Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Dev Orig Health Dis ; 9(4): 361-372, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29582717

RESUMO

Well-controlled intrauterine development is an essential condition for many aspects of normal adult physiology and health. This process is disrupted by poor maternal nutrition status during pregnancy. Indeed, physiological adaptations occur in the fetus to ensure nutrient supply to the most vital organs at the expense of the others, leading to irreversible consequences in tissue formation and differentiation. Evidence indicates that maternal undernutrition in early life promotes changes in key hormones, such as glucocorticoids, growth hormones, insulin-like growth factors, estrogens and androgens, during fetal development. These alterations can directly or indirectly affect hormone release, hormone receptor expression/distribution, cellular function or tissue organization, and impair tissue growth, differentiation and maturation to exert profound long-term effects on the offspring. Within the male reproductive system, maternal protein malnutrition alters development, structure, and function of the gonads, testes and prostate gland. Consequently, these changes impair the reproductive capacity of the male offspring. Further, permanent alterations in the prostate gland occur at the molecular and cellular level and thereby affect the onset of late life diseases such as prostatitis, hyperplasia and even prostate cancer. This review assembles current thoughts on the concepts and mechanisms behind the developmental origins of health and disease as they relate to protein malnutrition, and highlights the effects of maternal protein malnutrition on rat prostate development and homeostasis. Such insights on developmental trajectories of adult-onset prostate disease may help provide a foundation for future studies in this field.


Assuntos
Doenças Fetais/etiologia , Doenças Fetais/patologia , Desnutrição/complicações , Próstata/crescimento & desenvolvimento , Doenças Prostáticas/etiologia , Doenças Prostáticas/patologia , Adulto , Animais , Feminino , Humanos , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Gravidez
2.
Endocr Rev ; 36(6): E1-E150, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26544531

RESUMO

The Endocrine Society's first Scientific Statement in 2009 provided a wake-up call to the scientific community about how environmental endocrine-disrupting chemicals (EDCs) affect health and disease. Five years later, a substantially larger body of literature has solidified our understanding of plausible mechanisms underlying EDC actions and how exposures in animals and humans-especially during development-may lay the foundations for disease later in life. At this point in history, we have much stronger knowledge about how EDCs alter gene-environment interactions via physiological, cellular, molecular, and epigenetic changes, thereby producing effects in exposed individuals as well as their descendants. Causal links between exposure and manifestation of disease are substantiated by experimental animal models and are consistent with correlative epidemiological data in humans. There are several caveats because differences in how experimental animal work is conducted can lead to difficulties in drawing broad conclusions, and we must continue to be cautious about inferring causality in humans. In this second Scientific Statement, we reviewed the literature on a subset of topics for which the translational evidence is strongest: 1) obesity and diabetes; 2) female reproduction; 3) male reproduction; 4) hormone-sensitive cancers in females; 5) prostate; 6) thyroid; and 7) neurodevelopment and neuroendocrine systems. Our inclusion criteria for studies were those conducted predominantly in the past 5 years deemed to be of high quality based on appropriate negative and positive control groups or populations, adequate sample size and experimental design, and mammalian animal studies with exposure levels in a range that was relevant to humans. We also focused on studies using the developmental origins of health and disease model. No report was excluded based on a positive or negative effect of the EDC exposure. The bulk of the results across the board strengthen the evidence for endocrine health-related actions of EDCs. Based on this much more complete understanding of the endocrine principles by which EDCs act, including nonmonotonic dose-responses, low-dose effects, and developmental vulnerability, these findings can be much better translated to human health. Armed with this information, researchers, physicians, and other healthcare providers can guide regulators and policymakers as they make responsible decisions.


Assuntos
Disruptores Endócrinos/toxicidade , Animais , Compostos Benzidrílicos , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/epidemiologia , Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/epidemiologia , Endocrinologia , Exposição Ambiental , Feminino , Herbicidas , Humanos , Masculino , Neoplasias Hormônio-Dependentes/epidemiologia , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/epidemiologia , Sistemas Neurossecretores/efeitos dos fármacos , Obesidade/induzido quimicamente , Obesidade/epidemiologia , Praguicidas , Fenóis , Ácidos Ftálicos/toxicidade , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/epidemiologia , Reprodução/efeitos dos fármacos , Sociedades Médicas , Glândula Tireoide/efeitos dos fármacos
3.
Endocr Rev ; 36(6): 593-602, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26414233

RESUMO

This Executive Summary to the Endocrine Society's second Scientific Statement on environmental endocrine-disrupting chemicals (EDCs) provides a synthesis of the key points of the complete statement. The full Scientific Statement represents a comprehensive review of the literature on seven topics for which there is strong mechanistic, experimental, animal, and epidemiological evidence for endocrine disruption, namely: obesity and diabetes, female reproduction, male reproduction, hormone-sensitive cancers in females, prostate cancer, thyroid, and neurodevelopment and neuroendocrine systems. EDCs such as bisphenol A, phthalates, pesticides, persistent organic pollutants such as polychlorinated biphenyls, polybrominated diethyl ethers, and dioxins were emphasized because these chemicals had the greatest depth and breadth of available information. The Statement also included thorough coverage of studies of developmental exposures to EDCs, especially in the fetus and infant, because these are critical life stages during which perturbations of hormones can increase the probability of a disease or dysfunction later in life. A conclusion of the Statement is that publications over the past 5 years have led to a much fuller understanding of the endocrine principles by which EDCs act, including nonmonotonic dose-responses, low-dose effects, and developmental vulnerability. These findings will prove useful to researchers, physicians, and other healthcare providers in translating the science of endocrine disruption to improved public health.


Assuntos
Disruptores Endócrinos/toxicidade , Animais , Compostos Benzidrílicos/toxicidade , Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/epidemiologia , Endocrinologia , Exposição Ambiental , Epigênese Genética , Feminino , Interação Gene-Ambiente , Herbicidas/toxicidade , Humanos , Masculino , Neoplasias Hormônio-Dependentes/induzido quimicamente , Neoplasias Hormônio-Dependentes/epidemiologia , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/epidemiologia , Sistemas Neurossecretores/efeitos dos fármacos , Obesidade/induzido quimicamente , Obesidade/epidemiologia , Praguicidas/toxicidade , Fenóis/toxicidade , Ácidos Ftálicos/toxicidade , Bifenilos Policlorados/toxicidade , Neoplasias da Próstata/induzido quimicamente , Neoplasias da Próstata/epidemiologia , Reprodução/efeitos dos fármacos , Sociedades Médicas , Glândula Tireoide/efeitos dos fármacos
4.
J Dev Orig Health Dis ; 1(6): 365-70, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24795802

RESUMO

While early-life estrogens are thought to play a physiologic role in prostate gland development, inappropriate estrogenic exposures either in dose, type or temporally can reprogram the prostate gland and increase susceptibility to abnormal prostate growth with aging including carcinogenesis. This review discusses the evidence for developmental estrogenic reprogramming that leads to adult prostate disease in a rat model. We propose that estrogen imprinting of the prostate is mediated through both structural reorganization of the gland early in life and epigenomic reprogramming that permits life-long memory of the inappropriate developmental exposures including heightened sensitivity to rising estradiol levels with aging. Complex interactions between early epigenetic programming and later-life experiences results in an emergence of multiple epigenomic outcomes, with some contributing to carcinogenesis with aging.

5.
Biol Reprod ; 70(5): 1504-17, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14749301

RESUMO

Objectives of the study were to determine developmental changes in morphology and expression of androgen receptor (AR) and estrogen receptor (ER)alpha in the body of the rat penis exposed neonatally to diethylstilbestrol (DES). Male pups received DES at a dose of 10 microg per rat on alternate days from Postnatal Day 2 to Postnatal Day 12. Controls received olive oil vehicle only. Tissue samples were collected on Days 18 (prepuberty), 41 (puberty), and 120 (adult) of age. DES-induced abnormalities were evident at 18 days of age and included smaller, lighter, and thinner penis, loss of cavernous spaces and associated smooth muscle cells, and increased deposition of fat cells in the corpora cavernosa penis. Fat cells virtually filled the entire area of the corpora cavernosa at puberty and adulthood. Plasma testosterone (T) was reduced to an undetectable level, while LH was unaltered in all treated groups. AR-positive cells were ubiquitous and their profile (incidence and staining intensity) did not differ between control and treated rats of the respective age groups. Conversely, ERalpha-positive cells were limited to the stroma of corpus spongiosus in all age groups of both control and treated rats, but the expression in treated rats at 18 days was up-regulated in stromal cells of corpora cavernosa, coincident with the presence of morphological abnormalities. Hence, this study reports for the first time DES-induced developmental, morphological abnormalities in the body of the penis and suggests that these abnormalities may have resulted from decreased T and/or overexpression of ERalpha.


Assuntos
Animais Recém-Nascidos , Dietilestilbestrol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios não Esteroides/farmacologia , Pênis/efeitos dos fármacos , Pênis/patologia , Envelhecimento/metabolismo , Animais , Feminino , Imuno-Histoquímica , Hormônio Luteinizante/sangue , Masculino , Pênis/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/metabolismo , Testosterona/sangue , Distribuição Tecidual
6.
J Urol ; 166(6): 2267-72, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11696749

RESUMO

PURPOSE: Gap junctions composed of connexin proteins have an essential role in intercellular communication and differentiation. Dysregulation of connexin expression is believed to have a role in carcinogenesis. The human prostate has been reported to express connexin 32 and 43. However, the expression pattern in prostate cancer is controversial, while to our knowledge connexin expression has not been reported in benign prostatic hyperplasia (BPH). To understand the potential involvement in prostate disease connexin 32 and 43 expression was evaluated in a series of normal prostate, BPH and prostate cancer specimens that were surgically removed due to bladder outlet obstruction. MATERIALS AND METHODS: Frozen sections of 23 normal, 43 BPH and 40 cancer involved prostates were evaluated for the presence, staining intensity and pattern of connexin 32 and 43 by immunocytochemical testing. RESULTS: In all specimens examined connexin 43 stain was punctate along the borders of the basal epithelial cells, whereas connexin 32 immunolocalized to luminal epithelial cells. In normal prostate connexin 43 and 32 were present in 87% and 65% of specimens, respectively, at low to moderate stain intensity. Importantly none of the normal samples were negative foreach connexin. In BPH specimens there was a marked increase in the incidence and intensity of connexin 43 and 32 immunostaining within epithelial cells. In addition, 23% of BPH samples showed strong connexin 43 expression in stromal cells. In contrast, connexin was decreased in prostate cancer specimens, of which 65% and 38% were negative for connexin 43 and 32, respectively, and 28% were negative for each type. In poorly differentiated tumors connexin 43 and 32 were present in only 10% and 40% of tumors, respectively, at low immunostaining intensity. CONCLUSIONS: In normal human prostate basal cells communicate via connexin 43 gap junctions, whereas luminal cells communicate via connexin 32 gap junctions. In BPH gap junctional intercellular communication is increased in epithelial and stromal cells, which may have a role in BPH pathogenesis. In prostate cancer gap junctional intercellular communication is decreased, is as indicated by decreased expression of connexin 43 and 32 with severe loss in poorly differentiated prostate cancer. These alterations in connexin expression may have a role in dedifferentiation and tumor progression.


Assuntos
Adenocarcinoma/metabolismo , Conexina 43/biossíntese , Conexinas/biossíntese , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , Conexina 43/análise , Conexinas/análise , Humanos , Imuno-Histoquímica , Masculino , Proteína beta-1 de Junções Comunicantes
7.
Cancer Res ; 61(19): 7310-7, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11585771

RESUMO

The growth and development of the prostate gland are regulated by the androgen and the androgen receptor (AR). Despite our molecular understanding of the roles of the AR regulating; a downstream target gene transcription, the direct or indirect (stromally mediated) actions of the androgen in controlling prostate cell growth and differentiation are still unclear. In this report, an invasive; and metastatic human prostate tumor cell line, androgen-repressed human prostate cancer cell line (ARCaP), either transduced with wild-type human AR (hAR) or a control neomycin-resistant plasmid DNA, was used to evaluate the direct role of AR in regulating prostate tumor cell growth and gene transcription. Results showed that: (a) introduction of wild-type hAR to ARCaP cells restored positive androgen regulation of prostate tumor cell growth in vitro through an enhanced cell-cycle progression from G(0)/G(1) to S and G(2)-M phases; (b) hAR was shown to transactivate glucocorticoid-responsive element but not prostate-specific antigen promoter-directed reporter gene expression; and (c) hAR-transduced ARCaP cells exhibited reduced growth, invasion, and migratory behavior in vitro and tumor growth in vivo. These results suggest that the introduction of hAR into the invasive human prostate cancer ARCaP cell line restored its androgen-regulated cell growth, decreased the rate of tumor growth, and selectively activated AR target gene expression. These cellular functions in response to androgen are commonly associated with increased differentiation of prostate epithelial cells.


Assuntos
Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/fisiologia , Ativação Transcricional/fisiologia , Androgênios/fisiologia , Animais , Adesão Celular/fisiologia , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Metribolona/farmacologia , Camundongos , Camundongos Nus , Receptores Androgênicos/genética , Congêneres da Testosterona/farmacologia , Transfecção , Células Tumorais Cultivadas
8.
Biol Reprod ; 65(5): 1496-505, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11673267

RESUMO

Brief exposure of rats to high doses of natural estrogens early in life results in permanent alterations of the prostate gland, which include differentiation defects, altered gene expression, and dysplasia with aging. Whether low-dose treatments can cause similar effects in the developing prostate remains controversial. The current project was designed to determine the dose-response relationship of the prostate gland to estradiol exposure during the developmentally critical neonatal period in the rat. Male Sprague-Dawley (SD) rats were treated on Days 1, 3, and 5 of life by s.c. injections of a 7-log range of doses (0.015 microg/kg to 15.0 mg/kg) of beta-estradiol-3-benzoate (EB) in 25 microl of peanut oil (Arachis) as vehicle. In a separate block, neonatal Fisher 344 (F344) rats received 0.15, 15.0, or 1500.0 microg EB/kg. Rats were killed on Postnatal Day (PND) 35 or 90, and the prostates were microdissected, weighed, and frozen for immunohistochemistry. Preputial separation and hepatic testosterone hydroxlase activities were monitored and measured to determine the onset of puberty. On PND 35, there was an increase in prostate weights of SD rats treated with low doses of EB and a decrease in prostate weights of SD rats treated with high doses. The low-dose effect was entirely abolished by PND 90, and only high-dose suppression of organ sizes was found. The transient nature of the effect in low-dose animals suggests an advancement of puberty as the cause for increased reproductive organ weights on PND 35. F344 rats were more sensitive than SD rats to the suppressive effects of high doses of neonatal EB on PND 90. Despite this heightened responsiveness in the F344 rats, a low-dose estrogenic effect on adult prostate weights was not observed. Thus, in the rat model a sustained effect at low doses of natural estrogens is not present in the prostate glands.


Assuntos
Animais Recém-Nascidos , Estradiol/análogos & derivados , Estradiol/administração & dosagem , Próstata/efeitos dos fármacos , Envelhecimento , Animais , Peso Corporal/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Próstata/crescimento & desenvolvimento , Próstata/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Receptores Androgênicos/metabolismo , Maturidade Sexual
9.
Biol Reprod ; 65(5): 1506-17, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11673268

RESUMO

Environmental contaminants with estrogenic properties have been cause for heightened concern about their possible role in inducing adverse health effects. Brief exposure of rodents to high doses of natural estrogens early in life results in permanent alterations of the male reproductive tissues, but the question of whether environmentally relevant doses can cause the same effects remains controversial. The current project was designed to determine the dose-response relationship between neonatal estradiol exposure and the development of the male reproductive tract in the rat. Neonatal male Sprague-Dawley (SD) and Fisher 344 (F344) rats were exposed to beta-estradiol-3-benzoate (EB) at concentrations ranging from 0.015 microg/kg body weight (BW) to 15.0 mg/kg BW and 0.15 microg/kg BW to 1.5 mg/kg BW, respectively. Results showed an inverted U-shaped dose-response profile for testis and epididymis weights in 35-day-old SD rats, with increased organ sizes at the low-dose end of the treatment. This effect was transient and was not sustained into adulthood. Increased hepatic testosterone hydroxylase activities in low-dose animals suggest an advancement of puberty as the cause for increased reproductive organ weights. On postnatal day (PND) 90, a stimulatory low-dose response to EB was present in SD rat testicular and epididymal weights, however at one order of magnitude lower dose than that seen on PND 35, suggesting a separate effect. All SD male reproductive tract organs and serum hormones showed a permanent inhibitory response to high doses of neonatal EB. F344 rats exhibited greater estrogen sensitivity on PND 90. Despite this heightened responsiveness, F344 rats did not exhibit a low-dose effect for any endpoint. These low-dose responses to estradiol are organ and strain specific.


Assuntos
Animais Recém-Nascidos , Hidrocarboneto de Aril Hidroxilases , Estradiol/análogos & derivados , Estradiol/administração & dosagem , Genitália Masculina/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Envelhecimento , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Epididimo/efeitos dos fármacos , Epididimo/crescimento & desenvolvimento , Estradiol/farmacologia , Genitália Masculina/crescimento & desenvolvimento , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Esteroide Hidroxilases/metabolismo , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento , Testosterona/sangue
10.
J Mass Spectrom ; 36(8): 882-8, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11523087

RESUMO

Since retinoic acid (RA) and RA receptors are key developmental regulators during organogenesis, they might participate in the abnormal development of the prostate caused by early estrogen exposure. In order to test this assumption, a sensitive analytical method that can differentiate 9-cis, 13-cis, and all-trans RA in small tissue samples ( approximately 8 mg) is required. Since retinol is the metabolic precursor to RA, simultaneous quantification of retinol would also provide valuable information. Here, we report a liquid chromatography-mass spectrometry method for simultaneous determination of retinol and 9-cis, 13-cis, and all-trans RA in rat prostate. Mass spectrometric signal responses for RA were compared using positive ion atmospheric-pressure chemical ionization (APCI) and electrospray, as well as positive ion and negative ion APCI. Positive ion APCI was selected for all subsequent analysis for its better sensitivity, and to provide simultaneous determination of retinol and RA. Ventral prostate tissue samples were homogenized and extracted following simple protein precipitation without derivatization. Baseline separation of 9-cis, 13-cis, and all-trans RA standards was obtained by using a non-porous silica C18 column. Selected ion monitoring of the ions m/z 301 and m/z 269 was carried out for mass spectrometric quantitative analysis. The ion of m/z 301 corresponded to the protonated molecule of RA, whereas the ion of m/z 269 corresponded to loss of water or acetic acid from the protonated molecule of retinol or the internal standard retinyl acetate respectively. The method has a linear response over a concentration range of at least three orders of magnitude. The limit of quantitation was determined to be 702 fmol all-trans RA injected on-column. The method showed excellent intra- and inter-assay reproducibility and good recovery, and is suitable for analyzing RA and retinol in small tissue samples (approximately 8 mg).


Assuntos
Isotretinoína/análise , Próstata/química , Tretinoína/análise , Vitamina A/análise , Alitretinoína , Animais , Cromatografia Líquida/métodos , Isotretinoína/química , Masculino , Espectrometria de Massas/métodos , Ratos , Tretinoína/química , Vitamina A/química
11.
Cancer Res ; 61(16): 6089-97, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11507058

RESUMO

Neonatal exposure of rodents to high doses of estrogen permanently imprints the growth and function of the prostate and predisposes this gland to hyperplasia and severe dysplasia analogous to prostatic intraepithelial neoplasia with aging. Because the rodent prostate gland expresses estrogen receptor (ER)-alpha within a subpopulation of stromal cells and ERbeta within epithelial cells, the present study was undertaken to determine the specific ER(s) involved in mediating prostatic developmental estrogenization. Wild-type (WT) mice, homozygous mutant ER (ERKO) alpha -/- mice, and betaERKO -/- mice were injected with 2 microg of diethylstilbestrol (DES) or oil (controls) on days 1, 3, and 5 of life. Reproductive tracts were excised on days 5 or 10 (prepubertal), day 30 (pubertal), day 90 (young adult), or with aging at 6, 12, and 18 months of age. Prostate complexes were microdissected and examined histologically for prostatic lesions and markers of estrogenization. Immunocytochemistry was used to examine expression of androgen receptor, ERalpha, ERbeta, cytokeratin 14 (basal cells), cytokeratin 18 (luminal cells), and dorsolateral protein over time in the treated mice. In WT-DES mice, developmental estrogenization of the prostate was observed at all of the time points as compared with WT-oil mice. These prostatic imprints included transient up-regulation of ERalpha, down-regulation of androgen receptor, decreased ERbeta levels in adult prostate epithelium, lack of DLP secretory protein, and a continuous layer of basal cells lining the ducts. With aging, epithelial dysplasia and inflammatory cell infiltrate were observed in the ventral and dorsolateral prostate lobes. In contrast, the prostates of alphaERKO mice exhibited no response to neonatal DES either immediately after exposure or throughout life up to 18 months of age. Furthermore, neonatal DES treatment of betaERKO mice resulted in a prostatic response similar to that observed in WT animals. The present findings indicate that ERalpha is the dominant ER form mediating the developmental estrogenization of the prostate gland. If epithelial ERbeta is involved in some component of estrogen imprinting, its role would be considered minor and would require the presence of ERalpha expression in the prostatic stromal cells.


Assuntos
Dietilestilbestrol/farmacologia , Estrogênios/fisiologia , Próstata/efeitos dos fármacos , Próstata/crescimento & desenvolvimento , Receptores de Estrogênio/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
12.
Endocrinology ; 142(1): 359-69, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11145599

RESUMO

Brief exposure to estrogens during the neonatal period interrupts rat prostatic development by reducing branching morphogenesis and by blocking epithelial cells from entering a normal differentiation pathway. Upon aging, ventral prostates exhibit extensive hyperplasia and dysplasia suggesting that neonatal estrogens may predispose the prostate gland to preneoplastic lesions. To determine whether these prostatic lesions may be manifested through aberrant cell-to-cell communications, the present study examined specific gap junction proteins, Connexins (Cx) 32, and Cx 43, and the cell adhesion molecule, E-cadherin, in the developing, adult and aged rat prostate gland. Male rat pups were given 25 microgram estradiol benzoate or oil on days 1, 3, and 5 of life. Prostates were removed on days 1, 4, 5, 6, 10, 15, 30, or 90 or at 16 months, and frozen sections were immunostained for E-cadherin, Cx 43, and Cx 32. Colocalization studies were performed with immunofluorescence using specific antibodies for cell markers. Gap junctions in undifferentiated epithelial cells at days 1-10 of life were composed of Cx 43, which always colocalized with basal cell cytokeratins (CK 5/15). Cx 32 expression was first observed between days 10-15 and colocalized to differentiated luminal cells (CK 8/18). Cx 43 and Cx 32 never colocalized to the same cell indicating that gap junction intercellular communication differs between basal and luminal prostatic cells. While epithelial connexin expression was not initially altered in the developing prostates following estrogen exposure, adult prostates of neonatally estrogenized rats exhibited a marked decrease in Cx 32 staining and an increased proportion of Cx 43 expressing cells. In the developing prostate, E-cadherin was localized to lateral surfaces of undifferentiated epithelial cells and staining intensity increased as the cells differentiated into luminal cells. By day 30, estrogenized prostates had small foci of epithelial cells that did not immunostain for E-cadherins. In the adult and aged prostates of estrogenized rats, larger foci with differentiation defects and dysplasia were associated with a decrease or loss in E-cadherin staining. The present findings suggest that estrogen-induced changes in the expression of E-cadherin, Cx32 and Cx43 may result in impaired cell-cell adhesion and defective cell-cell communication and may be one of the key mechanisms through which changes toward a dysplastic state are mediated. These findings are significant in light of the data on human prostate cancers where carcinogenesis and progression are associated with loss of E-cadherin and a switch from Cx32 to Cx43 expression in the epithelium.


Assuntos
Envelhecimento/fisiologia , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Conexinas/metabolismo , Células Epiteliais/fisiologia , Estradiol/farmacologia , Próstata/fisiologia , Animais , Conexina 43/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Humanos , Masculino , Próstata/citologia , Próstata/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteína beta-1 de Junções Comunicantes
13.
Reprod Fertil Dev ; 13(4): 241-52, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11800163

RESUMO

Brief exposure of rodents to estrogens during early development alters prostate branching morphogenesis and cellular differentiation in a dose-dependant manner. If estrogenic exposures are high, these disturbances lead to permanent imprints of the prostate, which include reduced growth, differentiation defects of the epithelial cells, altered secretory function and reduced responsiveness to androgens in adulthood. This process, referred to as neonatal imprinting or developmental estrogenization, is associated with an increased incidence of prostatic lesions with aging, which include hyperplasia, inflammation and dysplasia. To better understand how early estrogenic exposures can permanently alter prostate growth and function and predispose the gland to neoplasia, the effects of estrogens on prostatic steroid receptors, cell-cell communication molecules and key developmental genes were examined. Transient and permanent alterations in the expression of prostatic androgen receptors, estrogen receptors alpha (ERalpha) and beta, and retinoic acid receptors are observed. It is proposed that the estrogen-induced alterations in these critical transcription factors play a fundamental role in initiating prostatic growth and differentiation defects. Down-stream effects of the altered steroid receptor expression include disruption of TGFbeta paracrine communication, altered expression of gap junction connexin molecules and loss of epithelial cadherin on epithelial cells. Additionally, specific disruptions in the expression of prostatic developmental genes are observed in response to neonatal estrogen. An extended developmental period of hoxa-13 expression, a lack of hoxd-13 increase with maturation, and an immediate and sustained suppression of hoxb-13 was noted within prostatic tissue. A transient decrease in Nkx3.1 expression in the developing prostate was also observed. Thus subtle and overt alterations in Hox-13 and Nkx3.1 genes may be involved in the altered prostate phenotype in response to neonatal estrogen exposure. In summary, estrogen imprinting of the prostate gland is mediated through up-regulated levels of stromal ERalpha, which initiates alterations in steroid receptor expression within the developing gland. Rather than being an androgen-dominated process, as occurs normally, prostatic development is regulated by alternate steroids, including estrogens and retinoids, in the estrogenized animal. This, in turn, leads to disruptions in the coordinated expression of critical developmental genes including TGFbeta, Hox-13 genes and Nkx3.1. Since a precise temporal expression pattern of these and other molecules is normally required for appropriate differentiation of the prostatic epithelium and stroma, the estrogen-initiated disruption in this pattern would lead to permanent differentiation defects of the prostate gland. It is hypothesized that these molecular and cellular changes initiated early in life predispose the prostate to the neoplastic state upon aging.


Assuntos
Estrogênios/metabolismo , Próstata/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Caderinas/metabolismo , Comunicação Celular , Conexinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fator 3-alfa Nuclear de Hepatócito , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Próstata/efeitos dos fármacos , Próstata/patologia , Ratos , Ratos Sprague-Dawley , Receptores de Esteroides/efeitos dos fármacos , Receptores de Esteroides/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Endocrinology ; 141(9): 3471-7, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10965920

RESUMO

Neonatal exposure to high doses of estrogen results in permanent suppression of prostate growth and reduced sensitivity to androgens in adulthood. It is unclear whether alterations in prostate growth are due to a direct effect of estrogens on the gland or are the result of hypothalamic-pituitary-gonadal axis suppression and a subsequent reduction in androgen levels. Therefore, the aim of this study was to determine whether estrogens have a direct effect on the prostate using a defined method of culturing neonatal prostates. Newborn rat ventral prostates were microdissected and cultured in the presence of testosterone, which resulted in branching morphogenesis and ductal canalization. Solid cords of epithelium differentiated into acini lined by tall columnar epithelial cells; these acini were surrounded by stromal cells, expressing smooth muscle alpha-actin. When cultured in the presence of 17beta-estradiol or diethylstilbestrol in addition to testosterone, androgen-induced prostatic growth was reduced, and differentiation was altered. Although estrogen-treated explants were smaller than controls, quantification of epithelial, stromal, and luminal volumes using unbiased stereology revealed significant changes; the proportion of epithelial cells and lumen decreased, and the proportion of stroma increased compared with control values. Concurrent with this reduced growth rate, we observed a disturbance in the branching pattern and a reduction in ductal canalization. Specifically, stromal differentiation and organization were disrupted, so that a discontinuous smooth muscle layer was observed around the epithelial ducts, and epithelial differentiation was altered. The effects of estrogens were not accompanied by a decrease in androgen response via the androgen receptor, because immunolocalization of this receptor remained constant. These data demonstrate that high doses of estrogens are growth inhibitory and have direct effects on prostate development in vitro, which may occur in vivo in addition to indirect effects via suppression of the hypothalamic-pituitary-gonadal axis.


Assuntos
Androgênios/fisiologia , Estrogênios/farmacologia , Próstata/crescimento & desenvolvimento , Actinas/metabolismo , Antagonistas de Receptores de Andrógenos , Animais , Animais Recém-Nascidos , Imuno-Histoquímica , Queratinas/metabolismo , Masculino , Técnicas de Cultura de Órgãos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Sprague-Dawley
15.
Mayo Clin Proc ; 75 Suppl: S32-5, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10959213

RESUMO

The androgen receptor is a member of a superfamily of nuclear transcription factors that mediate the action of steroid hormones. When activated by ligand binding, these transcription factors bind to specific DNA sequences on target genes and regulate the transcriptional activity of those genes. Various genes have been identified that are regulated by androgens, including prostate-specific antigen (PSA). PSA contains an upstream glucocorticoid response element/androgen response element, so it is believed that androgens directly regulate its transcription rate. Despite this, posttranscriptional regulation through stability of the messenger RNA product is believed to be a major mechanism of androgens' effects.


Assuntos
Biologia Molecular , Proteínas Nucleares/genética , Antígeno Prostático Específico/genética , Receptores Androgênicos/genética , Fatores de Transcrição/genética , Transcrição Gênica/genética , Androgênios/fisiologia , Sequência de Bases/genética , Proteínas de Ligação a DNA/genética , Humanos , Ligantes , Proteínas Nucleares/química , Processamento Pós-Transcricional do RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/química , Elementos de Resposta/genética , Fatores de Transcrição/química
16.
Prostate ; 40(2): 115-24, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10386472

RESUMO

Estrogens can have profound effects on prostate growth and differentiation. These effects were thought to be mediated by the classical estrogen receptor; however, the discovery of a second estrogen receptor has redefined the estrogen signaling pathway and may have broad implications on estrogen-responsive tissues, including the prostate. The new estrogen receptor, named estrogen receptor-beta (ERbeta), is preferentially expressed in the prostate and maintains some characteristics that are different from ERalpha. Establishing the distribution and function of ERbeta in the various estrogen-responsive tissues is critical to defining its pharmacological and physiological impact. Differential expression of ERbeta may facilitate development of tissue-specific estrogen agonists and antagonists, a goal in the treatment of diseases in estrogen-sensitive tissues such as breast cancer. This article reviews the current knowledge on ERbeta and its potential impact on the prostate.


Assuntos
Próstata , Receptores de Estrogênio , Animais , Sítios de Ligação , DNA/metabolismo , Dimerização , Humanos , Masculino , Próstata/química , Próstata/citologia , Próstata/fisiologia , Receptores de Estrogênio/química , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/fisiologia , Ativação Transcricional
17.
Endocrinology ; 140(6): 2801-13, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10342871

RESUMO

Exposure of male rats to estrogens during the neonatal period retards prostate branching morphogenesis, blocks epithelial differentiation, and predisposes the adult prostate to hyperplasia and dysplasia. The mechanism of neonatal estrogenization is not well understood. The present study evaluated transforming growth factor-beta (TGFbeta) in the neonatally estrogenized ventral prostate to determine whether this paracrine/autocrine factor may in part mediate the effects ofestrogen on the developing prostate gland. Immunocytochemistry using antibodies against active TGFbeta1 and its latency-associated peptide localized this molecule to the periductal smooth muscle cells in the developing prostate. Although neonatal estrogenization increased the accumulation of total and active TGFbeta1 in the smooth muscle layer as early as day 6 of life, it was physically separated from the epithelial ducts by a proliferating layer of fibroblasts surrounding the basement membrane. RT-PCR demonstrated that alterations in TGFbeta1 levels were not due to alterations in TGFbeta1 transcription. TGFbeta2 and TGFbeta3 were primarily immunolocalized to differentiating epithelial cells in developing prostates, and this was markedly dampened between days 10-30 after neonatal estrogen exposure. Immunocytochemistry for TGFbeta signaling components revealed that neonatal estrogenization transiently reduced TGFbeta type I receptor levels in the prostate epithelium, but not in stroma, between days 6-15, whereas there was no effect on TGFbeta type II receptor. Levels of the intracellular signal Smad2 (52 kDa) were detected in epithelial cells but were not altered after estrogenization. To analyze the functional status of the TGFbeta signaling pathway, immunocytochemistry was performed for p21(cip-1/waf-1), a cyclin-dependent kinase inhibitor that is inducible by TGFbeta1 in the prostate. Transient nuclear localization of p21(cip-1/waf-1) was normally observed in epithelial cells between days 6-15 and was associated with entry of cells into a terminal differentiation pathway. Neonatal estrogenization prevented this transient expression of p21(cip-1/waf-1). The present findings demonstrate that the TGFbeta signaling system is perturbed at several levels in the estrogenized prostate, which may in part account for the epithelial cell differentiation blockade as well as the proliferation of periductal fibroblasts in this model.


Assuntos
Ciclinas/análise , Estrogênios/farmacologia , Próstata/efeitos dos fármacos , Fator de Crescimento Transformador beta/análise , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/análise , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Imuno-Histoquímica , Masculino , Próstata/química , Próstata/citologia , Ratos , Receptores Androgênicos/análise , Receptores de Fatores de Crescimento Transformadores beta/análise , Proteína Smad2 , Transativadores/análise , Fator de Crescimento Transformador beta/imunologia
18.
J Urol ; 161(5): 1504-8, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10210383

RESUMO

PURPOSE: Sperm retrieved by testicular sperm extraction is routinely used to attempt pregnancy by in vitro fertilization-intracytoplasmic sperm injection. We evaluated the efficacy of cryopreserving testicular sperm collected by testicular sperm extraction at diagnostic biopsy. MATERIALS AND METHODS: A total of 73 men with obstructive and 42 with nonobstructive azoospermia underwent testicular sperm extraction at diagnostic biopsy. Sperm was retrieved and cryopreserved in all cases of obstruction and in 15 of nonobstructive azoospermia cases. Before freezing we determined sperm count, motility, morphology and viability, and after thawing we assessed sperm motility and viability. In 17 couples a total of 20 cycles of in vitro fertilization-intracytoplasmic sperm injection were performed and fertilization, cleavage and pregnancy rates were determined in cases of obstruction and nonobstruction. RESULTS: Sperm count and morphology were lower in the testicular biopsies of men with nonobstructive versus obstructive azoospermia. Motility was low or absent in all testicular sperm extraction specimens. Importantly, pre-freeze (63%) and post-thaw (31%) viability was the same in both patient groups. After in vitro fertilization-intracytoplasmic sperm injection using frozen and thawed testicular sperm the fertilization, cleavage, implantation and clinical pregnancy rates were 60, 86, 16 and 50%, respectively. Using cryopreserved sperm we observed no differences in outcome of any in vitro fertilization-intracytoplasmic sperm injection procedure in patients with obstructive versus nonobstructive azoospermia. CONCLUSIONS: Cryopreservation of testicular sperm provides enough good quality sperm after thawing to result in excellent in vitro fertilization-intracytoplasmic sperm injection outcomes. Cryopreservation does not adversely affect intracytoplasmic sperm injection outcomes, including pregnancy rate. Therefore, we recommend routine testicular sperm extraction and cryopreservation of sperm at testicular biopsy.


Assuntos
Criopreservação , Oligospermia , Motilidade dos Espermatozoides , Espermatozoides , Feminino , Fertilização in vitro , Humanos , Masculino , Gravidez/estatística & dados numéricos , Testículo
19.
Endocrinology ; 140(1): 405-15, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9886852

RESUMO

The purpose of this study was to examine whether changes in extracellular matrix (ECM) molecules are associated with the growth inhibition and differentiation defects of the prostate gland following neonatal exposure to estradiol. Using immunocytochemistry (ICC), laminin and collagen IV were localized to the basement membrane (BM) as well to the basal lamina of the periductal smooth muscle of the control developing prostates. In contrast, fibronectin and collagen III were localized throughout the stromal ECM. Exposure to neonatal estrogen altered the staining profile for specific ECM molecules. In the estrogenized rats, a thick layer of cells negative for laminin and collagen IV was observed adjacent to the BM. Electron microscopy and ICC for alpha-actin, fibronectin, and vimentin identified this multicellular layer of periductal cells as differentiated fibroblasts. Peripheral to these fibroblasts, actin-positive smooth muscle formed a second layer of periductal stromal cells. PCNA labeling showed that estrogen exposure increased the fibroblast proliferation. Because many periductal fibroblasts were positive for estrogen receptor alpha (ER alpha) in estrogenized rats, a direct effect of estradiol on their proliferation is suggested. Gelatinolytic gels revealed that estrogen exposure did not alter the activity of matrix metalloproteinases associated with tissue remodeling during prostate morphogenesis. However, the periductal fibroblast layer in estrogenized prostates was devoid of urokinase- and tissue-plasminogen activator, which may potentially alter the localized proteolysis involved in matrix remodeling. It is proposed that proliferation of a multicellular layer of periductal fibroblasts in estrogenized prostates results in a physical barrier that constrains branching morphogenesis and blocks paracrine communications between smooth muscle and epithelial cells which normally regulate differentiation.


Assuntos
Estrogênios/farmacologia , Matriz Extracelular/ultraestrutura , Fibroblastos/efeitos dos fármacos , Próstata/ultraestrutura , Animais , Divisão Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Feminino , Fibroblastos/citologia , Imuno-Histoquímica , Masculino , Camundongos , Microscopia Eletrônica , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Próstata/citologia , Próstata/efeitos dos fármacos , Coelhos , Ratos , Ratos Sprague-Dawley
20.
Toxicol Appl Pharmacol ; 150(2): 254-70, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9653056

RESUMO

In the male Holtzman rat, in utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure decreases prostate weight without inhibiting testicular androgen production or decreasing circulating androgen concentrations. Therefore, the present study sought to characterize effects of TCDD exposure on prostate development, from very early outgrowth from the urogenital sinus (Gestation Day [GD] 20) until rapid growth and differentiation are essentially complete (Postnatal Day [PND] 32). Pregnant Holtzman rats were administered a single dose of TCDD (1.0 microgram/kg po) or vehicle on GD 15 and offspring were exposed via placental transfer (GD 20 euthanasia) or placental and subsequent lactational transfer until euthanasia (if before PND 21) or weaning. Results show that the prostatic epithelial budding process was impaired by in utero TCDD exposure, as evidence by significant decreases in the number of buds emerging from dorsal, lateral, and ventral aspects of the GD 20 urogenital sinus. Ventral prostate cell proliferation index was significantly decreased on PND 1 but was similar to or higher than control at later times, whereas apoptosis was an extremely rare event in ventral prostates from both control and TCDD-exposed animals. Delays were noted in the differentiation of pericordal smooth muscle cells and luminal epithelial cells. In addition, ventral prostates from approximately 40% of TCDD-exposed animals examined on PNDs 21 and 32 exhibited alterations in the histological arrangement of cell types that could not be explained by a developmental delay. Compared to controls, these ventral prostates exhibited a disorganized, hyperplastic epithelium containing fewer luminal epithelial cells and an increased density or continuous layer of basal epithelial cells, as well as thicker periductal smooth muscle sheaths. In addition, in ventral prostates from TCDD-exposed animals, the intensity of androgen receptor staining was relatively low in the central and distal epithelium, and the number of androgen receptor-positive cells was relatively high in the periductal stroma. These data suggest that in utero and lactational TCDD exposure interferes with prostate development by decreasing very early epithelial growth, delaying cytodifferentiation, and, in the most severely affected animals, producing alterations in epithelial and stromal cell histological arrangement and the spatial distribution of androgen receptor expression that may be of permanent consequence.


Assuntos
Lactação , Dibenzodioxinas Policloradas/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Próstata/efeitos dos fármacos , Receptores Androgênicos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , DNA/análise , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Masculino , Tamanho do Órgão , Gravidez , Próstata/embriologia , Próstata/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA