Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Front Endocrinol (Lausanne) ; 13: 989523, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36329893

RESUMO

Objective: Obesity increases the risk of certain cancers, especially tumours that reside close to adipose tissue (breast and ovarian metastasis in the omentum). The obesogenic and tumour micro-environment share a common pathogenic feature, oxygen deprivation (hypoxia). Here we test how hypoxia changes the metabolome of adipocytes to assist cancer cell growth. Methods: Human and mouse breast and ovarian cancer cell lines were co-cultured with human and mouse adipocytes respectively under normoxia or hypoxia. Proliferation and lipid uptake in cancer cells were measured by commercial assays. Metabolite changes under normoxia or hypoxia were measured in the media of human adipocytes by targeted LC/MS. Results: Hypoxic cancer-conditioned media increased lipolysis in both human and mouse adipocytes. This led to increased transfer of lipids to cancer cells and consequent increased proliferation under hypoxia. These effects were dependent on HIF1α expression in adipocytes, as mouse adipocytes lacking HIF1α showed blunted responses under hypoxic conditions. Targeted metabolomics of the human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes media revealed that culture with hypoxic-conditioned media from non-malignant mammary epithelial cells (MCF10A) can alter the adipocyte metabolome and drive proliferation of the non-malignant cells. Conclusion: Here, we show that hypoxia in the adipose-tumour microenvironment is the driving force of the lipid uptake in both mammary and ovarian cancer cells. Hypoxia can modify the adipocyte metabolome towards accelerated lipolysis, glucose deprivation and reduced ketosis. These metabolic shifts in adipocytes could assist both mammary epithelial and cancer cells to bypass the inhibitory effects of hypoxia on proliferation and thrive.


Assuntos
Adipócitos , Neoplasias Ovarianas , Humanos , Camundongos , Animais , Feminino , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Adipócitos/metabolismo , Hipóxia/metabolismo , Hipóxia/patologia , Proliferação de Células , Lipídeos/farmacologia , Neoplasias Ovarianas/metabolismo , Microambiente Tumoral
2.
Oncogene ; 36(18): 2565-2576, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27869162

RESUMO

Epithelial ovarian cancer (EOC) has poor prognosis and rapid recurrence because of widespread dissemination of peritoneal metastases at diagnosis. Multiple pathways contribute to the aggressiveness of ovarian cancer, including hypoxic signaling mechanisms. In this study, we have determined that the hypoxia-inducible histone demethylase KDM4B is expressed in ∼60% of EOC tumors assayed, including primary and matched metastatic tumors. Expression of KDM4B in tumors is positively correlated with expression of the tumor hypoxia marker CA-IX, and is robustly induced in EOC cell lines exposed to hypoxia. KDM4B regulates expression of metastatic genes and pathways, and loss of KDM4B increases H3K9 trimethylation at the promoters of target genes like LOXL2, LCN2 and PDGFB. Suppressing KDM4B inhibits ovarian cancer cell invasion, migration and spheroid formation in vitro. KDM4B also regulates seeding and growth of peritoneal tumors in vivo, where its expression corresponds to hypoxic regions. This is the first demonstration that a Jumonji-domain histone demethylase regulates cellular processes required for peritoneal dissemination of cancer cells, one of the predominant factors affecting prognosis of EOC. The pathways regulated by KDM4B may present novel opportunities to develop combinatorial therapies to improve existing therapies for EOC patients.


Assuntos
Biomarcadores Tumorais/genética , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias Ovarianas/genética , Neoplasias Peritoneais/genética , Peritônio/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Proteínas de Neoplasias/biossíntese , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Prognóstico , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
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
4.
Endocrinology ; 142(6): 2328-35, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356679

RESUMO

Immature female rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) before gonadotropin-induced follicle development and ovulation ovulate significantly fewer ova compared with controls. This study was designed to investigate potential ovarian-specific mechanisms of TCDD-mediated inhibition of ovulation. Immature hypophysectomized rats were treated with TCDD (32 microg/kg) or corn oil vehicle. Follicle development was initiated by injection of 10 IU PMSG 24 h after TCDD, and ovulation was induced 52 h after PMSG by injection of 10 IU hCG. The number of ova flushed from the oviduct was assessed the morning after hCG injection, and ovaries were collected at multiple times throughout the treatment schedule for histological analysis and analysis of FSH and hCG receptor binding and ovarian cAMP levels. In addition, serum levels of estradiol and progesterone were determined. Control rats ovulated 9.3 +/- 1.5 ova, whereas TCDD-treated rats ovulated 0.6 +/- 0.3. Gonadotropin receptor binding was evaluated 52 h after PMSG at the usual time of hCG injection to induce ovulation. Both FSH binding and hCG binding were significantly reduced in animals treated with TCDD. Serum estradiol levels in control animals were increased by 52 h after PMSG administration. In contrast, serum levels of estradiol in TCDD-treated animals remained low. In response to the ovulatory dose of hCG, serum levels of both estradiol and progesterone increased in control animals. Steroid levels also increased in TCDD-treated animals, but did not reach the peak levels observed in controls. TCDD treatment further resulted in lower ovarian cAMP levels at 52 h post-PMSG and at 5 h post-hCG. Together the data indicate that TCDD treatment altered the ability of the ovary to respond to PMSG, resulting in the development of follicles not comparable to controls (lower gonadotropin binding, lower estradiol production, lower levels of cAMP). It appears that critical steps in the development and maturation of follicles are disrupted by TCDD.


Assuntos
Estradiol/biossíntese , Folículo Ovariano/crescimento & desenvolvimento , Ovulação , Progesterona/biossíntese , Receptores da Gonadotropina/metabolismo , Animais , Membrana Celular/metabolismo , Gonadotropina Coriônica/metabolismo , Gonadotropina Coriônica/farmacologia , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/metabolismo , Gonadotropinas Equinas/farmacologia , Tamanho do Órgão , Ovário/anatomia & histologia , Ovário/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacologia , Progesterona/sangue , Ratos , Ratos Sprague-Dawley
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
J Reprod Immunol ; 35(2): 87-99, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9421794

RESUMO

Estrogens and progesterone could be among the environmental signals that govern uterine immune cell synthesis of pro-inflammatory substances. In order to investigate this possibility, we first mapped expression of the inducible nitric oxide synthase (iNOS) and tumor necrosis factor-alpha (TNF-alpha) genes in the leukocytes of cycling and pregnant mouse uteri, then tested the ability of estradiol-17 beta (E2) and progesterone to influence gene expression. Immunohistochemistry, in situ hybridization, and other experimental approaches, revealed that the iNOS and TNF-alpha genes are expressed in mouse uterine mast cells, macrophages and natural killer cells (uNK). Gene expression in each cell type was noted to be dependent upon stage of the cycle or stage of gestation, implying potential relationships with levels of female hormones and state of cell differentiation or activation. Further in vivo and in vitro experiments showed that individual hormones have cell type-specific effects on synthesis of iNOS and TNF-alpha that are exerted at the level of transcription. In uterine mast cells, iNOS and TNF-alpha are promoted by E2 whereas preliminary studies in macrophages suggest that transcription and translation of the two genes are unaffected by E2 but are inhibited by progesterone. Uterine NK cell production of iNOS and TNF-alpha is strongly related to cell differentiation, which is initiated and sustained by progesterone. Collectively, the results indicate that regulation of synthesis of pro-inflammatory molecules by hematopoietic cells in cycling and pregnant uterus comprises a new and potentially critical role for female steroid hormones.


Assuntos
Estradiol/farmacologia , Leucócitos/metabolismo , Óxido Nítrico Sintase/biossíntese , Progesterona/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Feminino , Humanos , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Mamíferos , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Gravidez , Fator de Necrose Tumoral alfa/genética , Útero/citologia , Útero/metabolismo
15.
J Reprod Fertil ; 106(2): 285-90, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8699412

RESUMO

Signals transduced by binding of tumour necrosis factor alpha (TNF) and lymphotoxin alpha (LT-alpha) trimers to high-affinity cell membrane receptors, TNF-RI (p55/p60) and TNF-RII (p75/p80), affect many cell functions. In this study, expression of the gene encoding TNF-RI in uteri of cyclic mice was mapped using in situ hybridization. TNF-RI hybridization signals fluctuated during the cycle. Signal intensity was highest during dioestrus-II, when mRNA encoding TNF-RI was present in endometrial epithelial and stroma cells, as well as in myometrial smooth muscle and connective tissue cells. The ability of oestradiol and progesterone to modulate steady state concentrations of mRNA encoding TNF-RI in uterine cells was assessed by using in situ and northern blot hybridization procedures. Seven days after ovariectomy, low concentrations of mRNA encoding TNF-RI were detected by northern analysis and weak in situ hybridization signals were identified in epithelia and some myometrial connective tissue cells. Administration of oestradiol, progesterone or oestradiol plus progesterone to ovariectomized animals stimulated temporal and cell type-specific changes in steady state concentrations of mRNA encoding TNF-RI that were unique to each hormonal regimen. Maximal induction of mRNA encoding TNF-RI required 24 h of oestradiol stimulation and 72 h of progesterone stimulation. In uteri treated with oestradiol plus progesterone, the oestradiol pattern predominated over the progesterone pattern. Thus, multiple cell types in cyclic mouse uteri express the gene encoding TNF-RI, and expression in specific cells is controlled by female steroid hormones.


Assuntos
Antígenos CD/análise , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônios Esteroides Gonadais/farmacologia , RNA Mensageiro/análise , Receptores do Fator de Necrose Tumoral/análise , Útero/metabolismo , Animais , Antígenos CD/genética , Northern Blotting , Epitélio/metabolismo , Estradiol/farmacologia , Feminino , Hibridização In Situ , Camundongos , Camundongos Endogâmicos , Miométrio/metabolismo , Ovariectomia , Progesterona/farmacologia , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral
16.
Placenta ; 17(1): 27-32, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8710809

RESUMO

Heterodimers of transporter for antigen processing proteins, Tap-1 and Tap-2, are essential components of the pathway that leads to expression of conventional HLA-A, -B class I transplantation antigens on cell surfaces. In this study, expression of the Tap-1 gene in trophoblast cells, some of which display novel and unconventional HLA class I molecules that include HLA-G and an HLA-C-like antigen, was investigated by using in situ hybridization to identify Tap-1 mRNA and immunohistochemistry to detect Tap-1 protein. The experiments were done on semiserial sections of paraformaldehyde-fixed tissues. In first trimester placentas, expression of the Tap-1 gene correlated with expression of HLA class I antigens in trophoblast cells. HLA-G/C positive extravillous cytotrophoblast cells exhibited high intensity in situ hybridization signals for Tap-1 mRNA and strong staining with anti-Tap-1 whereas Tap-1 gene products were rarely detected in HLA class I antigen negative syncytiotrophoblast and villous cytotrophoblast cells. Relationships were less definitive in term tissues. Although Tap-1 protein was detectable in extravillous cytotrophoblast cells (chorionic cytotrophoblast cells) as expected, HLA class I negative syncytiotrophoblast contained low intensity hybridization and immunostaining signals. Collectively, the data suggest that (1) as with conventional HLA class I antigens in other types of cells, the pathway leading to expression of novel HLA class I antigens in trophoblast cells includes transport of peptides by Tap-1, and that (2) deficiencies in Tap-1 might account in (whole or in) part for the failure of some trophoblast cells to express HLA class I antigens.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Expressão Gênica , Trofoblastos/metabolismo , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/análise , Feminino , Idade Gestacional , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imuno-Histoquímica , Hibridização In Situ , Placenta/química , Gravidez , RNA Mensageiro/análise , Trofoblastos/citologia
17.
Immunology ; 86(3): 469-74, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8550087

RESUMO

Human leucocyte antigen (HLA) class I antigen expression is closely controlled in placental trophoblast cells, which interface directly with genetically disparate maternal blood and tissues during pregnancy. In this study, the possibility that LMP7, a proteasome component that may be required for processing of class I-associated peptides, might be lacking or refractory to cytokine induction in trophoblast cells that fail to display HLA class I antigens was investigated. Analysis of Lmp7 mRNA and protein in paraformaldehyde-fixed placentas by in situ hybridization and immunohistochemistry revealed that both HLA class I-positive and HLA class I-negative trophoblast cells contain Lmp7 gene products. Consistent with these results, northern blot hybridization studies showed that HLA class I-positive (JEG-3) and HLA null (Jar) trophoblast-derived cell lines contain Lmp7 mRNA. After 48 hr of exposure to HLA class I-modulating cytokines, Lmp7 mRNA levels in JEG-3 cells were markedly increased by two interferons (IFN-beta, IFN-gamma) and tumour necrosis factor (TNF) whereas at the same time point, Jar cell Lmp7 mRNA was modestly enhanced by IFN-gamma. Collectively, the findings indicate that expression of HLA class I antigens in trophoblast cells is unlikely to be restricted by lack of Lmp7 gene products and suggest that endogenous placental cytokines may have different influences on Lmp7 mRNA levels in phenotypically distinct trophoblast subpopulations.


Assuntos
Cisteína Endopeptidases , Complexo Principal de Histocompatibilidade , Complexos Multienzimáticos , Placenta/química , Proteínas/análise , RNA Mensageiro/análise , Northern Blotting , Linhagem Celular , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Interferon beta/farmacologia , Interferon gama/farmacologia , Placenta/citologia , Gravidez , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Complexo de Endopeptidases do Proteassoma , Proteínas/genética , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
18.
Proc Soc Exp Biol Med ; 209(4): 325-42, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7638240

RESUMO

Tumor necrosis factor-alpha (TNF), originally identified as an inflammation-associated cytokine, is synthesized throughout the female reproductive tract as well as in placentas and embryos. Development, female sex steroid hormones, and lipopolysaccharide influence expression of this gene. The functions of TNF may be determined in part by differential expression of the two species of TNF receptors, both of which seem to be regulated by female sex steroid hormones. Evidence has accumulated that supports a role for this potent, pleiotropic cytokine in autocrine and paracrine processes central to reproduction, including gamete and follicle development, steroidogenesis, uterine cyclicity, placental differentiation, development of the embryo, and parturition.


Assuntos
Genitália Feminina/fisiologia , Ovário/fisiologia , Reprodução , Fator de Necrose Tumoral alfa/fisiologia , Animais , Embrião de Mamíferos/química , Embrião de Mamíferos/fisiologia , Feminino , Regulação da Expressão Gênica , Humanos , Ovário/química , Ovário/citologia , Ovário/efeitos dos fármacos , Placenta/química , Gravidez , Receptores do Fator de Necrose Tumoral/análise , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Útero/fisiologia
19.
Biol Reprod ; 52(3): 509-15, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7756446

RESUMO

The expression of tumor necrosis factor alpha (TNF) protein in mouse myometrial cells on each day of the estrous cycle and after ovariectomy, with and without replacement of estradiol (E2), progesterone (P4), or E2 + P4, was investigated. TNF protein was assessed by immunocytochemistry. In addition, the numbers of mast cells and the numbers of TNF-containing mast cells were determined under each condition. The total number of mast cells fluctuated during the estrous cycle; and at each stage, essentially 100% of the mast cells exhibited TNF immunoreactivity. In contrast, after ovariectomy and after ovariectomy with E2 replacement, only approximately 50% of the mast cells contained immunoreactive TNF. P4 treatment resulted in further decline in mast cell TNF whereby after 24 h of P4, only 3% of the mast cells were TNF-positive and by 72 h of treatment, no TNF-positive mast cells could be detected E2 and P4 in combination increased the total number of mast cells, and in a pattern reminiscent of normal cycling myometrium, over 90% of the mast cells exhibited TNF immunoreactivity after 24 h of treatment. TNF protein was detectable in muscle cells throughout the estrous cycle, with weak immunostaining observed on proestrus and more intense immunostaining on estrus, diestrus-I, and diestrus-II. After ovariectomy, only light immunostaining was observed in the muscle cells. Immunoreactive TNF was detected in muscle cells after each type of steroid treatment. E2 and E2 + P4 treatments resulted in a biphasic pattern of immunoreactive TNF expression.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Estradiol/fisiologia , Miométrio/metabolismo , Progesterona/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Estradiol/farmacologia , Estro/efeitos dos fármacos , Estro/fisiologia , Feminino , Imuno-Histoquímica , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Camundongos , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Miométrio/ultraestrutura , Ovariectomia , Progesterona/farmacologia
20.
J Leukoc Biol ; 57(1): 27-35, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7530281

RESUMO

Nitric oxide (NO), a potent and versatile free radical, is synthesized in macrophages and mast cells as well as in other types of cells by the inducible form of nitric oxide synthase (iNOS). In this study, cells containing iNOS were identified in the uteri of cycling mice by using a rabbit antibody generated to an iNOS-specific peptide. Macrophages were identified in semiserial sections of the same tissues with the monoclonal antibody, F4/80, and mast cells were identified by toluidine blue staining. In tissue sections of uteri obtained from mice in the four stages of the estrous cycle (8 to 11 mice per stage), iNOS immunoreactivity was strongest in diestrus-I uteri and weakest in diestrus-II uteri. Myometrial mast cells and endometrial epithelial cells were prominent locations of iNOS, and specific protein was also present in myometrial smooth muscle and macrophage-like cells in the endometrial stroma. Because cyclic variations suggested regulation of iNOS expression by ovarian steroid hormones, studies were done using ovariectomized mice. Seven days after ovariectomy, immunoreactive iNOS was low but detectable in mast cells and luminal epithelial cells. In the uteri of ovariectomized, estradiol-17 beta (E2)-treated mice, mast cells were iNOS+ after 24 h whereas epithelial cells were negative; the reverse was observed in progesterone (P4)-treated mice. Both mast cells and epithelial cells were iNOS+ in the uteri of mice that had received a combination of E2 + P4. These results indicate that several types of uterine cells produce iNOS and that expression of this enzyme in specific cell lineages is governed by ovarian steroid hormones. The data are consistent with the postulate that NO derived from uterine leukocytes and other types of cells plays a role in uterine cyclicity and preparation for pregnancy.


Assuntos
Aminoácido Oxirredutases/análise , Aminoácido Oxirredutases/biossíntese , Estradiol/farmacologia , Progesterona/farmacologia , Útero/enzimologia , Animais , Western Blotting , Endométrio/citologia , Endométrio/enzimologia , Indução Enzimática/efeitos dos fármacos , Estro/fisiologia , Feminino , Histocitoquímica , Macrófagos/citologia , Macrófagos/enzimologia , Mastócitos/citologia , Mastócitos/enzimologia , Camundongos , Miométrio/citologia , Miométrio/enzimologia , NADPH Desidrogenase/análise , Óxido Nítrico Sintase , Útero/citologia , Útero/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA