and D54 cells derived from human GBM due to an increase in cell proliferation. This induction was blocked with flutamide, an antagonist of AR. T also induced migration and invasion of GBM cells that flutamide partially blocked. These data suggest that T through AR contributes to the progression of GBM by promoting proliferation, migration, and invasion.

2.
Mol Cell Endocrinol ; 477: 81-89, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29894708

RESUMO

BACKGROUND AND AIMS: Glioblastoma is the most frequent and aggressive brain tumor due to its high capacity to migrate and invade normal brain tissue. The steroid hormone progesterone (P4) contributes to the progression of glioblastoma by promoting proliferation, migration, and cellular invasion through the activation of its intracellular receptor (PR). However, the use of PR antagonist RU486 partially blocks the effects of P4, suggesting the participation of signaling pathways such as those mediated by membrane receptors to P4 (mPRs). Therefore, this study aimed to investigate the effects of mPRα subtype activation on proliferation, migration, and invasion of human glioblastoma cells. METHODS: We treated human glioblastoma cell lines U87 and U251 with the specific mPRα agonist Org OD 02-0, and evaluated its effects on cell number, proliferation, migration, and invasion. Additionally, we measured the phosphorylation of the kinases Src and Akt in both cell lines upon Org OD 02-0 treatment. RESULTS: Org OD 02-0 (100 nM) augmented the number of U87 and U251 cells by increasing cell proliferation. The treatment with this agonist also increased U87 and U251 cell migration and invasion. Both proliferation and cell invasion decreased when mPRα expression was silenced. Finally, we observed that Org OD 02-0 increased the content of p-Src and p-Akt in both cell lines. CONCLUSION: Our data suggest that P4 produces its effects in human glioblastoma progression not only by PR interaction but also through cell signaling pathways activated by mPRα.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Membrana Celular/metabolismo , Movimento Celular , Glioblastoma/metabolismo , Glioblastoma/patologia , Receptores de Progesterona/metabolismo , Neoplasias Encefálicas/enzimologia , Contagem de Células , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glioblastoma/enzimologia , Humanos , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Progesterona/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Progesterona/agonistas , Quinases da Família src/metabolismo
3.
Int J Mol Sci ; 19(3)2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29543748

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive brain tumors. In these malignancies, progesterone (P4) promotes proliferation, migration, and invasion. The P4 metabolite allopregnanolone (3α-THP) similarly promotes cell proliferation in the U87 human GBM cell line. Here, we evaluated global changes in gene expression of U87 cells treated with 3α-THP, P4, and the 5α-reductase inhibitor, finasteride (F). 3α-THP modified the expression of 137 genes, while F changed 90. Besides, both steroids regulated the expression of 69 genes. After performing an over-representation analysis of gene ontology terms, we selected 10 genes whose products are cytoskeleton components, transcription factors, and proteins involved in the maintenance of DNA stability and replication to validate their expression changes by RT-qPCR. 3α-THP up-regulated six genes, two of them were also up-regulated by F. Two genes were up-regulated by P4 alone, however, such an effect was blocked by F when cells were treated with both steroids. The remaining genes were regulated by the combined treatments of 3α-THP + F or P4 + F. An in-silico analysis revealed that promoters of the six up-regulated genes by 3α-THP possess cyclic adenosine monophosphate (cAMP) responsive elements along with CCAAT/Enhancer binding protein alpha (CEBPα) binding sites. These findings suggest that P4 and 3α-THP regulate different sets of genes that participate in the growth of GBMs.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Pregnanolona/farmacologia , Transcriptoma/efeitos dos fármacos , Inibidores de 5-alfa Redutase/farmacologia , Linhagem Celular Tumoral , Citoesqueleto/genética , Citoesqueleto/metabolismo , Finasterida/farmacologia , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima
4.
Anim Reprod Sci ; 188: 123-129, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29175176

RESUMO

Chromosomal sex and steroid hormones play a determining role in brain sexual differentiation during chick embryonic development. Hormone effects on the brain are associated with the expression pattern of their intracellular receptors, which is sexually dimorphic in many species. We determined by Western blot the content of progesterone, estrogen, and androgen receptors (PR-A and PR-B, ERα, and AR, respectively) in the cortex, cerebellum, tectum, and hypothalamus of female and male newly hatched chicks. Males presented a higher content of PR-B in the tectum whereas females exhibited a higher content of PR-A in the hypothalamus. ERα was only detected as a band of 66kDa, and it showed a higher content in the cerebellum and tectum of females as compared to these regions in males. Besides, males exhibited a higher content of AR in the tectum than females. Our study suggests that newly hatched chicks show a sexual dimorphism in the expression of sex hormone receptors in brain regions involved in sexual behavior such as the hypothalamus, and in non-sexual behavior such as the optic tectum and the cerebellum.


Assuntos
Galinhas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Animais , Feminino , Masculino , Isoformas de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores Sexuais
5.
Biomed Res Int ; 2017: 1295087, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28168193

RESUMO

Progesterone-induced blocking factor (PIBF) is a progesterone (P4) regulated protein expressed in different types of high proliferative cells including astrocytomas, the most frequent and aggressive brain tumors. It has been shown that PIBF increases the number of human astrocytoma cells. In this work, we evaluated PIBF regulation by P4 and the effects of PIBF on proliferation, migration, and invasion of U87 and U251 cells, both derived from human glioblastomas. PIBF mRNA expression was upregulated by P4 (10 nM) from 12 to 24 h. Glioblastoma cells expressed two PIBF isoforms, 90 and 57 kDa. The content of the shorter isoform was increased by P4 at 24 h, while progesterone receptor antagonist RU486 (10 µM) blocked this effect. PIBF (100 ng/mL) increased the number of U87 cells on days 4 and 5 of treatment and induced cell proliferation on day 4. Wound-healing assays showed that PIBF increased the migration of U87 (12-48 h) and U251 (24 and 48 h) cells. Transwell invasion assays showed that PIBF augmented the number of invasive cells in both cell lines at 24 h. These data suggest that PIBF promotes proliferation, migration, and invasion of human glioblastoma cells.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Proteínas da Gravidez/metabolismo , Fatores Supressores Imunológicos/metabolismo , Neoplasias Encefálicas/genética , Contagem de Células , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Humanos , Peso Molecular , Invasividade Neoplásica , Proteínas da Gravidez/genética , Progesterona/farmacologia , Isoformas de Proteínas/metabolismo , Fatores Supressores Imunológicos/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
6.
Steroids ; 119: 36-42, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28119080

RESUMO

Allopregnanolone (3α-THP) is one of the main reduced progesterone (P4) metabolites that is recognized as a neuroprotective and myelinating agent. 3α-THP also induces proliferation of different neural cells. It has been shown that P4 favors the progression of glioblastomas (GBM), the most common and aggressive primary brain tumors. However, the role of 3α-THP in the growth of GBMs is unknown. Here, we studied the effects of 3α-THP on the number of cells, proliferation and gene expression in U87 cell line derived from a human GBM. 3α-THP (10, 100nM and 1µM) increased the number of U87 cells, and at 10nM exerted a similar increase in both the number of total and proliferative U87 cells as compared with P4 (10nM). Interestingly, finasteride (F; 100nM), an inhibitor of 5α-reductase (5αR), an enzyme necessary to metabolize P4 and produce 3α-THP, blocked the increase in the number of U87 cells induced by P4. By using RT-qPCR, we determined that U87 cells express 5α-R isoenzymes 1 and 2 (5αR1 and 5αR2), being 5αR1 the predominant one in these cells. 3α-THP (10nM) increased the expression of TGFß1, EGFR, VEGF and cyclin D1 genes. P4 increased TGFß1 and EGFR expression, and this effect was blocked by F. These data provide evidence that P4, through its metabolite 3α-THP, can promote in part cell proliferation of human GBM cells by changing the expression of genes involved in tumor progression.


Assuntos
Glioblastoma/metabolismo , Pregnanolona/farmacologia , Progesterona/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colestenona 5 alfa-Redutase/metabolismo , Ciclina D1/metabolismo , Receptores ErbB/metabolismo , Humanos , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Mol Cell Endocrinol ; 439: 317-327, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27663075

RESUMO

Many progesterone (P4) actions are mediated by its intracellular receptor (PR), which has two isoforms (PR-A and PR-B) differentially transcribed from separate promoters of a single gene. In glioblastomas, the most frequent and aggressive brain tumors, PR-B is the predominant isoform. In an in silico analysis we showed putative CCAAT/Enhancer Binding Protein (C/EBP) binding sites at PR-B promoter. We evaluated the role of C/EBPß in PR-B expression regulation in glioblastoma cell lines, which expressed different ratios of PR and C/EBPß isoforms (LAP1, LAP2, and LIP). ChIP assays showed a significant basal binding of C/EBPß, specific protein 1 (Sp1) and estrogen receptor alpha (ERα) to PR-B promoter. C/EBPß knockdown increased PR-B expression and treatment with estradiol (E2) reduced C/EBPß binding to the promoter and up-regulated PR-B expression. P4 induced genes were differently regulated when CEBP/ß was silenced. These data show that C/EBPß negatively regulates PR-B expression in glioblastoma cells.


Assuntos
Neoplasias Encefálicas/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Receptores de Progesterona/genética , Sítios de Ligação/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Simulação por Computador , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Humanos , Progesterona/farmacologia , Regiões Promotoras Genéticas , Ligação Proteica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/metabolismo , Fator de Transcrição Sp1/metabolismo
8.
J Steroid Biochem Mol Biol ; 172: 198-206, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27717886

RESUMO

Intracellular progesterone receptors (PRs) and protein kinases C (PKCs) are known regulators of cancer cell proliferation and metastasis. Both PRs and PKCs are found overexpressed in grade IV human astrocytomas, also known as glioblastomas, which are the most frequent and aggressive brain tumors. In the present study, we investigated whether PR activation by PKC induces the migration and invasion of glioblastoma derived cell lines and if PKCα and δ isoforms are involved in PR activation. We observed that PKC activation with tetradecanoylphorbol acetate (TPA) increases the migration and invasion capacity of two human glioblastoma derived human cell lines (U251 MG and U87) and that the treatment with the PR receptor antagonist RU486 blocks these processes. Interestingly, the pharmacological inhibition of the isoenzymes PKCα and PKCδ also resulted in a blocked PR transcriptional activity. Also, TPA-dependent PR activation increases the expression of progesterone-induced blocking factor (PIBF), a known PR target gene. These results hint to an existing cross-talk between PKCs and PRs in regulating the infiltration process of human glioblastomas.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neuroglia/metabolismo , Proteína Quinase C-alfa/genética , Proteína Quinase C-delta/genética , Receptores de Progesterona/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Humanos , Mifepristona/farmacologia , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Transdução de Sinais , Fatores Supressores Imunológicos/genética , Fatores Supressores Imunológicos/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
9.
Steroids ; 105: 19-25, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26639431

RESUMO

Astrocytomas are the most common and aggressive primary brain tumors in humans. Invasiveness of these tumors has been attributed in part to deregulation of cell motility-dependent cytoskeletal dynamics that involves actin-binding proteins such as cofilin. Progesterone (P4) has been found to induce migration and invasion of cells derived from breast cancer and endothelium. However, the role of P4 in migration and invasion of astrocytoma cells as well as its effects on astrocytomas cytoskeleton remodeling is not known. In this work we evaluated these aspects in D54 and U251 cells derived from human astrocytomas from the highest degree of malignancy (grade IV, glioblastoma). Our results showed that in scratch-wound assays P4 increased the number of D54 and U251 cells migrating from 3 to 48 h. Both RU486, a P4 receptor (PR) antagonist, and an oligonucleotide antisense against PR significantly blocked P4 effects. Transwell assays showed that P4 significantly increased the number of invasive cells at 24h. As in the case of migration, this effect was blocked by RU486. Finally, by Western blotting, an increase in the cofilin/p-cofilin ratio at 15 and 30 min and a decrease at 30 and 60 min in U251 and D54 cells, respectively, was observed after P4, P4+RU486 and RU486 treatments. These data suggest that P4 increases human astrocytoma cells migration and invasion through its intracellular receptor, and that cofilin activation by P4 is independent of PR action.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Astrocitoma/metabolismo , Astrocitoma/patologia , Movimento Celular/efeitos dos fármacos , Progesterona/farmacologia , Linhagem Celular Tumoral , Humanos , Mifepristona/farmacologia , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Receptores de Progesterona/metabolismo
10.
Front Psychiatry ; 6: 165, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26635640

RESUMO

In the brain of female mammals, including humans, a number of physiological and behavioral changes occur as a result of sex hormone exposure. Estradiol and progesterone regulate several brain functions, including learning and memory. Sex hormones contribute to shape the central nervous system by modulating the formation and turnover of the interconnections between neurons as well as controlling the function of glial cells. The dynamics of neuron and glial cells morphology depends on the cytoskeleton and its associated proteins. Cytoskeletal proteins are necessary to form neuronal dendrites and dendritic spines, as well as to regulate the diverse functions in astrocytes. The expression pattern of proteins, such as actin, microtubule-associated protein 2, Tau, and glial fibrillary acidic protein, changes in a tissue-specific manner in the brain, particularly when variations in sex hormone levels occur during the estrous or menstrual cycles or pregnancy. Here, we review the changes in structure and organization of neurons and glial cells that require the participation of cytoskeletal proteins whose expression and activity are regulated by estradiol and progesterone.

11.
J Steroid Biochem Mol Biol ; 154: 176-85, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26275946

RESUMO

Progesterone (P) participates in the regulation of the growth of several tumors, including astrocytomas, the most common and malignant human brain tumors. It has been reported that P induces astrocytomas growth in part by its interaction with its intracellular receptors (PR). Recently, it has been reported that membrane progesterone receptors (mPRs) are expressed in ovarian and breast cancer cells, and that P could exert some actions through these receptors, however, it is unknown whether mPRs are expressed in astrocytomas. In this work, U251 and U87 cell lines derived from human astrocytomas grade IV were used to study the expression, localization and hormonal regulation of three mPRs subtypes. Using RT-qPCR and Western blot techniques, we found that mPRα and mPRß are clearly expressed at mRNA and protein levels in astrocytoma cells whereas mPRγ was barely expressed in these cells. Immunofluorescence staining showed that mPRα and mPRß were mainly located in the cell surface. Flow cytometry assays demonstrated that in U251 and U87 cells, mPRß is expressed by a higher percentage of both permeabilized and non-permeabilized cells as compared with mPRα. The percentage of cells expressing mPRγ was very low. P and estradiol (E) (10, 100 nM and 1 µM) decreased mPRα protein content at 12 h. In contrast, both P (100 nM and 1 µM) and E (10 and 100 nM) increased mPRß content. Finally, by in silico analysis, we identified that mPRα, mPRß and mPRγ promoters contain several progesterone and estrogen response elements. Our results indicate that mPRs are expressed in human astrocytoma cells, exhibiting a differential regulation by E and P. These data suggest that some P actions in astrocytoma cells may be mediated by mPRs.


Assuntos
Astrocitoma/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo , Astrocitoma/patologia , Linhagem Celular Tumoral , Humanos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Receptores de Progesterona/genética
12.
Int J Endocrinol ; 2015: 674915, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26064112

RESUMO

The CCAAT/enhancer-binding protein beta (C/EBPß) is a transcription factor expressed in different areas of the brain that regulates the expression of several genes involved in cell differentiation and proliferation. This protein has three isoforms (LAP1, LAP2, and LIP) with different transcription activation potential. The role of female sex hormones in the expression pattern of C/EBPß isoforms in the rat brain has not yet been described. In this study we demonstrate by western blot that the expression of the three C/EBPß isoforms changes in different brain areas during the estrous cycle. In the cerebellum, LAP2 content diminished on diestrus and proestrus and LIP content diminished on proestrus and estrus days. In the prefrontal cortex, LIP content was higher on proestrus and estrus days. In the hippocampus, LAP isoforms presented a switch on diestrus day, since LAP1 content was the highest while that of LAP2 was the lowest. The LAP2 isoform was the most abundant one in all the three brain areas. The LAP/LIP ratio changed throughout the cycle and was tissue specific. These results suggest that C/EBPß isoforms expression changes in a tissue-specific manner in the rat brain due to the changes in sex steroid hormone levels presented during the estrous cycle.

13.
Endocrinology ; 156(3): 1010-22, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25514083

RESUMO

Progesterone regulates cancer cell proliferation and invasion through its receptors (PR-A and PR-B), whose phosphorylation modifies their transcriptional activity and induce their degradation. We identified by in silico analysis a putative residue (Ser400) in PR that might be phosphorylated by protein kinase C (PKC), a family of enzymes involved in the proliferation and infiltration of astrocytomas, the most frequent and aggressive brain tumors. A grade III human astrocytoma-derived cell line was used to study the role of PKC in PR phosphorylation, transcriptional activity, and degradation. Treatment with PKC activator [tetradecanoyl phorbol acetate (TPA)] increased PR phosphorylation in Ser400 after 5 minutes, which in turn induced PR transcriptional activity and its subsequent degradation by the 26S proteasome 3-5 hours after treatment. Silencing or inhibition of PKCα and PKCδ blocked PR phosphorylation and degradation induced by TPA. Both PR isoforms were associated with PKCα and reached the maximum association after 5 minutes of TPA addition. These data correlated with immunnofluorescence assays in which nuclear colocalization of PKCα with PR increased after TPA treatment. We observed a 2-fold increase in cell proliferation after PKC activation with TPA that was reduced with the PR antagonist, RU486. The PR S400A mutant revealed that this residue is essential for PKC-mediated PR phosphorylation and degradation. Our results show a key participation of PKCα and PKCδ in PR regulation and function.


Assuntos
Astrocitoma/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , Receptores de Progesterona/metabolismo , Substituição de Aminoácidos , Linhagem Celular Tumoral , Humanos , Isoenzimas , Fosforilação , Proteína Quinase C-alfa/genética , Proteína Quinase C-delta/genética , Piridinas , Receptores de Progesterona/genética , Transcrição Gênica
14.
Methods Mol Biol ; 1165: 89-98, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24839021

RESUMO

Steroid hormone receptors (SHR) are important transcription factors for regulating different physiological and pathological processes. Their altered expression has been strongly associated to cancer progression. Epigenetic marks such as DNA methylation have been proposed as one of the regulatory mechanisms for SHR expression in cancer. DNA methylation occurs at CpG dinucleotides, which form clusters known as CpG islands. These islands are mostly observed at promoter regions of housekeeping genes, and their aberrant methylation in cancer cells is associated with silencing of tumor-suppressor gene expression. SHR genes are characterized for presenting alternative promoters with different CpG island content, which are prone to be methylated. The method of choice for studying DNA methylation is bisulfite sequencing, since it provides information about the methylation pattern at single-nucleotide level. The method is based on the deamination of cytosine residues to uracil after treatment with sodium bisulfite. The converted DNA is amplified by a polymerase chain reaction, cloned, and sequenced. Here, we describe a protocol for bisulfite sequencing suitable for analyzing different CpG regions in SHR genes.


Assuntos
Metilação de DNA , Receptores Citoplasmáticos e Nucleares/genética , Análise de Sequência de DNA/métodos , Clonagem Molecular , Ilhas de CpG/genética , Metilação de DNA/efeitos dos fármacos , Primers do DNA/genética , Escherichia coli/citologia , Escherichia coli/genética , Vetores Genéticos/genética , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Sulfitos/farmacologia , Transformação Genética
15.
Steroids ; 78(5): 500-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23474171

RESUMO

Many progesterone (P4) effects are mediated by its intracellular receptor (PR), which has two isoforms, PR-A and PR-B, each of them with different function and regulation. Differential PR expression in cancer cells has been associated to a PR isoform-specific promoter methylation. In astrocytomas, the most frequent and aggressive brain tumors, PR isoforms expression is directly correlated to the tumor's evolution grade. However, there is no evidence of the role of epigenetic regulation of PR expression in astrocytomas. We evaluated the effect of the demethylating agent 5-aza-2'-deoxycytidine (5AzadC) and the histone deacetylase inhibitor trichostatin A (TSA) on PR expression in human astrocytoma cell lines U373 (grade III) and D54 (grade IV) by RT-PCR and Western blot. Total PR expression increased with 5 µM 5AzadC treatment, whereas PR-B expression increased with 5 and 10 µM 5AzadC treatment in U373 cells, but not in D54 cells. In U373 cells, PR-A protein content augmented with 10 µM 5AzadC treatment, while PR-B content increased with 5 and 10 µM 5AzadC. PR-B expression was not modified by the TSA concentrations that were used, and the combination with 5AzadC did not change the effects of the latter. The study of 5AzadC effects on the number of astrocytoma cells showed that P4 treatment increased the number of U373 cells, whereas 5AzadC and the combined treatment with P4 reduced it. Our results suggest that PR-B expression is regulated by methylation and not by histone acetylation in U373 cells, and that DNA demethylation reduced the number of U373 cells.


Assuntos
Astrocitoma/patologia , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Acetilação/efeitos dos fármacos , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Decitabina , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/farmacologia , Gradação de Tumores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
16.
Biochim Biophys Acta ; 1823(2): 379-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22142990

RESUMO

Estradiol (E2) regulates several cellular functions through the interaction with estrogen receptor subtypes, ERα and ERß, which present different functional and regulation properties. ER subtypes have been identified in human astrocytomas, the most common and aggressive primary brain tumors. We studied the role of ER subtypes in cell growth of two human astrocytoma cell lines derived from tumors of different evolution grades: U373 and D54 (grades III and IV, respectively). E2 significantly increased the number of cells in both lines and the co-administration with an ER antagonist (ICI 182, 780) significantly blocked E2 effects. ERα was the predominant subtype in both cell lines. E2 and ICI 182, 780 down-regulated ERα expression. The number of U373 and D54 cells significantly increased after PPT (ERα agonist) treatment but not after DPN (ERß agonist) one. To determine the role of SRC-1 and SRC-3 coactivators in ERα induced cell growth, we silenced them with RNA interference. Coactivator silencing blocked the increase in cell number induced by PPT. The content of proteins involved in proliferation and metastasis was also determined after PPT treatment. Western blot analysis showed that in U373 cells the content of PR isoforms (PR-A and PR-B), EGFR, VEGF and cyclin D1 increased after PPT treatment while in D54 cells only the content of EGFR was increased. Our results demonstrate that E2 induces cell growth of human astrocytoma cell lines through ERα and its interaction with SRC-1 and SRC-3 and also suggest differential roles of ERα on cell growth depending on astrocytoma grade.


Assuntos
Astrocitoma/fisiopatologia , Neoplasias Encefálicas/fisiopatologia , Linhagem Celular Tumoral , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Coativador 1 de Receptor Nuclear/metabolismo , Coativador 3 de Receptor Nuclear/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/fisiologia , Ciclina D1/genética , Ciclina D1/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Receptor alfa de Estrogênio/genética , Humanos , Coativador 1 de Receptor Nuclear/genética , Coativador 3 de Receptor Nuclear/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA