Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biol Reprod ; 92(5): 122, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25882702

RESUMO

Progesterone (P4) and the synthetic glucocorticoid dexamethasone (Dex) inhibit luminal epithelial (LE) proliferation in neonatal mouse uteri. This study determined the roles of progesterone receptor and estrogen receptor 1 (PR and ESR1, respectively) in P4- and Dex-induced inhibition of LE proliferation using PR knockout (PRKO) and Esr1 knockout (Esr1KO) mice. Wild-type (WT), heterozygous, and homozygous PRKO female pups were injected with vehicle, P4 (40 µg/g body weight), or Dex (4 or 40 µg/g body weight) on Postnatal Day 5, then 24 h later immunostained for markers of cell proliferation. In WT and heterozygous mice, P4 sharply reduced LE proliferation, and Dex produced dose-responsive decreases equaling those of P4 at the higher dose. Critically, although both doses of Dex similarly decreased proliferation compared to vehicle-treated PRKOs, treatment of PRKO pups with the high Dex dose (40 µg/g) did not inhibit LE as much as treatments of WT mice with this Dex dose or with P4. Stromal proliferation was stimulated by P4 in WT but not PRKO mice, and Dex did not alter stromal proliferation. Uteri of all genotypes strongly expressed glucocorticoid receptor (GR), demonstrating that impaired Dex effects in PRKOs did not reflect GR deficiency. Furthermore, inhibition of LE proliferation by Dex (40 µg/g body weight) in Esr1KO mice was normal, so this process does not involve ESR1. In summary, inhibitory Dex effects on LE proliferation occur partially through non-PR-mediated mechanisms, presumably GR, as indicated by Dex inhibition of LE proliferation in PRKOs. However, maximal inhibitory Dex effects on uterine LE proliferation are not seen in PRKO mice with even high Dex, indicating that maximal Dex effects in WT mice also involve PR.


Assuntos
Dexametasona/farmacologia , Células Epiteliais/efeitos dos fármacos , Progesterona/farmacologia , Receptores de Progesterona/metabolismo , Útero/fisiologia , Animais , Animais Recém-Nascidos , Células Epiteliais/fisiologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Receptores de Progesterona/genética , Útero/citologia , Útero/efeitos dos fármacos
2.
Gynecol Oncol ; 131(1): 198-206, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23851291

RESUMO

OBJECTIVE: The objective of this study is to determine whether an altered DNA replication process is responsible for some of genetic damage observed in ovarian cancer. METHODS: The replication fidelity of the DNA synthetic process was evaluated in both malignant and non-malignant human ovarian cells. The types of replication errors produced were identified. In addition, kinetic analyses of the efficiency of ovarian cancer DNA polymerases for misincorporating nucleotides were performed. RESULTS: We report for the first time that ovarian cancer cells harbor an error promoting DNA replication apparatus which contributes to the decrease in DNA synthetic fidelity exhibited by these cells. Our study also shows that the decrease in DNA replication fidelity was not a result of an increased DNA replication activity. In addition, it was observed that the higher rate of DNA replication errors does not result in significant differences in the type of DNA replication-errors made during the DNA replication process; just the relative abundance. A detailed kinetic analysis of the efficiency of misincorporating nucleotides demonstrated that the DNA polymerases within the ovarian cancer cells exhibited a significant propensity for creating purine-pyrimidine nucleotide mismatches relative to non-malignant ovarian cells, while being only slightly more efficient at incorrectly pairing a purine nucleotide with a purine nucleotide. CONCLUSIONS: All together, these data suggest that the systematic analysis of the DNA replication process in ovarian cancer could uncover information on some of the molecular mechanisms that drive the accumulation of genetic damage, and probably contribute to the pathogenesis of the disease.


Assuntos
Carcinoma/genética , Replicação do DNA , DNA de Neoplasias/biossíntese , DNA Polimerase Dirigida por DNA , Complexos Multienzimáticos , Mutação , Neoplasias Ovarianas/genética , Linhagem Celular Tumoral , DNA Polimerase Dirigida por DNA/metabolismo , Nucleotídeos de Desoxiadenina/metabolismo , Feminino , Humanos , Cinética , Óperon Lac/genética , Ovário/citologia
3.
Carcinogenesis ; 32(8): 1162-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21606321

RESUMO

Mice treated neonatally with diethylnitrosamine (DEN) develop liver tumors in a male-dominant manner, reflecting the male bias in human hepatocellular carcinoma. Evidence suggests that estrogen, androgen, prolactin (PRL) and growth hormone (GH) modify liver tumorigenesis. We determined the roles of estrogen receptor-α (ERα) and prolactin receptor (PRLR) using receptor null mice, ERαKO (C57Bl/6J) and PRLR-KO (129Ola-X-C57BL/6), in the neonatal-DEN model of liver tumorigenesis. In both mouse strains, females had reduced tumorigenesis compared with males (P < 0.01), regardless of ERα or PRLR status. Tumorigenesis was not affected by ovariectomy in C57Bl/6J mice but it was increased by ovariectomy in the mixed strain, 129Ola-X-C57BL/6, regardless of PRLR status. ERαKO males had 47% fewer tumors than ERα wild-type males (P < 0.01). On the other hand, estradiol treatment protected against tumorigenesis in males only in the presence of ERα. As evidenced by liver gene expression, lack of ERα did not alter the pattern of GH secretion in males but resulted in the male GH pattern in females. These observations indicate that ERα is not required for lower tumorigenesis in females, but it is required for the protective effects of exogenously delivered estradiol. Unexpectedly, the results indicate that ERα plays a role in promotion of liver tumors in males. In addition, it can be concluded that sex differences in liver tumorigenesis cannot be explained by the sexually dimorphic pattern of GH secretion. The results also rule out PRL as the mediator of the protective effect of the ovaries.


Assuntos
Dietilnitrosamina/toxicidade , Receptor alfa de Estrogênio/fisiologia , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Receptores da Prolactina/fisiologia , Animais , Estradiol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento/metabolismo , Humanos , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovariectomia , Prolactina/farmacologia
4.
Radiat Res ; 194(5): 557-565, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33045089

RESUMO

Astronauts participating in prolonged space missions constitute a population of individuals who are at an increased risk for cataractogenesis due to exposure to densely ionizing charged particles. Using a rat model, we have previously shown that after irradiation of eyes with either low-linear energy transfer (LET) 60Co γ rays or high-LET 56Fe particles, the rate of progression of anterior and posterior subcapsular cataracts was significantly greater in ovariectomized females implanted with 17-ß-estradiol (E2) compared to ovariectomized or intact rats. However, our additional low-LET studies indicated that cataractogenesis may be a modifiable late effect, since we have shown that the modulation of cataractogenesis is dependent upon the timing of administration of E2. Interestingly, we found that E2 protected against cataractogenesis induced by low-LET radiation, but only if administered after the exposure; if administered prior to and after irradiation, for the entire period of observation, then E2 enhanced progression and incidence of cataracts. Since most radioprotectors tested to date are unsuccessful in protecting against the effects of high-LET radiation, we wished to determine whether the protection mediated by E2 against radiation cataractogenesis induced by low-LET radiation would also be observed after high-LET irradiation. Female 56-day-old Sprague-Dawley rats were treated with E2 at various times relative to the time of single-eye irradiation with 2 Gy of 56Fe ions. We found that administration of E2 before irradiation and throughout the lifetime of the rat enhanced cataractogenesis compared to ovariectomized animals. The enhancing effect was slightly reduced when estrogen was removed after irradiation. However, in contrast to what we observed after γ-ray irradiation, there was no inhibition of cataractogenesis if E2 was administered only after 56Fe-ion irradiation. We conclude that protection against cataractogenesis by estrogen is dependent upon the type and ionization density of radiation that the lens was exposed to. The lack of inhibition of radiation cataractogenesis in rats that receive E2 treatment after high-LET irradiation may be attributed to the qualitative differences in the types of DNA damage induced with high-LET radiation compared to low-LET radiation or how damage may be modified at the DNA or tissue level after irradiation.


Assuntos
Catarata/prevenção & controle , Radioisótopos de Cobalto , Estradiol/uso terapêutico , Raios gama/efeitos adversos , Íons Pesados/efeitos adversos , Ferro , Lesões Experimentais por Radiação/prevenção & controle , Medicina Aeroespacial , Animais , Catarata/etiologia , Esquema de Medicação , Implantes de Medicamento , Estradiol/administração & dosagem , Incidência , Transferência Linear de Energia , Ovariectomia , Ratos , Ratos Sprague-Dawley
5.
Environ Health Perspect ; 117(1): 93-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19165393

RESUMO

BACKGROUND: A previous study from our laboratory showed that polybrominated diphenyl ethers (PBDEs) were metabolized to hydroxylated PBDEs (HO-PBDEs) in mice and that para-HO-PBDEs were the most abundant and, potentially, the most toxic metabolites. OBJECTIVE: The goal of this study was to determine the concentrations of HO-PBDEs in blood from pregnant women, who had not been intentionally or occupationally exposed to these flame retardants, and from their newborn babies. METHODS: Twenty human blood samples were obtained from a hospital in Indianapolis, Indiana, and analyzed for both PBDEs and HO-PBDEs using electron-capture negative-ionization gas chromatographic mass spectrometry. RESULTS: The metabolite pattern of HO-PBDEs in human blood was quite different from that found in mice; 5-HO-BDE-47 and 6-HO-BDE-47 were the most abundant metabolites of BDE-47, and 5'-HO-BDE-99 and 6'-HO-BDE-99 were the most abundant metabolites of BDE-99. The relative concentrations between precursor and corresponding metabolites indicated that BDE-99 was more likely to be metabolized than BDE-47 and BDE-100. In addition, three bromophenols were also detected as products of the cleavage of the diphenyl ether bond. The ratio of total hydroxylated metabolites relative to their PBDE precursors ranged from 0.10 to 2.8, indicating that hydroxylated metabolites of PBDEs were accumulated in human blood. CONCLUSIONS: The quite different PBDE metabolite pattern observed in humans versus mice indicates that different enzymes might be involved in the metabolic process. Although the levels of HO-PBDE metabolites found in human blood were low, these metabolites seemed to be accumulating.


Assuntos
Bifenil Polibromatos/sangue , Animais , Éteres , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Hidroxilação , Camundongos , Estados Unidos
6.
Radiat Res ; 172(1): 129-33, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19580515

RESUMO

Radiation cataractogenesis is an important consideration for radiotherapy patients and for astronauts. Data in the literature suggest that gender and/or estrogen may play a role in the incidence of age-related cataracts. However, few data exist on the effect of gender on radiation-induced cataractogenesis. We compared the incidence and rate of progression of cataracts induced by ionizing radiation in male and female Sprague-Dawley rats. Male rats were implanted with either an empty silastic capsule or a capsule containing 17-beta-estradiol. Ovary-intact female rats were implanted with empty capsules. All rats received a single dose of 10 Gy (60Co gamma rays) to the right eye only. Lens opacification was measured at 2-4-week intervals with a slit lamp. The incidence of radiation-induced cataracts was significantly increased in male rats compared to female rats (P=0.034). There was no difference in the rate of cataract progression between the three groups. Our data suggest there is a gender-related difference in radiation-induced cataractogenesis, but the increased incidence of radiation cataractogenesis in male rats compared to female rats cannot be attributed to estrogen levels, since there was no difference in cataract incidence between male rats implanted with empty capsules and those implanted with capsules containing 17-beta-estradiol.


Assuntos
Catarata/etiologia , Catarata/patologia , Estradiol/metabolismo , Lesões Experimentais por Radiação/complicações , Lesões Experimentais por Radiação/patologia , Caracteres Sexuais , Animais , Progressão da Doença , Feminino , Raios gama/efeitos adversos , Cristalino/patologia , Cristalino/efeitos da radiação , Modelos Lineares , Masculino , Ratos , Ratos Sprague-Dawley
7.
Mol Cancer Ther ; 7(7): 2096-108, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18645020

RESUMO

Breast cancer resistance to the antiestrogens tamoxifen (OHT) and fulvestrant is accompanied by alterations in both estrogen-dependent and estrogen-independent signaling pathways. Consequently, effective inhibition of both pathways may be necessary to block proliferation of antiestrogen-resistant breast cancer cells. In this study, we examined the effects of apigenin, a dietary plant flavonoid with potential anticancer properties, on estrogen-responsive, antiestrogen-sensitive MCF7 breast cancer cells and two MCF7 sublines with acquired resistance to either OHT or fulvestrant. We found that apigenin can function as both an estrogen and an antiestrogen in a dose-dependent manner. At low concentrations (1 mumol/L), apigenin stimulated MCF7 cell growth but had no effect on the antiestrogen-resistant MCF7 sublines. In contrast, at high concentrations (>10 mumol/L), the drug inhibited growth of MCF7 cells and the antiestrogen-resistant sublines, and the combination of apigenin with either OHT or fulvestrant showed synergistic, growth-inhibitory effects on both antiestrogen-sensitive and antiestrogen-resistant breast cancer cells. To further elucidate the molecular mechanism of apigenin as either an estrogen or an antiestrogen, effects of the drug on estrogen receptor-alpha (ERalpha); transactivation activity, mobility, stability, and ERalpha-coactivator interactions were investigated. Low-dose apigenin enhanced receptor transcriptional activity by promoting interaction between ERalpha and its coactivator amplified in breast cancer-1. However, higher doses (>10 mumol/L) of apigenin inhibited ERalpha mobility (as determined by fluorescence recovery after photobleaching assays), down-regulated ERalpha and amplified in breast cancer-1 expression levels, and inhibited multiple protein kinases, including p38, protein kinase A, mitogen-activated protein kinase, and AKT. Collectively, these results show that apigenin can function as both an antiestrogen and a protein kinase inhibitor with activity against breast cancer cells with acquired resistance to OHT or fulvestrant. We conclude that apigenin, through its ability to target both ERalpha-dependent and ERalpha-independent pathways, holds promise as a new therapeutic agent against antiestrogen-resistant breast cancer.


Assuntos
Apigenina/farmacologia , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Moduladores de Receptor Estrogênico/farmacologia , Receptor alfa de Estrogênio/metabolismo , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Feminino , Recuperação de Fluorescência Após Fotodegradação , Fulvestranto , Genes Reporter , Histona Acetiltransferases/metabolismo , Humanos , Hidroxitestosteronas/farmacologia , Coativador 3 de Receptor Nuclear , Ligação Proteica/efeitos dos fármacos , Proteínas Quinases/metabolismo , Transporte Proteico/efeitos dos fármacos , Transativadores/metabolismo , Transcrição Gênica/efeitos dos fármacos
8.
Mol Cancer Ther ; 7(7): 1993-2002, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18645009

RESUMO

Epithelial ovarian cancer (EOC) arises from the epithelial layer covering the surface of ovaries and i.p. metastasis is commonly observed at diagnosis. Sphingosine-1-phosphate (S1P), a bioactive lipid signaling molecule, is potentially involved in EOC tumorigenesis. We have found that S1P is elevated in human EOC ascites. We show that physiologically relevant concentrations of S1P stimulate migration and invasion of EOC cells but inhibit migration of human ovarian surface epithelial (HOSE) cells. In addition, S1P inhibits lysophosphatidic acid (LPA)-induced cell migration in HOSE but not in EOC cells. We have provided the first line of evidence that the expression levels of S1P receptor subtypes are not the only determinants for how cells respond to S1P. Although S1P(1) is expressed and functional in HOSE cells, the inhibitory effect mediated by S1P(2) is dominant in those cells. The cellular preexisting stress fibers are also important determinants for the migratory response to S1P. Differential S1P-induced morphology changes are noted in EOC and HOSE cells. Preexisting stress fibers in HOSE cells are further enhanced by S1P treatment, resulting in the negative migratory response to S1P. By contrast, EOC cells lost stress fibers and S1P treatment induces filopodium-like structures at cell edges, which correlates with increased cell motility. In addition, inhibition of the protein kinase C pathway is likely to be involved in the inhibitory effect of S1P on LPA-induced cell migration in HOSE cells. These findings are important for the development of new therapeutics targeting S1P and LPA in EOC.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Epiteliais/patologia , Lisofosfolipídeos/farmacologia , Neoplasias Ovarianas/patologia , Esfingosina/análogos & derivados , Ascite/patologia , Linhagem Celular Tumoral , Forma Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Feminino , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Neoplasias Ovarianas/enzimologia , Proteína Quinase C/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/farmacologia , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/patologia
9.
Endocr Relat Cancer ; 15(2): 475-83, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18509000

RESUMO

Numerous epidemiological observations point to sex differences in lung cancer etiology and progression. The present study was aimed at understanding the bases of these sex differences. To test the effect of estradiol on tumor progression, we used a mouse model based on conditional Kras expression and concurrent deletion of Tp53 following inhalation of an adenoviral vector expressing Cre recombinase (AdeCre). Ovariectomized females and males were treated with estradiol via a continuous-release capsule. Tumor multiplicity, tumor volume, and histological grade were determined at 10 weeks after AdeCre administration. Cell proliferation was monitored by Ki67 immunohistochemistry at 4 and 10 weeks after AdeCre administration. At 10 weeks, female mice had more than twice the number of tumors evident on the surface of the lungs than male mice; ovariectomy eliminated this sex difference. The estrogen treatment significantly increased tumor number and volume in ovariectomized females and in males. Histological character of the tumors ranged from adenoma to adenocarcinoma. Ovary-intact females exhibited higher grade tumors than ovariectomized females or males. Progression to higher histological grade was stimulated by estrogen in male mice but not in ovariectomized females. At 10 weeks after AdeCre administration, tumor cell Ki67-labeling varied widely, precluding assessment of an estrogen effect; however, at 4 weeks, Ki67 labeling of lung parenchymal cells was increased 3.5-fold by estrogen. In conclusion, estrogen acts as a promoter for lung adenocarcinoma in a genetically defined lung cancer model; estrogen-induced cell proliferation in the oncogene-initiated cells is likely to play a role in this tumor promoter activity.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Estrogênios/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Feminino , Immunoblotting , Integrases/genética , Masculino , Camundongos , Camundongos Transgênicos , Ovariectomia , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores de Estrogênio/metabolismo , Caracteres Sexuais , Proteína Supressora de Tumor p53/genética
10.
Environ Health Perspect ; 116(10): 1315-21, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18941571

RESUMO

BACKGROUND: Polybrominated diphenyl ethers (PBDEs) are widely found in the environment and are suspected endocrine disruptors. We previously identified six hydroxylated metabolites of PBDE (OH-PBDEs) in treated mice. OBJECTIVE: We tested the hypothesis that OH-PBDEs would interact with and alter activity of estrogen receptor-alpha (ER-alpha). METHODS: We tested estrogenicity using two assays: 3H-estradiol (3H-E2) displacement from recombinant ER-alpha and induction of reporter gene (ERE-luciferase) in cultured cells. We incubated the PBDE mixture DE-71 with rat liver microsomes and tested the resultant metabolite mixture for estrogenic activity. We also determined relative estrogenic potential of individual hydroxylated PBDE congeners. RESULTS: Reporter gene activity was increased by DE-71 that had been subjected to microsomal metabolism. DE-71 did not displace E2 from ER-alpha, but all six of the OH-PBDE metabolites did. para-Hydroxylated metabolites displayed a 10- to 30-fold higher affinity for ER-alpha compared with ortho-hydroxylated PBDEs, and one produced a maximal effect 30% higher than that produced by E2. Coadministration of E2 and DE-71, or certain of its metabolites, yielded reporter activity greater than either chemical alone. Two ortho-OH-PBDEs were antiestrogenic in the reporter assay. CONCLUSIONS: The observations--that the DE-71 mixture did not displace 3H-E2 from ER-alpha while the hydroxylated metabolites did-suggest that the weak estrogenic effects of DE-71 are due to metabolic activation of individual congeners. However, the behavior of DE-71 and its metabolites, when co-administered with E2, suggest a secondary, undetermined mechanism from classical ER-alpha activation.


Assuntos
Receptor alfa de Estrogênio/efeitos dos fármacos , Éteres Fenílicos/toxicidade , Bifenil Polibromatos/toxicidade , Animais , Biotransformação , Receptor alfa de Estrogênio/metabolismo , Genes Reporter , Éteres Difenil Halogenados , Hidroxilação , Ligantes , Microssomos Hepáticos/efeitos dos fármacos , Éteres Fenílicos/metabolismo , Éteres Fenílicos/farmacocinética , Bifenil Polibromatos/metabolismo , Bifenil Polibromatos/farmacocinética , Ratos , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo
11.
Environ Health Perspect ; 116(5): 605-11, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18470304

RESUMO

BACKGROUND: Polybrominated diphenyl ethers (PBDEs) are widely found in the environment, and they may act as endocrine disruptors. OBJECTIVE: Our goal in this study was to test the PBDE mixture DE-71 for estrogenic activity. METHODS: We used proliferation of cultured breast cancer cells (MCF-7) and trophic effects in the reproductive tracts of ovariectomized mice as estrogen bioassays. DE-71 was administered to mice by subcutaneous injection (sc) or oral gavage (po), alone or in combination with estradiol, for 3 or 34 days. Liver weights and cytochrome P450 enzyme activities were also measured. RESULTS: DE-71 increased MCF-7 cell proliferation, and this was prevented by antiestrogen. DE-71 cotreatment reduced the effect of estradiol in MCF-7 cells. In the mouse 3-day assay, DE-71 administered alone had no effect on uterine weight, uterine epithelial height (UEH), or vaginal epithelial thickness (VET); however, when DE-71 was administered as a cotreatment, it potentiated estradiol's effect on uterine weight. DE-71 administered sc to BALB/c mice for 34 days slightly increased UEH and VET, and attenuated the estradiol-induced increase in UEH; these effects were not seen in BALB/c mice treated po or in C57BL/6 mice treated sc. DE-71 increased liver weight in BALB/c, C57BL/6, and estrogen receptor-alpha knockout mice. We also found an increase in liver cytochrome P450 1A (CYP1A) and CYP2B activities when DE-71 was administered po, but only CYP2B increased after sc treatment. CONCLUSION: DE-71 behaves as a weak estrogen. In mice, the treatment route and duration determined if DE-71 was estrogenic. BALB/c mice are more susceptible to DE-71 effects in estrogen target tissues than C57BL/6 mice. DE-71 increased liver weight independently of estrogen receptor-alpha.


Assuntos
Moduladores de Receptor Estrogênico/farmacologia , Éteres Fenílicos/farmacologia , Bifenil Polibromatos/farmacologia , Animais , Bioensaio , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/análise , Disruptores Endócrinos , Estradiol/administração & dosagem , Feminino , Éteres Difenil Halogenados , Fígado/citologia , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovariectomia , Éteres Fenílicos/administração & dosagem , Bifenil Polibromatos/administração & dosagem
12.
Radiat Res ; 170(6): 758-64, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19138041

RESUMO

Cataractogenesis is a complication of radiotherapy when the eye is included in the treatment field. Low doses of densely ionizing space radiation may also result in an increased risk of cataracts in astronauts. We previously reported that estrogen (17-beta-estradiol), when administered to ovariectomized rats commencing 1 week before gamma irradiation of the eye and continuously thereafter, results in a significant increase in the rate and incidence of cataract formation and a decreased latent period compared to an ovariectomized control group. We therefore concluded that estrogen accelerates progression of radiation-induced opacification. We now show that estrogen, if administered continuously, but commencing after irradiation, protects against radiation cataractogenesis. Both the rate of progression and incidence of cataracts were greatly reduced in ovariectomized rats that received estrogen treatment after irradiation compared to ovariectomized rats. As in our previous study, estradiol administered 1 week prior to irradiation at the time of ovariectomy and throughout the period of observation produced an enhanced rate of cataract progression. Estrogen administered for only 1 week prior to irradiation had no effect on the rate of progression but resulted in a slight reduction in the incidence. We conclude that estrogen may enhance or protect against radiation cataractogenesis, depending on when it is administered relative to the time of irradiation, and may differentially modulate the initiation and progression phases of cataractogenesis. These data have important implications for astronauts and radiotherapy patients.


Assuntos
Catarata/etiologia , Catarata/prevenção & controle , Estrogênios/farmacologia , Lesões Experimentais por Radiação/prevenção & controle , Animais , Catarata/patologia , Estrogênios/administração & dosagem , Olho/efeitos dos fármacos , Olho/patologia , Olho/efeitos da radiação , Feminino , Doses de Radiação , Lesões Experimentais por Radiação/patologia , Radioterapia (Especialidade) , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
13.
Environ Health Perspect ; 115(7): 1052-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17637922

RESUMO

BACKGROUND: Previous studies have shown that polybrominated diphenyl ethers (PBDEs) behave as weak estrogens in animal and cell culture bioassays. In vivo metabolites of PBDEs are suspected to cause these effects. OBJECTIVES: To identify candidate metabolites, mouse plasma samples were collected after continuous oral and subcutaneous exposure to DE-71, a widely used commercial pentabromodiphenyl ether product, for 34 days. METHODS: Samples were extracted, separated into neutral and phenolic fractions, and analyzed by gas chromatographic mass spectrometry. RESULTS: In the plasma samples of orally treated animals, 2,2',4,4',5,5'-hexabromodiphenyl ether (BDE-153) represented 52% of total measurable PBDEs, whereas it represented only 4.3% in the DE-71 mixture. This suggested that BDE-153 was more persistent than other congeners in mice. Several metabolites were detected and quantitated: 2,4-dibromophenol, 2,4,5-tribromophenol, and six hydroxylated PBDEs. The presence of the two phenols suggested cleavage of the ether bond of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) and 2,2',4,4',5-pentabromodiphenyl ether (BDE-99), respectively. The hydroxylated (HO)-PBDEs might come from hydroxylation or debromination/hydroxylation. Among the quantitated hydroxylated metabolites, the most abundant was 4-HO-2,2',3,4'-tetra-BDE, which suggested that there was a bromine shift during the hydroxylation process. para-HO-PBDEs have been proposed to behave as endocrine disruptors. CONCLUSIONS: THERE SEEM TO BE THREE METABOLIC PATHWAYS: cleavage of the diphenyl ether bond, hydroxylation, and debromination/hydroxylation. The cleavage of the diphenyl ether bond formed bromophenols, and the other two pathways formed hydroxylated PBDEs, of which para-HO-PBDEs are most likely formed from BDE-47. These metabolites may be the most thyroxine-like and/or estrogen-like congeners among the HO-PBDEs.


Assuntos
Bifenil Polibromatos/sangue , Animais , Cromatografia Gasosa-Espectrometria de Massas , Hidroxilação , Camundongos
14.
Chem Biol ; 13(8): 891-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16931338

RESUMO

Bisphenol A (BPA) is a widely used plasticizer whose estrogenic properties may impact hormone-responsive disorders and fetal development. In vivo, BPA appears to have greater activity than is suggested by its estrogen receptor (ER) binding affinity. This may be a result of BPA sulfation/desulfation providing a pathway for selective uptake into hormone-responsive cells. BPA is a substrate for estrogen sulfotransferase, and bisphenol A sulfate (BPAS) and disulfate are substrates for estrone sulfatase. Although the sulfated xenobiotics bind poorly to the ER, both stimulated the growth of receptor-positive breast tumor cells. Treatment of MCF-7 cells with BPAS leads to desulfation and uptake of BPA. No BPAS is found inside the cells. These findings suggest a mechanism for the selective uptake of BPA into cells expressing estrone sulfatase. Therefore, sulfation may increase the estrogenic potential of xenobiotics.


Assuntos
Neoplasias da Mama/metabolismo , Fenóis/farmacocinética , Sulfotransferases/efeitos dos fármacos , Óxidos de Enxofre/química , Ésteres do Ácido Sulfúrico/farmacocinética , Compostos Benzidrílicos , Sítios de Ligação , Catálise , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Espectroscopia de Ressonância Magnética/métodos , Estrutura Molecular , Fenóis/síntese química , Fenóis/química , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Sensibilidade e Especificidade , Sulfotransferases/química , Sulfotransferases/metabolismo , Ésteres do Ácido Sulfúrico/síntese química , Ésteres do Ácido Sulfúrico/química
15.
Radiat Res ; 165(1): 9-15, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16392957

RESUMO

Cataractogenesis is a widely reported late effect that is observed in patients receiving total-body irradiation (TBI) prior to bone marrow transplantation or radiotherapy for ocular or head and neck cancers. Recent studies indicate that estrogens may protect against age-related and drug-induced cataracts. Moreover, other reports suggest that estrogen possesses antioxidant properties. Since the effect of estrogen on radiation cataractogenesis is unknown, we wished to determine whether estrogen modulates radiation-induced opacification of the lens. Intact or ovariectomized Sprague-Dawley rats were treated with either 17-beta-estradiol or an empty silastic capsule. The right orbit was then irradiated with either 10 or 15 Gy of (60)Co gamma rays using a Leksell Gamma Knife, and lenses were examined at various times postirradiation with a slit lamp or evaluated for light transmission. We found that for ovariectomized rats irradiated with 15 Gy, the lens opacity and the incidence of cataract formation in the estradiol-treated group were significantly increased compared to the control group at the end of the 25-week period of observation. Cataract incidence was also high in irradiated eyes of ovary-intact animals at 25 weeks postirradiation but was greatly reduced in the ovariectomized control group, with less than half of irradiated eyes showing evidence of cataractogenesis. Thus, after irradiation with 15 Gy of gamma rays, estrogen increased the incidence of cataract formation. We also observed that although the incidence of cataract formation in rats irradiated with 10 Gy and receiving continuous estrogen treatment was not altered compared to rats in the control group that did not receive estrogen, the latent period for posterior subcapsular cataract formation decreased and the severity of the anterior cataract increased. Taken together, our data suggest that estrogen accelerates progression of radiation-induced opacification.


Assuntos
Catarata/patologia , Catarata/prevenção & controle , Estrogênios/administração & dosagem , Lesões por Radiação/patologia , Lesões por Radiação/prevenção & controle , Radiocirurgia/efeitos adversos , Animais , Catarata/etiologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Estrogênios/metabolismo , Feminino , Raios gama/efeitos adversos , Ovariectomia , Lesões por Radiação/etiologia , Tolerância a Radiação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
16.
Mol Endocrinol ; 17(3): 356-65, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12554766

RESUMO

Steroid hormone receptors, including estrogen receptor-alpha (ERalpha), are ligand-activated transcription factors, and hormone binding leads to depletion of receptor levels via preteasome-mediated degradation. NEDD8 (neural precursor cell-expressed developmentally down-regulated) is an ubiquitin-like protein essential for protein processing and cell cycle progression. We recently demonstrated that ubiquitin-activating enzyme (Uba)3, the catalytic subunit of the NEDD8-activating enzyme, inhibits ERalpha transcriptional activity. Here we report that Uba3-mediated inhibition of ERalpha transactivation function is due to increased receptor protein turnover. Coexpression of Uba3 with ERalpha increased receptor degradation by the 26S proteasome. Inhibition of NEDD8 activation and conjugation diminished polyubiquitination of ERalpha and blocked proteasome-mediated degradation of receptor protein. The antiestrogen ICI 182,780 is known to induce ER degradation. In human MCF7 breast cancer cells modified to contain a disrupted NEDD8 pathway, ICI 182,780 degradation of ERalpha was impaired, and the antiestrogen was ineffective at inhibiting cell proliferation. This study provides the first evidence linking nuclear receptor degradation with the NEDD8 pathway and the ubiquitin-proteasome system, suggesting that the two pathways can act together to modulate ERalpha turnover and cellular responses to estrogens. Based on our observation that an intact NEDD8 pathway is essential for the antiproliferation activity of the ICI 182,780 in ERalpha positive breast cancer cells, we propose that disruptions in the NEDD8 pathway provide a mechanism by which breast cancer cells acquire antiestrogen resistance while retaining expression of ERalpha.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/metabolismo , Cisteína Endopeptidases/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacologia , Complexos Multienzimáticos/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Receptores de Estrogênio/metabolismo , Ubiquitinas/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Divisão Celular/fisiologia , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio , Feminino , Fulvestranto , Regulação Neoplásica da Expressão Gênica/fisiologia , Células HeLa , Humanos , Immunoblotting , Proteína NEDD8 , Neoplasias Hormônio-Dependentes/genética , Complexo de Endopeptidases do Proteassoma , Receptores de Estrogênio/genética , Ativação Transcricional/fisiologia , Transfecção , Células Tumorais Cultivadas , Enzimas Ativadoras de Ubiquitina
17.
Mol Endocrinol ; 16(2): 315-30, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11818503

RESUMO

Coregulator proteins, coactivators and corepressors, have a profound influence on steroid receptor activity and play a role in regulating receptor levels. To identify novel coregulators of nuclear receptors, we used the ligand-binding and hinge region of ERalpha as bait in a yeast two-hybrid screen of a cDNA library derived from rat uterine luminal epithelium. We report the cloning and characterization of a cDNA encoding a protein homologous to yeast and human ubiquitin-activating enzyme 3 (Uba3), the catalytic subunit of the activating enzyme of the ubiquitin-like NEDD8 (neural precursor cellexpressed developmentally down-regulated) conjugation pathway (known as neddylation). Sequence analysis revealed that Uba3 contains multiple nuclear receptor (NR)-interacting motifs (NR boxes), which are known to mediate interactions between coregulatory proteins and ligand-activated NRs. Yeast two-hybrid and glutathione-S-transferase pull-down assays demonstrated that Uba3 directly interacts with ligand-occupied ERalpha and ERbeta. Transient transfection of Uba3 in mammalian cells inhibited ER-mediated transactivation in a time-dependent fashion; Uba3 had no effect on the initial events of transcriptional activation by liganded ER, but it blocked the progressive increase in target gene expression during continuous stimulation. Uba3 also inhibited transactivation by AR and PR in mammalian cells but had no effect on a steroid receptor-independent transactivation pathway. An enzymatically silent form of Uba3 did not inhibit ER-induced transcription, and a Uba3-binding fragment of amyloid precursor protein-binding protein, the other subunit of the NEDD8-activating enzyme, partially overcame Uba3-mediated inhibition, demonstrating that the neddylation activity of Uba3 is required for its inhibition of steroid receptor transactivation. Thus, Uba3 inhibits transcription induced by steroid hormone receptors through a novel mechanism that involves the neddylation pathway. Understanding the mechanisms controlling hormone responsiveness of target tissues, such as the uterus and mammary gland, may lead to novel insights of therapeutic intervention.


Assuntos
Receptores de Esteroides/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae , Ativação Transcricional , Ubiquitinas/genética , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Perfilação da Expressão Gênica , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Proteína NEDD8 , Ligação Proteica , Subunidades Proteicas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Receptores de Esteroides/metabolismo , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido , Enzimas Ativadoras de Ubiquitina , Ubiquitinas/química , Leveduras
18.
Cancer Lett ; 179(2): 113-9, 2002 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-11888665

RESUMO

Tumor promoter effects of steroid hormones were tested in ovariectomized rats treated with the tumor initiator, methylnitrosourea. Continuous treatment with low doses of estrone induced tumor in 55% of experimental animals while intact control animals had a 96-100% incidence. Although progesterone alone produced no tumors in ovariectomized, methylnitrosourea-treated animals, estrone and progesterone acted synergistically, inducing tumorigenesis in 88% of treated rats. These results point to an important interaction between estrogens and progestins in promoting mammary tumorigenesis.


Assuntos
Alquilantes/toxicidade , Estrona/farmacologia , Neoplasias Mamárias Experimentais/patologia , Metilnitrosoureia/toxicidade , Progesterona/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia , Hipófise/efeitos dos fármacos , Hipófise/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Útero/efeitos dos fármacos , Útero/crescimento & desenvolvimento
19.
Environ Health Perspect ; 111(9): 1249-52, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12842781

RESUMO

Polybrominated diphenyl ethers (PBDEs) are widely used as flame retardants in consumer goods, such as plastics, electronics, textiles, and construction material. PBDEs have been found in human milk, fat, and blood samples. Rodent studies indicate that PBDEs may be detrimental to neurodevelopment, possibly by lowering thyroid hormone concentrations in blood. In the present study, we determined concentrations of PBDEs and thyroid hormones in human fetal and maternal serum. Patients presenting in labor to Indiana University and Wishard Memorial County hospitals in Indianapolis, who were older than 18 years, were recruited to participate. Twelve paired samples of maternal and cord blood were obtained and analyzed using gas chromatographic mass spectrometry; thyroid hormone concentrations were determined by radioimmunoassay. Six congeners of PBDE were measured in maternal and fetal serum samples. The concentrations of total PBDEs found in maternal sera ranged from 15 to 580 ng/g lipid, and the concentrations found in fetal samples ranged from 14 to 460 ng/g lipid. Individual fetal blood concentrations did not differ from the corresponding maternal concentrations, indicating that measurement of maternal PBDE blood levels is useful in predicting fetal exposure; similarly, other reports have shown a high correlation between PBDE in mother's milk and fetal exposure. In accord with reports on other biologic samples, the tetrabrominated PBDE congener BDE-47 accounted for 53-64% of total PBDEs in the serum. The concentrations of PBDEs found in maternal and fetal serum samples were 20-106-fold higher than the levels reported previously in a similar population of Swedish mothers and infants. In this small sample, there was no apparent correlation between serum PBDEs and thyroid hormone concentrations. Our study shows that human fetuses in the United States may be exposed to relatively high levels of PBDEs. Further investigation is required to determine if these levels are specific to central Indiana and to assess the toxic potential of these exposure levels.


Assuntos
Exposição Ambiental , Sangue Fetal/química , Troca Materno-Fetal , Bifenil Polibromatos/sangue , Hormônios Tireóideos/sangue , Adulto , Feminino , Humanos , Bifenil Polibromatos/análise , Gravidez , Valores de Referência , Medição de Risco , Suécia , Estados Unidos
20.
Dev Growth Differ ; 30(3): 301-313, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37281486

RESUMO

The developing uterus, vagina, and cervix of mice whose age ranged from 16 days of gestation to 90 days postnatal were examined for nuclear estrogen receptors (ERs) by autoradiographic and whole cell uptake techniques. ERs were present within mesenchymal cells of these organs throughout the entire period of development and maturation. By contrast, nuclear ER first became detectable by autoradiography in the epithelium of vagina and uterus at 5 and 6 days postnatal, respectively. As a result of administration of the synthetic estrogen, diethylstilbestrol (DES), consecutively from 16 to 18 days of gestation, uterine and vaginal epithelial cell height was increased and epithelial secretory activity was elevated during the first 48 hr of postnatal life. Also, a single does of DES administered on the 2nd day after birth stimulated epithelial proliferation in the uterus as determined by 3 H-thymidine incorporation. These typical estrogenic effects occurred in the absence of nuclear ER within the epithelium. Prenatal DES treatment accelerated the onset of ER activity within the epithelium by 2 to 3 days relative to controls. The possibility that certain effects of estrogen on epithelial differentiation may be mediated indirectly via ER positive mesenchymal cells is discussed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA