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











Base de dados
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 305: 242-249, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27151233

RESUMO

It is well established that the prototypical aryl hydrocarbon receptor (AHR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can both cause and protect against carcinogenesis in non-transgenic rodents. But because these animals almost never develop prostate cancer with old age or after carcinogen exposure, whether AHR activation can affect cancer of the prostate remained unknown. We used animals designed to develop this disease, Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mice, to investigate the potential role of AHR signaling in prostate cancer development. We previously reported that AHR itself has prostate tumor suppressive functions in TRAMP mice; i.e., TRAMP mice in which Ahr was knocked out developed neuroendocrine prostate carcinomas (NEPC) with much greater frequency than did those with both Ahr alleles. In the present study we investigated effects of AHR activation by three different xenobiotics. In utero and lactational TCDD exposure significantly increased NEPC tumor incidence in TRAMP males, while chronic TCDD treatment in adulthood had the opposite effect, a significant reduction in NEPC incidence. Chronic treatment of adult TRAMP mice with the low-toxicity selective AHR modulators indole-3-carbinol or 3,3'-diindolylmethane did not significantly protect against these tumors. Thus, we demonstrate, for the first time, that ligand-dependent activation of the AHR can alter prostate cancer incidence. The nature of the responses depended on the timing of AHR activation and ligand structures.


Assuntos
Anticarcinógenos , Carcinógenos , Dibenzodioxinas Policloradas , Efeitos Tardios da Exposição Pré-Natal , Animais , Anticarcinógenos/farmacologia , Anticarcinógenos/toxicidade , Carcinógenos/farmacologia , Carcinógenos/toxicidade , Carcinoma Neuroendócrino/tratamento farmacológico , Feminino , Lactação , Masculino , Camundongos , Camundongos Transgênicos , Dibenzodioxinas Policloradas/farmacologia , Dibenzodioxinas Policloradas/toxicidade , Gravidez , Neoplasias da Próstata/tratamento farmacológico , Receptores de Hidrocarboneto Arílico/genética
2.
Toxicol Sci ; 150(2): 429-40, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26865671

RESUMO

Benign prostatic hyperplasia, prostate cancer, and changes in the ratio of circulating testosterone and estradiol often occur concurrently in aging men and can lead to lower urinary tract (LUT) dysfunction. To explore the possibility of a fetal basis for the development of LUT dysfunction in adulthood, Tg(CMV-cre);Nkx3-1(+/-);Pten(fl/+) mice, which are genetically predisposed to prostate neoplasia, were exposedin uteroand during lactation to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, 1 µg/kg po) or corn oil vehicle (5 ml/kg) after a single maternal dose on 13 days post coitus, and subsequently were aged without further manipulation, or at 8 weeks of age were exposed to exogenous 17 ß-estradiol (2.5 mg) and testosterone (25 mg) (T+E2) via slow release subcutaneous implants.In uteroand lactational (IUL) TCDD exposure in the absence of exogenous hormone treatment reduced voiding pressure in adult mice, but otherwise had little effect on mouse LUT anatomy or function. By comparison, IUL TCDD exposure followed by exogenous hormone treatment increased relative kidney, bladder, dorsolateral prostate, and seminal vesicle weights, hydronephrosis incidence, and prostate epithelial cell proliferation, thickened prostate periductal smooth muscle, and altered prostate and bladder collagen fiber distribution. We propose a 2-hit model whereby IUL TCDD exposure sensitizes mice to exogenous-hormone-induced urinary tract dysfunction later in life.


Assuntos
Envelhecimento/metabolismo , Poluentes Ambientais/toxicidade , Lactação , Sintomas do Trato Urinário Inferior/induzido quimicamente , Dibenzodioxinas Policloradas/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Animais Geneticamente Modificados , Poluentes Ambientais/farmacocinética , Etinilestradiol/farmacologia , Feminino , Predisposição Genética para Doença , Lactação/metabolismo , Sintomas do Trato Urinário Inferior/genética , Sintomas do Trato Urinário Inferior/metabolismo , Sintomas do Trato Urinário Inferior/patologia , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacocinética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Próstata/efeitos dos fármacos , Próstata/embriologia , Receptores de Hidrocarboneto Arílico/metabolismo , Glândulas Seminais/efeitos dos fármacos , Glândulas Seminais/embriologia , Testosterona/farmacologia , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/embriologia
3.
Toxicol Sci ; 113(1): 198-206, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19805408

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) dorsalizes the pattern of prostatic buds developing from the urogenital sinus (UGS) of male fetal mice, causing some buds to form in inappropriate positions while blocking formation of others. This teratogenic TCDD action significantly reduces prostate main duct number and causes ventral prostate agenesis in exposed males. The purpose of this study was to determine whether inhibition of fibroblast growth factor 10 (FGF10) signaling is mechanistically linked to mouse prostatic budding impairment by TCDD. In utero TCDD exposure induced aryl hydrocarbon receptor-responsive cytochrome P450 1b1 messenger RNA (mRNA) in ventral UGS regions where Fgf10 and fibroblast growth factor receptor 2 (Fgfr2) mRNA were expressed and where budding was most severely inhibited by TCDD. However, TCDD exposure did not reduce Fgf10 or Fgfr2 mRNA abundance in the UGS or alter their distribution. Addition of FGF10 protein to UGS organ culture media increased the abundance of UGS basal epithelial cells immunopositive for phosphorylated extracellular signal-regulated kinase (ERK). FGF10 also increased the number of 5-bromo-2'-deoxyuridine (BrdU)-labeled UGS epithelial cells and increased the number of prostatic buds formed per UGS. Addition of TCDD to UGS organ culture media did not alter FGF10-induced ERK activation in UGS basal epithelium but prevented FGF10-induced BrdU incorporation and blocked FGF10-induced prostatic bud formation. These results identify basal urogenital sinus epithelium cells as the key site of FGF10 action during fetal prostate development and suggest that TCDD likely acts downstream of FGFR2 and ERK to restrict UGS epithelial cell proliferation and prevent prostatic bud formation.


Assuntos
Anormalidades Induzidas por Medicamentos/etiologia , Células Epiteliais/efeitos dos fármacos , Fator 10 de Crescimento de Fibroblastos/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Próstata/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Teratogênicos/toxicidade , Anormalidades Induzidas por Medicamentos/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Proliferação de Células/efeitos dos fármacos , Citocromo P-450 CYP1B1 , Ativação Enzimática , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fator 10 de Crescimento de Fibroblastos/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Idade Gestacional , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Gravidez , Próstata/anormalidades , Próstata/metabolismo , RNA Mensageiro/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
4.
Toxicol Appl Pharmacol ; 239(1): 80-6, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19523480

RESUMO

Estrogens play an important role in prostatic development, health, and disease. While estrogen signaling is essential for normal postnatal prostate development, little is known about its prenatal role in control animals. We tested the hypothesis that estrogen signaling is needed for normal male prostatic bud patterning. Budding patterns were examined by scanning electron microscopy of urogenital sinus epithelium from wild-type mice, mice lacking estrogen receptor (ER)alpha, ERbeta, or both, and wild-type mice exposed to the antiestrogen ICI 182,780. Budding phenotypes did not detectably differ among any of these groups, strongly suggesting that estrogen signaling is not needed to establish the prototypical prostatic budding pattern seen in control males. This finding contributes to our understanding of the effects of low-level estrogen exposure on early prostate development. In utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can greatly alter the pattern in which prostatic buds form and reduce their number. For several reasons, including a prior observation that inhibitory effects of TCDD on prostatic budding in rats depend heavily on the sex of adjacent fetuses, we tested the hypothesis that estrogen signaling is needed for TCDD to disrupt prostatic budding. However, budding did not detectably differ among wild-type mice, or mice lacking ERalpha, ERbeta, or both, that were exposed prenatally to TCDD (5 microg/kg on embryonic day 13.5). Nor did ICI 182,780 detectably affect the response to TCDD. These results strongly suggest that estrogen signaling is not needed for TCDD to inhibit prostatic epithelial budding.


Assuntos
Disruptores Endócrinos/toxicidade , Estrogênios/fisiologia , Exposição Materna/efeitos adversos , Organogênese/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Próstata/efeitos dos fármacos , Animais , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/biossíntese , Receptor beta de Estrogênio/genética , Estrogênios/biossíntese , Feminino , Idade Gestacional , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Gravidez , Próstata/embriologia , Próstata/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Biochem Pharmacol ; 77(7): 1151-60, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19166822

RESUMO

The aryl hydrocarbon receptor (AhR) is a basic-helix-loop-helix transcription factor that binds halogenated aromatic hydrocarbons, polycyclic aromatic hydrocarbons, and endogenous compounds. We previously reported that AhR null (Ahr(-/-)) transgenic adenocarcinoma of the mouse prostate (TRAMP) mice on a C57BL/6J background develop prostate tumors with much greater frequency than AhR wild-type (Ahr(+/+)) TRAMP mice, suggesting that the AhR has tumor suppressor properties. Because AhR signaling pathway inactivation increased susceptibility to prostate tumorigenesis, we tested the hypothesis that a selective AhR modulator (SAhRM), 6-methyl-1,3,8-trichlorodibenzofuran (6-MCDF), can protect against prostate tumorigenesis. TRAMP mice on the standard C57BL/6JxFVB genetic background were fed 0, 10, or 40mg 6-MCDF/kg diet beginning at 8 weeks of age. Tumor incidence, pelvic lymph node metastasis, and serum vascular endothelial growth factor (VEGF) concentrations were determined at 140 days of age. Prostate tumor incidence and size were not significantly reduced in mice fed 6-MCDF. However, the frequency of pelvic lymph node metastasis was reduced fivefold in mice fed the 40mg 6-MCDF/kg diet. Serum VEGF concentrations were also reduced by 6-MCDF treatment, particularly in mice without prostate tumors, and 6-MCDF was shown to act directly on cultured prostates to inhibit VEGF secretion. Together, these results suggest that 6-MCDF inhibits metastasis, in part, by inhibiting prostatic VEGF production prior to tumor formation. This is the first report that 6-MCDF can confer protection against prostate cancer in vivo.


Assuntos
Benzofuranos/uso terapêutico , Neoplasias Pélvicas/prevenção & controle , Neoplasias Pélvicas/secundário , Neoplasias da Próstata/tratamento farmacológico , Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Benzofuranos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Neoplasias Pélvicas/patologia , Neoplasias da Próstata/patologia
6.
Dev Dyn ; 237(5): 1321-33, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18393306

RESUMO

Formation of prostatic buds from the urogenital sinus (UGS) to initiate prostate development requires localized action of several morphogenetic factors. This report reveals all-trans-retinoic acid (RA) to be a powerful inducer of mouse prostatic budding that is associated with reciprocal changes in expression of two regulators of budding: sonic hedgehog (Shh) and bone morphogenetic protein 4 (Bmp4). Localization of retinoid signaling and expression of RA synthesis, metabolism, and receptor genes in the UGS on embryonic days 14.5-17.5 implicate RA in the mechanism of bud initiation. In UGS organ culture, RA increased prostatic budding, increased Shh expression, and decreased Bmp4. Prostatic budding was stimulated in the absence of RA by recombinant SHH, by blocking BMP4 signaling with NOGGIN, or by combined treatment with SHH and NOGGIN in UGS organ culture media. These observations suggest that reciprocal changes in hedgehog and BMP signaling by RA may regulate bud initiation.


Assuntos
Antineoplásicos/farmacologia , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Hedgehog/metabolismo , Morfogênese , Próstata/efeitos dos fármacos , Próstata/embriologia , Tretinoína/farmacologia , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Antineoplásicos/metabolismo , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Próstata/anatomia & histologia , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Transdução de Sinais/fisiologia , Técnicas de Cultura de Tecidos , Tretinoína/metabolismo
7.
Toxicol Sci ; 104(1): 189-97, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18411234

RESUMO

The Hedgehog (Hh) signaling pathway is an essential regulator of embryonic development and appears to play important roles in postnatal repair and cancer progression and metastasis. The teratogenic Veratrum alkaloid cyclopamine is a potent Hh antagonist and is used experimentally both in vitro and in vivo to investigate the role of Hh signaling in diverse biological processes. Here, we set out to establish an administration regimen for cyclopamine-induced teratogenicity in the mouse. The dysmorphogenic concentration of cyclopamine was determined in vitro via mouse whole-embryo culture assays to be 2.0 microM. We administered cyclopamine to female C57BL/6J mice at varied doses by oral gavage, ip injection, or osmotic pump infusion and assessed toxicity and pharmacokinetic (PK) models. Bolus administration was limited by toxicity and rapid clearance. In vivo cyclopamine infusion at 160 mg/kg/day yielded a dam serum steady-state concentration of approximately 2 microM with a corresponding amniotic fluid concentration of approximately 1.5 microM. Gross facial defects were induced in 30% of cyclopamine-exposed litters, with affected embryos exhibiting cleft lip and palate. This is the first report describing the PKs and teratogenic potential of cyclopamine in the mouse and demonstrates that transient Hh signaling inhibition induces facial clefting anomalies in the mouse that mimic common human birth defects.


Assuntos
Fenda Labial/induzido quimicamente , Fissura Palatina/induzido quimicamente , Proteínas Hedgehog/antagonistas & inibidores , Teratogênicos/toxicidade , Alcaloides de Veratrum/toxicidade , Líquido Amniótico/química , Animais , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/efeitos dos fármacos , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Transdução de Sinais , Teratogênicos/farmacocinética , Alcaloides de Veratrum/administração & dosagem , Alcaloides de Veratrum/sangue , Alcaloides de Veratrum/farmacocinética
8.
Toxicol Sci ; 86(2): 387-95, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15888670

RESUMO

In utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure inhibits ventral, dorsolateral, and anterior prostate development in C57BL/6 mice. To determine if prostatic abnormalities persist into senescence, mice born to dams given TCDD (5 mug/kg, po) or vehicle on gestation day 13 were examined at 100 and 510 days of age. Half the mice were castrated ten days prior to necropsy in order to assess androgen dependence, while the remaining mice were sham castrated. Effects of TCDD on the dorsolateral and anterior prostate of senescent sham-castrated mice were relatively subtle, whereas the ventral prostate was rudimentary or absent. Castration of vehicle-exposed mice caused far greater reductions in prostate lobe weights, epithelial cell height, and androgen-dependent gene expression (MP25 and probasin) in young mice than in senescent ones, while cell proliferation was decreased by castration in young mice and increased in senescence. Responses to castration were similar at 100 days of age in vehicle- and TCDD-exposed mice. At 510 days, however, TCDD-exposed mice were substantially more responsive to castration by most indices than vehicle-exposed mice. These results demonstrate that prostatic androgen dependence in mice declines substantially with age in several key ways, and that in utero and lactational TCDD exposure protects against this decline. Surprisingly, TCDD increased the incidence of cribriform structures in dorsolateral prostate ducts, from 2-3% in vehicle-exposed senescent mice to 16% in sham-castrated and to 7% in castrated senescent mice. Collectively, these results demonstrate that effects of in utero and lactational TCDD exposure on the prostate persist into senescence, and suggest that in utero and lactational TCDD exposure retards the aging process in the prostate. However, because cribriform structures are often considered to be associated with prostate carcinogenesis, these results also suggest that TCDD exposure early in development may increase susceptibility to prostate cancer.


Assuntos
Envelhecimento/fisiologia , Lactação , Dibenzodioxinas Policloradas/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Próstata/efeitos dos fármacos , Proteína de Ligação a Androgênios/genética , Proteína de Ligação a Androgênios/metabolismo , Animais , Castração , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Próstata/crescimento & desenvolvimento , Próstata/patologia , RNA Mensageiro/metabolismo , Glândulas Seminais/efeitos dos fármacos , Glândulas Seminais/crescimento & desenvolvimento , Glândulas Seminais/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA