Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Environ Health Perspect ; 115(1): 48-52, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17366818

RESUMO

BACKGROUND: Prevalence and morbidity of allergic diseases have increased over the last decades. Based on the recently recognized differences in asthma prevalence between the sexes, we have examined the effect of endogenous estrogens on a key element of the allergic response. Some lipophilic pollutants have estrogen-like activities and are termed environmental estrogens. These pollutants tend to degrade slowly in the environment and to bioaccumulate and bioconcentrate in the food chain; they also have long biological half-lives. OBJECTIVES: Our goal in this study was to identify possible pathogenic roles for environmental estrogens in the development of allergic diseases. METHODS: We screened a number of environmental estrogens for their ability to modulate the release of allergic mediators from mast cells. We incubated a human mast cell line and primary mast cell cultures derived from bone marrow of wild type and estrogen receptor alpha (ER-alpha)-deficient mice with environmental estrogens with and without estradiol or IgE and allergens. We assessed degranulation of mast cells by quantifying the release of beta-hexosaminidase. RESULTS: All of the environmental estrogens tested caused rapid, dose-related release of beta-hexosaminidase from mast cells and enhanced IgE-mediated release. The combination of physiologic concentrations of 17beta-estradiol and several concentrations of environmental estrogens had additive effects on mast cell degranulation. Comparison of bone marrow mast cells from ER-alpha-sufficient and ER-alpha-deficient mice indicated that much of the effect of environmental estrogens was mediated by ER-alpha. CONCLUSIONS: Our findings suggest that estrogenic environmental pollutants might promote allergic diseases by inducing and enhancing mast cell degranulation by physiologic estrogens and exposure to allergens.


Assuntos
Poluentes Ambientais/toxicidade , Estrogênios não Esteroides/toxicidade , Mastócitos/efeitos dos fármacos , Animais , Antígenos de Dermatophagoides/farmacologia , Arocloros/toxicidade , Asma/imunologia , Degranulação Celular/efeitos dos fármacos , Linhagem Celular , Diclorodifenil Dicloroetileno/toxicidade , Dieldrin/toxicidade , Dinitrofenóis/farmacologia , Endossulfano/toxicidade , Estradiol/farmacologia , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Humanos , Imunoglobulina E/imunologia , Mastócitos/imunologia , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Praguicidas/toxicidade , Fenóis/toxicidade , Soroalbumina Bovina/farmacologia
2.
J Infect Dis ; 195(5): 680-3, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17262709

RESUMO

The protective effect of estrogen replacement on ascending urinary-tract infection (UTI) is controversial. We designed a study using an experimental model of UTI in which surgically menopausal mice were supplemented with estrogen and the susceptibility to UTI was evaluated after experimental Escherichia coli infection. The mean rate of E. coli infection in the group not treated with estrogen was 2 x 10(4) cfu/g of renal tissue, compared with 9 x 10(8) cfu/g (P<.001) in the estrogen-treated group. Surprisingly, despite the hypothesis that estrogen would protect mice from infection, estrogen treatment significantly increased the susceptibility of the mice to ascending UTI.


Assuntos
Cistite/prevenção & controle , Infecções por Escherichia coli/prevenção & controle , Estradiol/farmacologia , Estrogênios/metabolismo , Infecções Urinárias/prevenção & controle , Animais , Suscetibilidade a Doenças , Escherichia coli/classificação , Escherichia coli/metabolismo , Infecções por Escherichia coli/metabolismo , Feminino , Proteínas de Fímbrias , Menopausa , Camundongos , Camundongos Endogâmicos C3H , Ovariectomia , Infecções Urinárias/metabolismo
3.
Mol Immunol ; 44(8): 1977-85, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17084457

RESUMO

BACKGROUND: Allergic airway diseases are more common in females than in males during early adulthood. A relationship between female hormones and asthma prevalence and severity has been suggested, but the cellular and molecular mechanisms are not understood. OBJECTIVE: To elucidate the mechanism(s) by which estrogens enhance the synthesis and release of mediators of acute hypersensitivity. METHODS: Two mast cell/basophil cell lines (RBL-2H3 and HMC-1) and primary cultures of bone marrow derived mast cells, all of which naturally express estrogen receptor-alpha, were examined. Cells were incubated with physiological concentrations of 17-beta-estradiol with and without IgE and allergens. Intracellular Ca(2+) concentrations and the release of beta-hexosaminidase and leukotriene C(4) were quantified. RESULTS: Estradiol alone induced partial release of the preformed, granular protein beta-hexosaminidase from RBL-2H3, BMMC and HMC-1, but not from BMMC derived from estrogen receptor-alpha knock-out mice. The newly synthesized LTC(4) was also released from RBL-2H3. Estradiol also enhanced IgE-induced degranulation and potentiated LTC(4) production. Intracellular Ca(2+) concentration increased prior to and in parallel with mediator release. Estrogen receptor antagonists or Ca(2+) chelation inhibited these estrogenic effects. CONCLUSION: Binding of physiological concentrations of estradiol to a membrane estrogen receptor-alpha initiates a rapid onset and progressive influx of extracellular Ca(2+), which supports the synthesis and release of allergic mediators. Estradiol also enhances IgE-dependent mast cell activation, resulting in a shift of the allergen dose response.


Assuntos
Asma/imunologia , Sinalização do Cálcio/imunologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/imunologia , Mastócitos/imunologia , Caracteres Sexuais , Alérgenos/imunologia , Alérgenos/farmacologia , Animais , Asma/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Linhagem Celular , Estradiol/imunologia , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Imunoglobulina E/imunologia , Imunoglobulina E/farmacologia , Leucotrieno C4/imunologia , Leucotrieno C4/metabolismo , Masculino , Mastócitos/metabolismo , Camundongos , Camundongos Knockout , Ratos , beta-N-Acetil-Hexosaminidases/imunologia , beta-N-Acetil-Hexosaminidases/metabolismo
4.
Clin Cancer Res ; 12(19): 5641-7, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17000652

RESUMO

PURPOSE: The risk of developing breast cancer is positively correlated with exposure to increased levels of estrogen and/or an increased duration of estrogen exposure. Many different mechanisms have been proposed to explain the association of estrogens with breast cancer risk; however, the well-documented immune modulatory properties of estrogen have received little attention. In part, this is due to a lack of suitable models for studying this relationship. EXPERIMENTAL DESIGN: We have developed an animal model using estrogen receptor (ER)-negative human breast cancer cell line, MDA-MB-468, xenografted into severe combined immunodeficient (SCID) mice. We also generated the ER-alpha knockout (ER-alphaKO) mice on the SCID background and then tested the ability of 17beta-estradiol to stimulate growth of xenografted ER-negative human breast cancer tumors in wild-type and ER-alphaKO SCID mice. We quantified vascularization of tumors, macrophage recruitment to the tumor site by immunocytochemistry, and inflammatory cytokine production. RESULTS: We show that estrogen treatment of C57BL/6/SCID mice promotes the growth of xenografted ER-negative tumors in wild-type mice and this estrogen-induced tumor growth is abrogated in ER-alphaKO mice. Tumor neovascularization of estrogen-treated mice was unchanged versus control; however, estrogen treatment of the C57BL/6/SCID host suppressed macrophage recruitment to and inflammatory cytokine production at the tumor site. CONCLUSIONS: These data are consistent with estrogen modulation of the inflammatory response as a contributing factor in estrogen-stimulated growth of an ER-negative tumor. This effect on the host innate immune response was mediated by ER-alpha.


Assuntos
Neoplasias da Mama/imunologia , Estradiol/farmacologia , Receptores de Estrogênio/metabolismo , Animais , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Feminino , Humanos , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Transplante Heterólogo , Células Tumorais Cultivadas
5.
Mol Endocrinol ; 20(9): 2020-35, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16645043

RESUMO

Estrogen receptor (ER)alpha activity is controlled by the balance of coactivators and corepressors contained within cells that are recruited into transcriptional complexes. The metastasis-associated protein (MTA) family has been demonstrated to be associated with breast tumor cell progression and ERalpha activity. We demonstrate that MTA2 expression is correlated with ERalpha protein expression in invasive breast tumors. We show that the MTA2 family member can bind to ERalpha and repress its activity in human breast cancer cells. Furthermore, it can inhibit ERalpha-mediated colony formation and render breast cancer cells resistant to estradiol and the growth-inhibitory effects of the antiestrogen tamoxifen. MTA2 participates in the deacetylation of ERalpha protein, potentially through its associated histone deacetylase complex 1 activity. We hypothesize that MTA2 is a repressor of ERalpha activity and that it could represent a new therapeutic target of ERalpha action in human breast tumors.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/metabolismo , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Acetilação , Sítios de Ligação , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Estrogênios/farmacologia , Histona Desacetilases/genética , Humanos , Lisina/genética , Lisina/metabolismo , Mutação/genética , Ligação Proteica , Proteínas Repressoras/genética , Transcrição Gênica/genética
6.
Mol Cell Endocrinol ; 246(1-2): 91-100, 2006 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-16388889

RESUMO

Regulation of breast cancer growth by estrogen is mediated by estrogen receptors (ER) in nuclear and extranuclear compartments. We assessed the structure and functions of extranuclear ER that initiate downstream signaling to the nucleus. ER, including full-length 66-kDa ER and a 46-kDa ER splice variant, are enriched in lipid rafts from MCF-7 cells with (MCF-7/HER-2) or without (MCF-7/PAR) HER-2 gene overexpression and co-localize with HER-1 and HER-2 growth factor receptors, as well as with lipid raft marker flotillin-2. In contrast, ER-negative MCF-7 cells do not express nuclear or lipid raft ER. ER knockdown with siRNA also elicits a marked loss of ER in MCF-7 cell rafts. In MCF-7/PAR cells, estrogen enhances ER association with membrane rafts and induces rapid phosphorylation of nuclear receptor coactivator AIB1, actions not detected in ER-negative cells. Thus, nuclear and lipid raft ER derive from the same transcript, and extranuclear ER co-localizes with HER receptors in membrane signaling domains that modulate downstream nuclear events leading to cell growth.


Assuntos
Neoplasias da Mama/metabolismo , Microdomínios da Membrana/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Humanos , Mutação , RNA/biossíntese , Receptores de Estrogênio/genética , Receptores de Fatores de Crescimento/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Immunol ; 175(9): 5716-23, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16237062

RESUMO

The effects of 17beta-estradiol (E2) on immune function have been extensively reported. The effects are dependent on concentration and duration of exposure and potential differences in signaling between the known E2 receptors, estrogen receptors (ER) alpha and ERbeta. Through the use of ER-deficient mice, we and others have begun to demonstrate the role of the two known receptors in modulating immune functional activities. Previous studies have shown that cells of the innate immune system have altered function (bactericidal capacity) and patterns of cytokine expression (increased proinflammatory cytokine expression) through amelioration of ERalpha signaling. In this study, we extend these studies to analysis of T cell differentiation and proliferation in APC-dependent and APC-independent in vitro assay systems. Our results demonstrate that ERalpha deficiency in splenic macrophages, but not CD11c+ splenic dendritic cells pulsed with OVA significantly enhances proliferative responses and IFN-gamma production by transgenic OVA peptide-specific (OT-II) CD4+ T cells when compared with Ag-pulsed APC from wild-type littermates. The addition of E2 in this culture system did not significantly affect the production of IFN-gamma. In addition, when purified CD4+ T cells from ERalpha-deficient and wild-type littermates were stimulated with anti-CD3/CD28 Ab in the absence of E2, there were no significant differences in IFN-gamma or IL-4 production. However, the addition of E2 significantly increased IL-4 secretion, as well as increased GATA-3 mRNA levels from ERalpha-replete CD4+ T cells, while this effect was abrogated in ERalpha-deficient CD4+ T cells.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Estradiol/farmacologia , Receptor alfa de Estrogênio/fisiologia , Fator de Transcrição GATA3/genética , Interleucina-4/genética , Ativação Linfocitária , Macrófagos/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Citocinas/biossíntese , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Antígenos de Histocompatibilidade Classe II/análise , Interferon gama/biossíntese , Interleucina-4/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise
8.
J Mol Endocrinol ; 35(1): 135-44, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16087727

RESUMO

The IGF-I receptor (IGF-IR) has an important role in breast cancer development and progression. Previous studies have suggested that the IGF-IR gene is negatively regulated by a number of transcription factors with tumor suppressor activity, including the Wilms' tumor protein WT1. The present study was aimed at evaluating the hypothesis that IGF-IR gene transcription in breast cancer cells is under inhibitory control by WT1 and, furthermore, that the mechanism of action of WT1 involves functional and physical interactions with estrogen receptor-alpha (ERalpha). Results of transient coexpression experiments showed that all four predominant isoforms of WT1 (including or lacking alternatively spliced exons 5 and 9) repressed IGF-IR promoter activity by 39-49%. To examine the potential interplay between WT1 and ERalpha in control of IGF-IR gene transcription we employed ER-depleted C4 cells that were generated by clonal selection of ER-positive MCF-7 cells that were maintained in estrogen-free conditions. IGF-IR levels in C4 cells were approximately 43% of the values in MCF-7 cells whereas WT1 levels in C4 cells were 4.25-fold higher than in MCF-7. Triple cotransfection experiments using an ERalpha expression vector in the absence or presence of WT1 expression vectors, along with an IGF-IR promoter reporter plasmid, revealed that ERalpha stimulated IGF-IR promoter activity whereas coexpression of WT1 abrogated the effect of ERalpha. In addition, co-immunoprecipitation experiments demonstrated a specific association between WT1 and ERalpha. Combined, our results suggest that WT1 suppresses IGF-IR gene transcription in breast cancer cells via a mechanism that involves protein-protein association with ERalpha. As a result of this interaction, the ability of ERalpha to transactivate the IGF-IR promoter is abrogated. These findings are consistent with a potential tumor suppressor role for WT1 in breast cancer and suggest that WT1 inactivation in tumoral cells may result in deregulated IGF-IR gene expression and enhanced mitogenic activation by locally produced and/or circulating IGFs.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor IGF Tipo 1/genética , Proteínas WT1/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Feminino , Genes do Tumor de Wilms , Humanos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transcrição Gênica , Proteínas WT1/genética
9.
Cancer Res ; 64(24): 9199-208, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15604293

RESUMO

Estrogen receptor (ER) alpha is mutated (lysine 303 to arginine, K303R) in approximately one third of premalignant breast hyperplasias, which renders breast cancer cells expressing the mutant receptor hypersensitive for proliferation in response to low doses of estrogen. It is known that ERalpha is posttranslationally modified by protein acetylation and phosphorylation by a number of secondary messenger signaling cascades. The K303R ERalpha mutation resides at a major protein acetylation site adjacent to a potential protein kinase A (PKA) phosphorylation site at residue 305 within the hinge domain of the receptor. Mutation of this phosphorylation site to aspartic acid to mimic constitutive phosphorylation blocks acetylation of the K303 ERalpha site and generates an enhanced transcriptional response similar to that seen with the naturally occurring K303R mutant receptor. Activation of PKA signaling by the cell-permeable cyclic AMP (cAMP) analog 8-bromo-cAMP further enhances estrogen sensitivity of the mutant receptor, whereas a specific PKA inhibitor antagonizes this increase. We propose that the hypersensitive ERalpha mutant breast cancer phenotype involves an integration of coupled acetylation and phosphorylation events by upstream signaling molecules.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/fisiologia , Acetilação , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptor alfa de Estrogênio/antagonistas & inibidores , Humanos , Fenótipo , Fosforilação , Transdução de Sinais/fisiologia
10.
Cancer Res ; 64(22): 8184-92, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15548683

RESUMO

Alterations in histones, chromatin-related proteins, and DNA methylation contribute to transcriptional silencing in cancer, but the sequence of these molecular events is not well understood. Here we demonstrate that on disruption of estrogen receptor (ER) alpha signaling by small interfering RNA, polycomb repressors and histone deacetylases are recruited to initiate stable repression of the progesterone receptor (PR) gene, a known ERalpha target, in breast cancer cells. The event is accompanied by acquired DNA methylation of the PR promoter, leaving a stable mark that can be inherited by cancer cell progeny. Reestablishing ERalpha signaling alone was not sufficient to reactivate the PR gene; reactivation of the PR gene also requires DNA demethylation. Methylation microarray analysis further showed that progressive DNA methylation occurs in multiple ERalpha targets in breast cancer genomes. The results imply, for the first time, the significance of epigenetic regulation on ERalpha target genes, providing new direction for research in this classical signaling pathway.


Assuntos
Neoplasias da Mama/metabolismo , Epigênese Genética , Inativação Gênica , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Sequência de Bases , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Primers do DNA , Humanos , Interferência de RNA , Receptores de Progesterona/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Gene ; 328: 143-51, 2004 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-15019994

RESUMO

The human progesterone receptor (PgR) gene has a complex promoter that produces alternate mRNAs encoding the PgRA (94 kDa) and PgRB (120 kDa) protein isoforms. Expression of PgR is induced by estradiol (E(2)) in the breast, reproductive tract and many cell lines despite the lack of a classical estrogen responsive element (ERE) in the promoter regions. We employed chromatin immunoprecipitation (ChIP) to analyze the sites of estrogen receptor alpha (ERalpha) and Sp1 occupancy of the PgR promoters in vivo. We also assessed the functional relevance of histone acetylation levels on the accessibility of transcription factors to the promoter and subsequent hormone-induced transcription. We utilized MCF-7 human breast cancer cells that express PgR in response to E(2) and the MCF-7 derived C4 cell strain that has lost PgR expression as a model system. We found that promoter-wide levels of histone acetylation were not decreased in C4 cells, but that access was partially blocked for Sp1 and completely blocked for ERalpha. The basal level of histone acetylation at six localized regions of the promoter did show some differences between cell lines, but it did not correlate with transcription factor binding. Furthermore, we found only a modest and highly localized change in histone acetylation levels in response to E(2) at only one of three sites of ERalpha binding in MCF-7 cells. This was at the B1 site at the distal 5' end of the promoter. This site also showed a significant decrease in basal histone acetylation in C4 compared to MCF-7 cells. We speculate that the histone acetylation level at this site may be a marker for chromatin structure that affects the access of transcription factors to the whole promoter.


Assuntos
Histonas/metabolismo , Receptores de Progesterona/genética , Fatores de Transcrição/metabolismo , Acetilação/efeitos dos fármacos , Sequência de Bases , Sítios de Ligação/genética , Western Blotting , Linhagem Celular Tumoral , Estradiol/farmacologia , Receptor alfa de Estrogênio , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Isoformas de Proteínas/genética , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/fisiologia , Elementos de Resposta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fator de Transcrição Sp1/metabolismo , Sítio de Iniciação de Transcrição
12.
J Leukoc Biol ; 75(6): 1166-72, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15020652

RESUMO

In this series of studies, we determined the potential role of intracellular estrogen receptors (ER), ERalpha and ERbeta, on macrophage function in response to bacterial stimuli. The sex hormone 17beta-estradiol (E(2)) and ER have been shown to modulate inflammatory responses as well as T helper cell type 1 (TH1)/TH2 responses. The mechanisms E(2) and its receptors use to alter these immune functions remain largely unknown. ERalpha and ERbeta possess complex actions in tissues where they are expressed. We have characterized the receptor repertoire of murine dendritic cells and thioglycollate-elicited peritoneal macrophages (PM). Both cell types express mRNA for ERalpha. Neither cell type expressed detectable amounts of ERbeta mRNA, as determined by reverse transcriptase-polymerase chain reaction using exon-specific primers spanning each of the seven intron/exon junctions. Primary macrophages from ERalpha- and ERbeta-deficient severe combined immunodeficiency mice [ERalpha knockout (KO) and ERssKO, respectively] were used to delineate the effects and potential mechanisms via which steroid receptors modulate macrophage function. ERalpha-deficient PM exposed ex vivo to lipopolysaccharide or Mycobacterium avium exhibited significant increases in tumor necrosis factor alpha (TNF-alpha) secretion as well as reduction in bacterial load when compared with wild-type (WT) PM. In contrast, ERbeta-deficient PM possessed no significant difference in TNF-alpha secretion or in bacterial load when compared with WT littermates. These studies suggest that ERalpha, but not ERbeta, modulates murine PM function.


Assuntos
Macrófagos Peritoneais/metabolismo , Mycobacterium avium/fisiologia , Receptores de Estrogênio/deficiência , Fator de Necrose Tumoral alfa/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Técnicas In Vitro , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID
13.
Cancer Biol Ther ; 3(5): 460-7, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15020841

RESUMO

A retrospective analysis of CALGB trial 9344 suggested paclitaxel administration following cyclophosphamide and doxorubicin adjuvant chemotherapy is most beneficial for patients with ERalpha negative (ERalpha-) breast cancer. Since the cytotoxic effects of paclitaxel are cell cycle dependent, we postulated that the relationship between ERalpha and the effectiveness of adjuvant paclitaxel reflects the observation that ERalpha positive (ERalpha+) breast cancers proliferate more slowly than ERalpha- breast cancers. Three in vitro models (MCF-7, T47D and ZR-75) were examined to compare growth rates and paclitaxel-induced apoptosis in ERalpha+ and ERalpha- clones of the same, originally ERalpha+ cell line. For the T47D and ZR-75 cell lines, loss of ERalpha was associated with a decrease in doubling time and an increase in paclitaxel sensitivity. However, when cell culture conditions were altered to achieve equivalent cell proliferation rates, no difference in paclitaxel sensitivity was observed. Similarly, an ERalpha- clone of MCF-7 cells that did not exhibit an enhanced growth rate compared to its ERalpha+ counterpart also did not show increased paclitaxel sensitivity. The combined apoptotic effects of tamoxifen and paclitaxel on MCF-7 cells were not synergistic or even clearly additive. In these in vitro models, the effectiveness of paclitaxel correlated more closely with growth rate than ERalpha expression. These data suggest that measurements of tumor proliferation may provide more accurate predictive markers for the benefits of adjuvant paclitaxel than ERalpha analysis.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Receptor alfa de Estrogênio/metabolismo , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Paclitaxel/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimioterapia Adjuvante , Meios de Cultura Livres de Soro , Resistencia a Medicamentos Antineoplásicos , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hormônio-Dependentes/metabolismo , Receptores de Esteroides/metabolismo , Células Tumorais Cultivadas
14.
Am J Physiol Heart Circ Physiol ; 285(5): H2150-7, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12881205

RESUMO

Estrogen has been shown to increase endothelium-dependent vasodilation and expression of endothelial nitric oxide (NO) synthase (eNOS); however, the role of estrogen receptors in mediating estrogen effects on endothelial function remains to be elucidated. The purpose of this study was to test the hypothesis that estrogen modulates NO-dependent vasodilation of coronary arteries through its action on estrogen receptor-alpha (ER-alpha) to increase protein levels of eNOS and Cu/Zn superoxide dismutase (SOD-1). Vasodilation to acetylcholine (ACh) and sodium nitroprusside was assessed in isolated coronary arteries from intact and ovariectomized female wild-type (WT) and ER-alpha knockout (ERalphaKO) mice. Protein levels for eNOS and SOD-1 were also evaluated. Vasodilation to ACh was not significantly altered in ERalphaKO mice compared with WT mice. Ovariectomy reduced responsiveness to ACh in ERalphaKO mice but not WT mice. Responses to sodium nitroprusside were not altered by disruption of ER-alpha or by ovariectomy. Supplementation with estrogen restored ACh-induced vasodilation in ovariectomized ERalphaKO mice. eNOS protein was reduced in ERalphaKO mice compared with WT mice. Ovariectomy caused a further reduction in eNOS protein in ERalphaKO mice, but this reduction was reversed by estrogen treatment. SOD-1 protein levels were increased by disruption of ER-alpha. Ovariectomy reduced SOD-1 protein in ERalphaKO mice, but this reduction was partially reversed by estrogen replacement. These results suggest that estrogen modulation of eNOS protein content is mediated in part through ER-alpha. NO-dependent responses are preserved in ERalphaKO mice, possibly through increased SOD-1 expression and enhanced bioavailability of NO.


Assuntos
Vasos Coronários/fisiologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/metabolismo , Receptores de Estrogênio/genética , Vasodilatação/fisiologia , Acetilcolina/farmacologia , Animais , Vasos Coronários/efeitos dos fármacos , Receptor alfa de Estrogênio , Estrogênios/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Nitroprussiato/farmacologia , Ovariectomia , Receptores de Estrogênio/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
15.
Environ Health Perspect ; 111(8): 994-1006, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12826473

RESUMO

Information concerning the fundamental mechanisms of action of both natural and environmental hormones, combined with information concerning endogenous hormone concentrations, reveals how endocrine-disrupting chemicals with estrogenic activity (EEDCs) can be active at concentrations far below those currently being tested in toxicological studies. Using only very high doses in toxicological studies of EEDCs thus can dramatically underestimate bioactivity. Specifically: a) The hormonal action mechanisms and the physiology of delivery of EEDCs predict with accuracy the low-dose ranges of biological activity, which have been missed by traditional toxicological testing. b) Toxicology assumes that it is valid to extrapolate linearly from high doses over a very wide dose range to predict responses at doses within the physiological range of receptor occupancy for an EEDC; however, because receptor-mediated responses saturate, this assumption is invalid. c) Furthermore, receptor-mediated responses can first increase and then decrease as dose increases, contradicting the assumption that dose-response relationships are monotonic. d) Exogenous estrogens modulate a system that is physiologically active and thus is already above threshold, contradicting the traditional toxicological assumption of thresholds for endocrine responses to EEDCs. These four fundamental issues are problematic for risk assessment methods used by regulatory agencies, because they challenge the traditional use of extrapolation from high-dose testing to predict responses at the much lower environmentally relevant doses. These doses are within the range of current exposures to numerous chemicals in wildlife and humans. These problems are exacerbated by the fact that the type of positive and negative controls appropriate to the study of endocrine responses are not part of traditional toxicological testing and are frequently omitted, or when present, have been misinterpreted.


Assuntos
Sistema Endócrino/efeitos dos fármacos , Exposição Ambiental , Poluentes Ambientais/toxicidade , Estrogênios/toxicidade , Receptores de Estrogênio/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Receptores de Estrogênio/fisiologia , Reprodutibilidade dos Testes , Projetos de Pesquisa , Medição de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...