Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 27(33): 4569-79, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18372911

RESUMO

Phosphoinositide 3-OH kinases (PI3Ks) are a group of major intracellular signaling molecules. In our previous study, we found that inhibition of PI3K activity suppressed the androgen receptor (AR)-mediated gene expression in prostate cancer cells. The AR has been considered as a critical determinant for the development and progression of human prostate cancers. In this study, we sought to identify the PI3K isoforms involved in AR transactivation. Using a gene-specific small interference RNA (siRNA) approach, we determined that the regulatory isoform p85alpha and the catalytic isoform p110beta, but not p110alpha, were required for androgen-stimulated AR transactivation and cell proliferation in prostate cancer cells. Consistently, overexpression of wild-type p110beta but not p110alpha gene led to androgen-independent AR transactivation. Silencing p110beta gene in prostate cancer cells abolished tumor growth in nude mice. Of the dual (lipid and protein) kinase activities, p110beta's lipid kinase activity was required for AR transactivation. Further analysis by a chromatin immunoprecipitation assay showed that p110beta is indispensable for androgen-induced AR-DNA interaction. Finally, gene expression analysis of clinical specimens showed that both p85alpha and p110beta were highly expressed in malignant prostate tissues compared to the nonmalignant compartments, and their expression levels correlated significantly with disease progression. Taken together, our data demonstrated that p85alpha and p110beta are essential for androgen-stimulated AR transactivation, and their aberrant expression or activation might play an important role in prostate cancer progression.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/biossíntese , Neoplasias da Próstata/enzimologia , Receptores Androgênicos/metabolismo , Ativação Transcricional , Antagonistas de Receptores de Andrógenos , Androgênios/farmacologia , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Isoenzimas/biossíntese , Isoenzimas/genética , Masculino , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Receptores Androgênicos/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
2.
Cell Death Differ ; 14(12): 2085-94, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17932503

RESUMO

Androgen receptor (AR) is a critical factor in the development and progression of prostate cancer. We and others recently demonstrated that eliminating AR expression leads to apoptotic cell death in AR-positive prostate cancer cells. To understand the mechanisms of AR-dependent survival, we performed a genome-wide search for AR-regulated survival genes. We found that serum/glucocorticoid-induced protein kinase-1 (SGK-1) mRNA levels were significantly upregulated after androgen stimulation, which was confirmed to be AR dependent. Promoter analysis revealed that the AR interacted with the proximal and distal regions of the sgk1 promoter, leading to sgk-1 promoter activation after androgen stimulation. Functional assays demonstrated that SGK-1 was indispensable for the protective effect of androgens on cell death induced by serum starvation. SGK-1 overexpression not only rescued cells from AR small-interfering RNA (siRNA)-induced apoptosis, but also enhanced AR transactivation, even in the absence of androgen. Additionally, SGK-1 siRNA reduced AR transactivation, indicating a positive feedback effect of SGK-1 expression on AR-mediated gene expression and cellular survival. Taken together, our data suggest that SGK-1 is an androgen-regulated gene that plays a pivotal role in AR-dependent survival and gene expression.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/genética , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/metabolismo , Ativação Transcricional/efeitos dos fármacos
3.
J Endocrinol ; 172(2): 247-53, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11834442

RESUMO

Numerous antral follicles develop during the second half of pregnancy in the golden hamster. However, mechanisms regulating follicular development during this period are unknown. Because inhibin and activin are related to follicular development, these hormones were studied to gain insight into any potential roles in follicular development. Plasma inhibin A and B suddenly increased from day 8 of pregnancy, reached peak levels on day 10 and gradually declined to term. Plasma activin A gradually increased from day 8 to day 15 of pregnancy, and this was followed by an abrupt decrease at day one of lactation. Ovariectomy on day 12 of pregnancy rapidly reduced plasma inhibin A and B, but not activin A levels. Hysterectomy or placentectomy on day 12 of pregnancy caused an abrupt decrease in the levels of plasma activin A and FSH, but not inhibin A and B at 6 h after surgery. Hysterectomy also induced atresia of large antral follicles at 24 h after surgery. These results indicate that antral follicles are the main source of circulating inhibin A and B, whereas uteri and placentae are the main source of circulating activin A. These results suggest that increased levels of activin A may be involved in folliculogenesis in the ovary during the second half of pregnancy in the golden hamster.


Assuntos
Ativinas/fisiologia , Subunidades beta de Inibinas/fisiologia , Folículo Ovariano/fisiologia , Placenta/metabolismo , Prenhez/fisiologia , Útero/metabolismo , Ativinas/biossíntese , Ativinas/sangue , Análise de Variância , Animais , Cricetinae , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Hormônio Foliculoestimulante/sangue , Idade Gestacional , Histerectomia , Subunidades beta de Inibinas/biossíntese , Subunidades beta de Inibinas/sangue , Inibinas/biossíntese , Inibinas/sangue , Mesocricetus , Ovariectomia , Placenta/cirurgia , Gravidez
4.
Reprod Toxicol ; 15(3): 269-74, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11390171

RESUMO

Immature female rats received 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) during an induced proestrus or diestrus. The inhibitory effect of TCDD on acute weight gain and the induction of hepatic ethoxyresorufin-o-deethylase (EROD) activity by TCDD were greatest during proestrus. In a second experiment, ovariectomized rats received estradiol cypionate (ECP) or progesterone followed by TCDD. TCDD and estradiol each alone significantly inhibited weight gain. Progesterone potentiated the effects of TCDD on weight gain. The highest dose of ECP was associated with greater induction of hepatic EROD activity by TCDD than seen with TCDD alone. Estradiol modulates the induction of hepatic EROD activity by TCDD. Differential effects of TCDD on acute weight gain during proestrus vs. diestrus in this model do not mimic changes induced by estrogen alone. Hepatic responses to TCDD may vary according to phase of the female reproductive cycle.


Assuntos
Citocromo P-450 CYP1A1/biossíntese , Poluentes Ambientais/toxicidade , Fígado/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Animais , Gonadotropina Coriônica/farmacologia , Diestro/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Estradiol/análogos & derivados , Estradiol/sangue , Estradiol/farmacologia , Feminino , Gonadotropinas Equinas/farmacologia , Humanos , Fígado/enzimologia , Modelos Animais , Ovariectomia , Ovário/patologia , Proestro/efeitos dos fármacos , Progesterona/farmacologia , Ratos , Ratos Sprague-Dawley , Aumento de Peso/efeitos dos fármacos
5.
Reprod Toxicol ; 15(3): 275-80, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11390172

RESUMO

Several studies have established that 2,3,7,8 tetrachloro-p-dioxin (TCDD) blocks ovulation. The main purpose of this study was to determine if induced ovulation was delayed temporarily by TCDD. The ovulation model used was that of the gonadotropin-primed intact or hypophysectomized rat. Immature intact female Sprague-Dawley rats (IIR) were given 32 microg TCDD/kg by gavage on day 24 of age. The next day equine chorionic gonadotropin (eCG) (5 IU) was injected sc to stimulate follicular development. The number of ova in the oviducts, the ovulation rate, and steroid concentrations were determined at 72, 96, 120, and 144 h after eCG. Immature female Sprague-Dawley rats (IHR) were hypophysectomized on day 23 of age. On day 26, the IHR were given 20 microg TCDD/kg by gavage. The next day eCG (10 IU) was injected sc to stimulate follicle development and at 52 h after eCG, 10 IU human chorionic gonadotropin (hCG) was given to induce ovulation. The same parameters as in IIR were determined in IHR at 72, 96, and 120 h after eCG. TCDD decreased body and ovarian weight gains in both IIR and IHR. In IIR, TCDD delayed ovulation by 24 to 48 h reducing the number of ova shed as well as the number of animals ovulating at 72 and 96 h after eCG. In IHR, however, TCDD reduced only the number of ova shed but caused no delay in ovulation. The IIR treated with TCDD had low levels of progesterone (P4) at 72 and 96 h after eCG but high levels of estradiol (E2) at the same time points. This sustained high level of E2 production coincided with a transient decrease in serum concentrations of androstenedione (A4). The alteration of steroid hormones by TCDD was restored to normal by 48 h after ovulation in IIR. Serum P4 concentration was not altered by TCDD in IHR at 72 h after eCG but was decreased thereafter. The delay in ovulation induced by TCDD in IIR indicates the disruption of the hypothalamus-pituitary-ovary axis during proestrus. The decrease in number of ova shed in IHR induced by exogenous gonadotropins indicates an additional direct ovarian effect of TCDD in blocking ovulation.


Assuntos
Poluentes Ambientais/toxicidade , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Contagem de Células , Gonadotropina Coriônica/farmacologia , Poluentes Ambientais/administração & dosagem , Feminino , Hormônios Esteroides Gonadais/sangue , Gonadotropinas Equinas/farmacologia , Humanos , Hipofisectomia , Modelos Animais , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/patologia , Ovário/fisiologia , Ovulação/fisiologia , Indução da Ovulação , Óvulo/citologia , Óvulo/efeitos dos fármacos , Dibenzodioxinas Policloradas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Toxicology ; 158(3): 91-107, 2001 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-11275352

RESUMO

Polychlorinated dibenzo-p-dioxins (PCDDs) can impinge on female fertility by preventing ovulation. In this review, the aspects of normal ovulatory physiology most relevant to our current understanding of PCDD action on the ovary are briefly reviewed. This is followed by a comprehensive assessment of data relevant to the effects of PCDDs during ovulation in the rat. PCDDs interrupt ovulation through direct effects on the ovary in combination with dysfunction of the hypothalamo-hypophyseal axis.


Assuntos
Anovulação/induzido quimicamente , Poluentes Ambientais/toxicidade , Ovulação/fisiologia , Dibenzodioxinas Policloradas/análogos & derivados , Dibenzodioxinas Policloradas/toxicidade , Animais , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Ratos
7.
Toxicol Appl Pharmacol ; 170(3): 181-90, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11162783

RESUMO

Sprague-Dawley rats (23-day-old) were dosed with TCDD (32 microg/kg) in corn oil or vehicle alone. Equine chorionic gonadotropin (eCG) was injected (5 IU, sc) 24 h later to induce follicular development. Another 24 h later, half of TCDD- or corn oil-treated rats were injected (sc) with 17 beta-estradiol-cypionate (ECP, at 0.004 to 0.5 mg/kg). Blood and ovaries were collected on expected proestrous (preovulatory period) at 51, 54, and 58 h after eCG injection as well as in the morning after ovulation (72 h after eCG). Serum concentrations of 17 beta-estradiol (E), progesterone (P), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) were determined by radioimmunoassay. The number of ova shed was measured at 72 h after injection of eCG by irrigating ova from oviducts. During the preovulatory period (approximately 58 h after eCG injection), a circulating level of 70-100 pg E/ml coincided with LH and FSH surges and later normal ovulation of 10 to 12 ova/rat was observed in controls. However, the same concentration of E was not associated with LH and FSH surges in rats treated with TCDD (32 microg/kg), resulting in reduced ovarian weight gain and reduction of ovulation by 70 to 80% (2-3 ova/rat). Blockage of the gonadotropin surge, reduced ovarian weight gain, and ovulation were all reversed completely by the lowest effective dose of ECP (0.1 mg/kg). At 72 h after eCG, serum P secretion was reduced and serum E levels were significantly increased compared to those of corn oil-treated controls. ECP alone had no effect on serum P levels at any time point, but in rats treated with TCDD and ECP, both the reduction of P (at 58 and 72 h) and the increase in E secretion (72 h) were completely reversed. Further studies confirmed that restoration by ECP of gonadotropin surges and associated ovulation could not be attained until circulating levels of E rose sufficiently high to trigger the LH and FSH surges. The new action threshold of E for inducing gonadotropin surges in rats treated with TCDD (32 microg/kg) was determined to be eight- to 10-fold higher than that in controls. Thus, it is apparent that TCDD decreased the responsiveness of the hypothalamus to E as a feedback inducer of preovulatory gonadotropin secretion.


Assuntos
Estradiol/farmacologia , Gonadotropinas/metabolismo , Hipotálamo/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Fase Folicular , Gonadotropinas/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Hormônio Luteinizante/sangue , Modelos Animais , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/patologia , Ovulação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
8.
Endocrine ; 15(3): 271-6, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11762698

RESUMO

The purpose of the present study was to investigate whether the tyrosine kinase inhibitor herbimycin with some selectivity to block Src would alter the stimulatory effects of follicle-stimulating hormone (FSH) and cyclic adenosine monophosphate (cAMP) on estradiol secretion by human granulosa cells. Granulosa cells were taken from ovaries of premenopausal women undergoing oophorectomy for reasons unrelated to ovarian pathology. Granulosa cells from follicles ranging from 5-20 mm in diameter were subjected to culture. Granulosa cells were cultured with human FSH (2 ng/mL) or cAMP (0-1 mM) and testosterone (1 microM) in the presence and absence of herbimycin (0-2 pM). Media were collected at 24, 48, and 72 h. Accumulation of cAMP, progesterone, and estradiol in the media was determined by radioimmunoassay. Herbimycin dose dependently inhibited the ability of FSH to induce increases in progesterone and estradiol secretion. Although herbimycin increased (p < 0.0001) the accumulation of cAMP in response to FSH, this was evident only at the high concentrations of herbimycin (2 microM). To determine whether herbimycin would inhibit the ability of exogenous cAMP to induce estradiol and progesterone secretion, granulosa cells were incubated with 0-1 mM cAMP in the presence and absence of various doses of herbimycin. Herbimycin inhibited cAMP-induced estradiol and progesterone secretion in granulosa cells. The results from seven experiments indicate that herbimycin inhibits FSH stimulation of estradiol and progesterone secretion and that this inhibition may be, in part, at post-cAMP site(s).


Assuntos
Inibidores Enzimáticos/farmacologia , Estradiol/metabolismo , Células da Granulosa/metabolismo , Progesterona/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinonas/farmacologia , Quinases da Família src/antagonistas & inibidores , Adulto , Benzoquinonas , Células Cultivadas , AMP Cíclico/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Humanos , Técnicas In Vitro , Lactamas Macrocíclicas , Folículo Ovariano/anatomia & histologia , Folículo Ovariano/citologia , Radioimunoensaio , Rifabutina/análogos & derivados
9.
Toxicology ; 147(1): 15-22, 2000 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-10837928

RESUMO

Several studies have shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has inhibitory effects on ovulation. This action may be the result of either direct effect(s) of TCDD on ovarian function or via altered secretion of pituitary luteinizing hormone (LH) and follicle stimulating hormone (FSH) which regulate ovarian follicular development and ovulation. To further evaluate the effects of TCDD on pituitary gonadotropins and their regulation, the potential role of gonadotropin-releasing hormone (GnRH) was investigated in the current study. Immature (23-day-old) female Sprague-Dawley rats were dosed with TCDD (32 microg/kg) in corn oil or vehicle alone. Equine chorionic gonadotropin (eCG) was injected subcutaneously (5 IU, sc) 24 h later to induce follicular development. Immediately prior to the expected time of the LH/FSH surges, 54 h after eCG injection, half of TCDD- or corn oil-treated rats were injected with GnRH (2 microg/rat, sc). Blood and ovaries were collected at 54, 56, 58, 60 and 72 h after eCG. Serum concentrations of 17beta-estradiol (E(2)), progesterone (P(4)), LH, and FSH were determined by radioimmunoassay. An indication of ovulation rate was assessed at 72 h after injection of eCG by irrigating the ova from oviducts. TCDD reduced the number of ova in the oviducts by 70-80% (2-3 ova/rat) and this was confirmed by the number of corpora lutea. GnRH partially restored ovulation (6-7 ova/rat) in TCDD-treated rats without reversing its effect on ovarian weight reduction. In controls, the LH and FSH surges at 58 h after eCG were significantly reduced at that time in TCDD-treated rats. However, in rats treated with TCDD and GnRH, a huge LH/FSH surges occurred at 56 h after eCG injection. GnRH alone enhanced E(2) and P(4) serum levels at 56-58 h after eCG injection. In rats treated with both TCDD and GnRH, E(2) secretion was significantly lower at 58, 60, and 72 h when compared with GnRH alone, whereas serum P(4) was only decreased at 72 h after eCG injection. The results indicate that exogenous GnRH induces LH and FSH surges in TCDD-treated rats, but only partially restores the inhibitory effects of TCDD on ovulation.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas/farmacologia , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/antagonistas & inibidores , Animais , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/sangue , Gonadotropinas/sangue , Hormônio Luteinizante/sangue , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Progesterona/sangue , Ratos , Ratos Sprague-Dawley
10.
Reprod Toxicol ; 14(3): 247-55, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10838126

RESUMO

Immature rats were treated with estradiol cypionate, (ECP, 0, 0.1, 1, or 2 mg/kg s.c.) followed 24 h later by TCDD (0 or 10 microg/kg orally). Follicular development was induced with eCG [5 or 10 IU subcutaneously (s.c.)] followed by an ovulatory dose of hCG (10 IU s. c.). Inhibition of ovulation by TCDD was potentiated by ECP in hypophysectomized but not intact rats. Only hypophysectomized rats exposed systemically to TCDD and ECP exhibited weight loss. Pair feeding mimicked the combined effects of TCDD and ECP in hypophysectomized rats. In another experiment, intact rats received ECP s.c. (0 or 2 mg/kg) and TCDD into the ovarian bursa (0 or 250 ng). Another group of intact rats received TCDD orally (10 microg/kg) and ECP into the ovarian bursa (0 or 1.5 microg). Blockade of ovulation by systemic or local TCDD was alleviated by ECP pretreatment. Estrogen increased the systemic toxicity of TCDD in rats whereas antagonizing its direct ovarian effects.


Assuntos
Poluentes Ambientais/toxicidade , Estradiol/farmacologia , Ovário/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Sinergismo Farmacológico , Estradiol/sangue , Feminino , Hipofisectomia , Injeções Subcutâneas , Tamanho do Órgão/efeitos dos fármacos , Ovário/patologia , Progesterona/sangue , Ratos , Ratos Sprague-Dawley
11.
Carcinogenesis ; 21(4): 585-91, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10753190

RESUMO

Mouse ovarian surface epithelial cells (MOSEC) were obtained from virgin, mature mice by mild trypsinization and were repeatedly passaged in vitro. Early passage cells (<20 passages) exhibited a cobblestone morphology and contact inhibition of growth. After approximately 20 passages in vitro, cobblestone morphology and contact inhibition of growth was lost. Tumor forming potential was determined by s.c. and i.p. injection of early and late passage cells into athymic and syngeneic C57BL6 mice. Subcutaneous tumors formed in approximately 4 months and were present only at the injection site. Intraperitoneal injection of late passage MOSEC into athymic and syngeneic mice resulted in growth of tumor implants throughout the abdominal cavity, and production of hemorrhagic ascitic fluid. Early passage MOSEC did not form tumors in vivo. Histopathologic analysis of tumors revealed a highly malignant neoplasm containing both carcinomatous and sarcomatous components. Late passage MOSEC expressed cytokeratin and did not produce ovarian steroids in response to gonadotropin stimulation in vitro. Ten clonal lines were established from late passage MOSEC. Each clone formed multiple peritoneal tumors and ascitic fluid after i.p. injection into C57BL6 mice. Three cell lines examined cytogenetically were polyploid with near-tetraploid modal chromosome numbers. Common clonal chromosome gains and losses included +5, +15, +19 and -X, -3, -4. One cell line had a clonal translocation between chromosomes 15 and 18 and another had a small marker chromosome; common structural abnormalities were not observed. These data describe the development of a mouse model for the study of events related to ovarian cancer in humans. The ability of the MOSEC to form extensive tumors within the peritoneal cavity, similar to those seen in women with Stage III and IV cancer, and the ability of the MOSEC to produce tumors in mice with intact immune systems, makes this model unique for investigations of molecular and immune interactions in ovarian cancer development.


Assuntos
Transformação Celular Neoplásica , Modelos Animais de Doenças , Neoplasias Ovarianas/patologia , Ovário/patologia , Animais , Células Cultivadas , Aberrações Cromossômicas , Células Epiteliais/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/genética
12.
Toxicol Appl Pharmacol ; 163(2): 115-24, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10698669

RESUMO

Previous studies have shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 1,2,3,7,8-pentachlorodibenzo-p-dioxin (PeCDD), and 1,2,3,4,7, 8-hexachlorodibenzo-p-dioxin (HxCDD), and their equipotent mixture block ovulation, reduce ovarian weight gain and alter preovulatory hormone levels in a similar manner. The objective of the current experiment was to investigate the effect of other structurally related compounds such as chlorinated furans and biphenyls on ovulation and related hormonal endpoints. The gonadotropin-primed immature female rat model was used to study the effect of 2,3,4,7, 8-pentachlorodibenzofuran (PeCDF), 3,3',4,4',5-pentachlorobiphenyl (PeCB), and 2,2',5,5' tetrachlorobiphenyl (TCB) and their mixture with polychlorinated dibenzo-p-dioxins (PCDDs) on ovulation. Rats were dosed on Day 23 of age at 0900 h with individual congeners (PeCDF, PeCB, TCB) or a mixture of five compounds, which included TCDD, PeCDD, HxCDD, in addition to PeCDF and PeCB. Equine choronic gonadotropin (eCG; 5 IU) was injected 24 h later to induce follicular development. Blood and ovaries were harvested, and ovarian weights determined at various times after eCG. Serum concentrations of 17beta-estradiol (E(2)), progesterone (P(4)), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) were determined by radioimmunoassay. At 72 h after injection of eCG, the number of ova shed was measured by irrigating the ova from oviducts. The slopes of the dose-responses for inhibition of ovulation generated by the individual PeCDF, PeCB, and/or their mixture with PCDDs were similar. PeCDF, PeCB, and the mixture increased serum concentrations of E(2) at 72 h after eCG injection, the day of expected ovulation; in contrast, serum P(4) and FSH were decreased at that same time point. Only the high doses of TCDD, PeCDF, and PeCB blocked LH and FSH surges at 58 h after eCG. The ovarian histology revealed that the effects of PeCDF, PeCB, and the mixture were very similar to those of PCDDs, consisting of ova in large preovulatory follicles and a lack of or reduced number of corpora lutea. Parallel dose-responses of the individual congeners (PeCDF and PeCB) and their equipotent mixture with PCDDs support the toxic equivalency (TEQ) concept for the blockage of ovulation. Thus, PCDDs, PCDFs, and PeCBs appear to block ovulation by the same or a very similar mechanism of action.


Assuntos
Benzofuranos/toxicidade , Ovulação/efeitos dos fármacos , Bifenilos Policlorados/toxicidade , Animais , Dibenzofuranos Policlorados , Dioxinas/toxicidade , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/farmacologia , Hormônio Luteinizante/sangue , Hormônio Luteinizante/farmacologia , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/patologia , Ratos , Ratos Sprague-Dawley
13.
Reprod Toxicol ; 13(6): 521-30, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10613400

RESUMO

The main purpose of this study was to investigate the direct effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on ovarian function including ovulation and steroidogenesis. In vivo effects of TCDD were investigated on ovulation and alteration of circulating and ovarian steroid hormones in immature hypophysectomized rats (IHR) primed with equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG). In addition, in vitro effects of TCDD on the steroidogenesis of granulosa cells (GC), theca-interstitial cells (TIC), and whole ovarian dispersates derived from the ovary of IHR were investigated. In the ovulation model, rats were hypophysectomized on Day 23 of age. On Day 26, the IHR were given 20 microg TCDD/kg by gavage. The next day eCG (10 IU) was injected sc to stimulate follicular development. Fifty-two hours after eCG, 10 IU hCG was given to induce ovulation. TCDD (20 microg/kg) blocked ovulation and reduced ovarian weight in IHR. Concentrations of progesterone (P4), androstenedione (A4), and estradiol (E2) in sera and ovaries were not altered by TCDD at 12, 24, 48, and 72 h after eCG. except for a two-fold increase in ovarian concentration of A4 at 48 h after TCDD. However, this higher concentration of A4 at 48 h after TCDD did not reflect that of A4 in sera and did not correlate with E2 in either sera or ovaries. In isolated GC from untreated IHR, TCDD (0.1 to 100 nM) had no significant effect on P4 and E2 after stimulation by LH or FSH. In TIC and whole ovarian dispersates containing GC, TIC, and other ovarian cells, TCDD (0.1 to 800 nM) had no effect on A4 and P4 secretion stimulated by LH. Using RT-PCR, AhR mRNA was shown to be expressed constitutively in the whole ovary of IHR with maximum down-regulation at 6 h after TCDD (20 microg/kg). Ovarian CYP1A1 was induced maximally at 6 h after TCDD, whereas CYP1B1 could not be detected. The induction of AhR related genes by TCDD in the ovary implies the existence of AhR-mediated signal transduction pathways. In summary, these results indicate that TCDD does not affect ovulation in IHR by altering ovarian steroidogenesis. It seems that inhibition of ovulation by TCDD is due to processes related to follicular rupture.


Assuntos
Poluentes Ambientais/toxicidade , Células da Granulosa/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Esteroides/biossíntese , Células Tecais/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/fisiologia , Humanos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovário/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Hidrocarboneto Arílico/biossíntese , Receptores de Hidrocarboneto Arílico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tecais/fisiologia
14.
Biol Reprod ; 61(6): 1616-21, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10570011

RESUMO

C57BL6 mice with targeted disruption of tumor necrosis factor (TNF) type 1 receptor (TNFRI) exhibited early vaginal opening when compared with wild-type mice (Day 24 +/- 0.6, n = 10, vs. 28 +/- 0.2, n = 11, P < 0.001). Equine CG- and hCG-treated TNFRI null mice ovulated more ova than did controls at two distinct times during the prepubertal period (Day 21: 13.4 +/- 1.7 vs. 7.3 +/- 1.4, P < 0.05; Day 25: 20.7 +/- 2.7 vs. 13.0 +/- 1.3, P < 0.05). Enhanced responsiveness to gonadotropins was not observed in adult mice. At 6 mo of age only 40% of TNFRI null mice exhibited estrous cycles. Those TNFRI null mice with estrous cycles spent significantly more time in diestrus and less time in estrus than controls. TNFRI null mice delivered significantly fewer litters (P < 0.001) than did C57BL6 and TNFRII null mice (TNFRI null 2.59 +/- 0.39; C57BL6 4.91 +/- 0.57; TNFRII null 5.40 +/- 0.60 litters/mo/10 pairs over a 12-mo period). Ovarian dispersates prepared on Day 25 of age from control and TNFRI knockout mice were cultured with and without 10 ng TNF/ml. TNF inhibited LH-stimulated progesterone and estradiol secretion by control dispersates but had no effect on cAMP. In contrast, TNF did not affect LH-stimulated accumulation of progesterone, estradiol, or cAMP by ovarian dispersates from TNFRI knockout mice. The results indicate that lack of TNFRI enhances ovarian responsiveness to gonadotropins during the prepubertal period and may be related to early vaginal opening. The lack of TNFRI is associated with early senescence and poor fertility. These studies demonstrate that the mechanism of TNF-mediated inhibition of steroidogenesis is most likely via TNFRI.


Assuntos
Ovário/fisiologia , Receptores do Fator de Necrose Tumoral/deficiência , Animais , Gonadotropina Coriônica/farmacologia , AMP Cíclico/metabolismo , Estro , Feminino , Hormônio Luteinizante/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovulação/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Maturidade Sexual , Fator de Necrose Tumoral alfa/farmacologia
15.
Endocrine ; 10(1): 19-23, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10403567

RESUMO

Tumor necrosis factor alpha (TNF) inhibits follicle-stimulating hormone- (FSH)induced estradiol secretion by granulosa cells in several species, including humans. One major inhibitory effect of TNF in rat granulosa cells is at the level of stimulatable adenylyl cyclase, resulting in reduced cAMP concentrations. The purpose of the present study was to investigate whether a reduction in cAMP secretion could account for the inhibitory effects of TNF on FSH-induced estradiol in human granulosa cells. Granulosa cells were taken from ovaries of premenopausal women undergoing oophorectomy for reasons unrelated to ovarian pathology. Women in this study were in various stages of the menstrual cycle or exhibited irregular cycles. Granulosa cells from follicles ranging from 5 to 10 mm diameter were subjected to culture for 48 and 96 h. Granulosa cells were cultured with human FSH (2 ng/mL) and testosterone (1 microM) in the presence and absence of human TNF (20 ng/mL). Media were collected at 48 h, fresh media and hormones added, and cultures continued for an additional 48 h. Accumulation of cAMP, progesterone, and estradiol in media were determined by radioimmunoassay (RIA). FSH induced significant increases in cAMP, progesterone, and estradiol by 96 h of culture. TNF inhibited the secretion of estradiol at 96 h without reducing the accumulation of cAMP and progesterone in media. Similar results were observed in the presence of 0.1 mM isobutylmethylxanthine (D3MX), a phosphodiesterase inhibitor that would prevent metabolism of cAMP to AMP. To determine whether TNF would inhibit the ability of cAMP to induce estradiol and progesterone secretion, granulosa cells were incubated with 0.1 mM cAMP in the presence and absence of TNF. TNF consistently inhibited the ability of cAMP to increase estradiol secretion. These results indicate that a pathway for TNF inhibition of FSH- or cAMP-induced estradiol secretion in human granulosa cells is at post-cAMP sites rather than inhibition of FSH-stimulatable adenylyl cyclase.


Assuntos
AMP Cíclico/metabolismo , Estradiol/metabolismo , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Adenilil Ciclases/metabolismo , Adulto , Células Cultivadas , Feminino , Células da Granulosa/efeitos dos fármacos , Humanos , Progesterona/metabolismo , Proteínas Recombinantes/farmacologia , Testosterona/farmacologia
16.
Toxicol Appl Pharmacol ; 157(2): 107-16, 1999 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10366543

RESUMO

Polychlorinated dibenzo-p-dioxins (PCDDs) are structural analogues, which produce a similar spectrum of biological and toxicological responses in animals, albeit with differential potencies. Very consistent structure-activity relationships have been found for acute toxicity and some biochemical effects among these compounds. For the current experiments, the gonadotropin-primed immature female rat model was used to study the effect of 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD), 1,2,3,7, 8-pentachlorodibenzo-p-dioxin (PeCDD), and 1,2,3,4,7, 8-hexachlorodibenzo-p-dioxin (HxCDD) on ovulation. Single doses of different PCDDs and their mixture were given orally to 23-day-old rats. Gonadotropin from pregnant mare's serum (PMSG) was injected (5 IU) 24 h later to induce follicular maturation. Rats were decapitated at various times after PMSG, blood was collected, and ovarian weight was measured. Serum concentrations of 17beta-estradiol (E2), progesterone (P4), luteinizing hormone (LH), follicle stimulating hormone (FSH), and prolactin (PrL) were determined by radioimmunoassay. Ovulation was measured at 72 h after injection of PMSG by counting ova flushed from oviducts. PCDDs dose dependently decreased the number of ova per ovary and reduced ovarian weight gain induced by PMSG. The slopes of the dose-response curves generated by individual PCDDs and/or their mixture were similar. PMSG-induced increase in serum E2 was enhanced on the day of expected ovulation by PCDDs; in contrast, serum P4 and FSH were decreased at that same time point. PCDDs also altered the temporal pattern of serum E2, FSH, and LH but not that of PrL. Histologically the effect of all three PCDDs consisted of ova trapped in preovulatory follicles and a lack of or reduced number of corpora lutea. The results indicate that the PCDDs, tested in the present model, have the same mode of action on ovulation and the reproductive hormones, e.g., LH, FSH, P4 and E2. Furthermore, the dose responses of the individual congeners are parallel to each other and also to that of their equipotent mixture, which represent a validation of the TEQ concept for one aspect of endocrine disruption, that is for inhibition of ovulation.


Assuntos
Hormônios Esteroides Gonadais/sangue , Gonadotropinas Hipofisárias/sangue , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/análogos & derivados , Dibenzodioxinas Policloradas/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estradiol/sangue , Feminino , Gonadotropinas Equinas/antagonistas & inibidores , Gonadotropinas Equinas/farmacologia , Tamanho do Órgão/efeitos dos fármacos , Ovário/efeitos dos fármacos , Ovário/fisiologia , Ovulação/sangue , Progesterona/sangue , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
17.
J Reprod Fertil ; 113(2): 337-41, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9861175

RESUMO

Tumour necrosis factor alpha (TNF-alpha) concentrations were measured during periods of controlled and natural follicular development and ovulation in rat ovaries. Concentrations of bioactive TNF-alpha were determined in the ovaries and sera of rats during puberty (the period of vaginal opening and the first ovulation) and in immature rats after gonadotrophin treatment. Ovaries and sera were collected from 33-, 35-, 37-, 39-, 41- and 43-day-old rats (n = 6 or 7 per group); vaginal opening occurs on day 35. The presence of ovarian follicles and corpora lutea or ova in the oviducts was assessed. For gonadotrophin treatment, a single subcutaneous injection of 5 iu equine chorionic gonadotrophin (eCG) was administered at 08:00 h to 28-day-old rats to stimulate follicular development. A single subcutaneous injection of 10 iu human chorionic gonadotrophin (hCG) was administered 48 h later to induce ovulation. Ovaries and sera from three to six animals per group were collected 0, 3, 24, 48, 51, 54, 57, 60 and 72 h after injection of eCG. At puberty, ovarian concentrations of TNF-alpha were highest (approximately 1.1 fg micrograms-1 ovarian protein) before vaginal opening and the first ovulation. After vaginal opening and ovulation at day 37, ovarian concentrations of TNF-alpha were markedly reduced (0.091 fg microgram-1 ovarian protein) and remained low up to day 43. Serum concentrations of TNF-alpha remained low throughout the period of vaginal opening and the first ovulation (8-32 pg ml-1). In 43-day-old rats serum concentrations of TNF-alpha increased (105 pg ml-1). In the immature ovaries of 28-day-old rats TNF-alpha concentrations were highest before injection of eCG (approximately 1.2 fg micrograms-1 ovarian protein) and decreased to approximately 0.4 fg microgram-1 protein 3 h after injection. TNF-alpha concentrations decreased further 24 h after eCG injection (< 0.1 fg microgram-1 protein) and remained low until 48 h after eCG injection. Serum concentrations of TNF-alpha did not change during the 48 h period after injection of eCG. hCG was administered 48 h after eCG, and ovarian and serum TNF-alpha concentrations increased transiently. Serum TNF-alpha concentrations increased 3 h after hCG and remained elevated until 9 h after injection, after which concentrations decreased. Ovarian concentrations of TNF-alpha increased 6 h after hCG, peaked (approximately 0.5 fg microgram-1 protein), and then declined. These results indicate that during puberty and the first ovulation, circulating and ovarian TNF-alpha concentrations change. In addition, exogenous gonadotrophins alter circulating and ovarian TNF-alpha concentrations. These data suggest that TNF-alpha has a role in follicular development and ovulation during puberty and in immature rats treated with gonadotrophins to induce ovulation.


Assuntos
Gonadotropina Coriônica/farmacologia , Gonadotropinas Equinas/farmacologia , Ovário/metabolismo , Maturidade Sexual/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Envelhecimento , Animais , Bioensaio , Feminino , Ovulação/efeitos dos fármacos , Ovulação/metabolismo , Ratos , Ratos Sprague-Dawley , Maturidade Sexual/efeitos dos fármacos , Estimulação Química
18.
Biol Reprod ; 59(6): 1385-91, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9828182

RESUMO

This study was designed to determine the source of tumor necrosis factor (TNF) alpha within the porcine corpus luteum (CL). 1) Sections of frozen or paraffin-embedded CL from various stages of the estrous cycle were incubated with the following primary antibodies: anti-human recombinant TNFalpha, anti-porcine macrophage-specific antigen, or anti-alpha-actin (marker of pericyte and smooth muscle cells). Dolichos biflorus lectin-peroxidase was used as an endothelial cell label. Positive immunostaining for TNFalpha was apparent in porcine CL throughout the estrous cycle. TNFalpha immunoreactivity was primarily localized in cells along septal/vascular tracts, and exhibited spatial and temporal distribution similar to that of cells labeled with anti-macrophage antibodies. Large luteal cells exhibited weak staining for TNFalpha in paraffin sections, whereas microvascular endothelial cells were consistently negative in both frozen and paraffin sections. 2) Enriched subpopulations of macrophages, endothelial cells, and large and small luteal cells were isolated by density gradient and immunomagnetic bead separation techniques. TNFalpha secretion by each subpopulation was determined by measuring bioactive TNFalpha in incubation media using a specific in vitro bioassay. Macrophage subpopulations secreted up to 100-fold greater quantities of bioactive TNFalpha (up to 400 pg/10(6) cells) than did other subpopulations. In contrast, endothelial cell and small luteal cell subpopulations released very small amounts (< 8 pg/10(6) cells) of bioactive TNFalpha. Large luteal cells secreted slightly greater amounts of TNFalpha (10-15 pg/10(6) cells). Local macrophages appear to be the primary source of TNFalpha in the porcine CL.


Assuntos
Corpo Lúteo/citologia , Macrófagos/metabolismo , Suínos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Corpo Lúteo/irrigação sanguínea , Corpo Lúteo/química , Endotélio Vascular/química , Estro , Feminino , Imuno-Histoquímica , Separação Imunomagnética , Macrófagos/química , Fator de Necrose Tumoral alfa/análise
19.
Hum Reprod ; 13(5): 1285-91, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9647561

RESUMO

This study determined effects of follicle stimulating hormone (FSH) alone and in combination with tumour necrosis factor (TNF), on granulosa cells from small (5-10 mm diameter) and large (>10-25 mm) follicles during follicular and luteal phases of the cycle and during periods of acyclicity. Granulosa cells were collected from ovaries of premenopausal women undergoing oophorectomy. The cells were cultured with human FSH (2 ng/ml) and testosterone (1 microM) in the presence or absence of human TNF-alpha (20 ng/ml). Media were removed at 48 and 96 h after culture and progesterone, oestradiol and cAMP in media were measured by radioimmunoassays. FSH stimulated the accumulation of oestradiol from granulosa cells of small follicles during the follicular and luteal phases but not during acyclicity; and TNF reduced oestradiol accumulation in the presence of FSH. Interestingly, in granulosa cells from small follicles, progesterone and cAMP secretion increased in response to FSH and neither was affected by TNF. Thus, TNF specifically inhibited the conversion of testosterone to oestradiol in granulosa cells from small follicles. FSH stimulated oestradiol production by granulosa cells of large follicles obtained only during the follicular phase of the cycle and TNF inhibited the FSH-induced oestradiol secretion. Granulosa cells obtained from large follicles during the luteal phase and during acyclicity did not accumulate oestradiol in response to FSH. However, FSH increased progesterone and cAMP secretion by granulosa cells obtained from large follicles during the follicular and luteal phases. During the luteal phase alone, TNF in combination with FSH increased progesterone accumulation above that of FSH alone. FSH did not increase progesterone, oestradiol or cAMP secretion by granulosa cells obtained from large follicles during acyclicity. Thus, FSH increases progesterone, oestradiol and cAMP secretion by granulosa cells of small follicles during the follicular and luteal phases and TNF appears to inhibit FSH-induced oestradiol secretion specifically in those cells. In large follicles, FSH-stimulated granulosa cell secretion of oestradiol is limited to the follicular phase and this effect can be inhibited by TNF. In addition, when granulosa cells of large follicles do not increase oestradiol secretion in response to FSH, TNF stimulates progesterone secretion.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/fisiologia , Folículo Ovariano/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Adulto , AMP Cíclico/metabolismo , Interações Medicamentosas , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/administração & dosagem , Fase Folicular/fisiologia , Humanos , Técnicas In Vitro , Fase Luteal/fisiologia , Pessoa de Meia-Idade , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia , Progesterona/metabolismo , Fator de Necrose Tumoral alfa/administração & dosagem
20.
Brain Res Dev Brain Res ; 107(1): 49-55, 1998 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-9602055

RESUMO

To investigate the effect of a delay in ovulation on postnatal growth and development in resultant rat offspring, a 1-day ovulatory delay was induced by sodium pentobarbital, animals mated, and the offspring monitored. There were no differences between control and 1-day delayed offspring in the number of live or dead births, number of males or females, nor in the ratio of sexes. Delayed pups had a slightly lower birth weight, but then recovered to weigh more than controls by day 12. In the first two weeks post-parturition, delayed pups displayed an earlier ability to reorient themselves in a negative geotaxis test, but no differences by the righting reflex and reflex suspension tests. At postnatal day (pnd) 28, delayed pups exhibited decreased activity in a continuous corridor test, but no alterations in gait. At this time, the brains of delayed animals revealed thickening of cortical layers V plus VI. There were significant correlations between various developmental endpoints (body weight, negative geotaxis, continuous corridor activity, and gait) and the cortical layer thicknesses. The results indicate that ovulatory delay produces changes in brain cortical thickness, with correlative changes in growth and behavior. Although the mechanisms by which ovulatory delay alters postnatal development and brain structure are unknown, ovulatory delay may alter the uterine environment during early pregnancy.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/psicologia , Comportamento Animal/fisiologia , Encéfalo/anatomia & histologia , Ovulação/fisiologia , Animais , Animais Recém-Nascidos/anatomia & histologia , Encéfalo/crescimento & desenvolvimento , Córtex Cerebral/anatomia & histologia , Feminino , Marcha/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Análise de Regressão , Fatores de Tempo , Aumento de Peso/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA