Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Oncogene ; 31(12): 1504-20, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21860425

RESUMO

WNT, RAS or phosphoinositide 3-kinase signaling pathways control specific stages of ovarian follicular development. To analyze the functional interactions of these pathways in granulosa cells during follicular development in vivo, we generated specific mutant mouse models. Stable activation of the WNT signaling effector ß-catenin (CTNNB1) in granulosa cells results in the formation of premalignant lesions that develop into granulosa cell tumors (GCTs) spontaneously later in life or following targeted deletion of the tumor suppressor gene Pten. Conversely, expression of oncogenic KRAS(G12D) dramatically arrests proliferation, differentiation and apoptosis in granulosa cells, and consequently, small abnormal follicle-like structures devoid of oocytes accumulate in the ovary. Because of the potent anti-proliferative effects of KRAS(G12D) in granulosa cells, we sought to determine whether KRAS(G12D) would block precancerous lesion and tumor formation in follicles of the CTNNB1-mutant mice. Unexpectedly, transgenic Ctnnb1;Kras-mutant mice exhibited increased GC proliferation, decreased apoptosis and impaired differentiation and developed early-onset GCTs leading to premature death in a manner similar to the Ctnnb1;Pten-mutant mice. Microarray and reverse transcription-PCR analyses revealed that gene regulatory processes induced by CTNNB1 were mostly enhanced by either KRAS activation or Pten loss in remarkably similar patterns and degree. The concomitant activation of CTNNB1 and KRAS in Sertoli cells also caused testicular granulosa cell tumors that showed gene expression patterns that partially overlapped those observed in GCTs of the ovary. Although the mutations analyzed herein have not yet been linked to adult GCTs in humans, they may be related to juvenile GCTs or to tumors in other tissues where CTNNB1 is mutated. Importantly, the results provide strong evidence that CTNNB1 is the driver in these contexts and that KRAS(G12D) and Pten loss promote the program set in motion by the CTNNB1.


Assuntos
Tumor de Células da Granulosa/genética , Proteínas de Membrana/genética , Neoplasias Ovarianas/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas/genética , Neoplasias Testiculares/genética , beta Catenina/genética , Proteínas ras/genética , Animais , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas p21(ras)
2.
Oncogene ; 30(32): 3522-36, 2011 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-21423204

RESUMO

Ovarian cancer is a complex and deadly disease that remains difficult to detect at an early curable stage. Furthermore, although some oncogenic (Kras, Pten/PI3K and Trp53) pathways that are frequently mutated, deleted or amplified in ovarian cancer are known, how these pathways initiate and drive specific morphological phenotypes and tumor outcomes remain unclear. We recently generated Pten(fl/fl); Kras(G12D); Amhr2-Cre mice to disrupt the Pten gene and express a stable mutant form of Kras(G12D) in ovarian surface epithelial (OSE) cells. On the basis of histopathologic criteria, the mutant mice developed low-grade ovarian serous papillary adenocarcinomas at an early age and with 100% penetrance. This highly reproducible phenotype provides the first mouse model in which to study this ovarian cancer subtype. OSE cells isolated from ovaries of mutant mice at 5 and 10 weeks of age exhibit temporal changes in the expression of specific Mullerian epithelial marker genes, grow in soft agar and develop ectopic invasive tumors in recipient mice, indicating that the cells are transformed. Gene profiling identified specific mRNAs and microRNAs differentially expressed in purified OSE cells derived from tumors of the mutant mice compared with wild-type OSE cells. Mapping of transcripts or genes between the mouse OSE mutant data sets, the Kras signature from human cancer cell lines and the human ovarian tumor array data sets, documented significant overlap, indicating that KRAS is a key driver of OSE transformation in this context. Two key hallmarks of the mutant OSE cells in these mice are the elevated expression of the tumor-suppressor Trp53 (p53) and its microRNA target, miR-34a-c. We propose that elevated TRP53 and miR-34a-c may exert negatively regulatory effects that reduce the proliferative potential of OSE cells leading to the low-grade serous adenocarcinoma phenotype.


Assuntos
Transformação Celular Neoplásica/genética , Células Epiteliais/metabolismo , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma Epitelial do Ovário , Linhagem Celular Transformada , Células Cultivadas , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , MicroRNAs/genética , Transplante de Neoplasias , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ovário/citologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética
3.
Biol Reprod ; 68(5): 1895-902, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12606366

RESUMO

An increase in metallothionein 1 (MT-1) mRNA was detected in the ovaries of immature Wistar rats that were primed with s.c. injection of 10 IU eCG followed 48 h later by 10 IU hCG s.c. to initiate the ovulatory process. Ovarian RNA was extracted at 0, 2, 4, 8, 12, 24, 72, 144, and 288 h after the primed animals were injected with hCG. These extracts were used for reverse transcription polymerase chain reaction (RT-PCR) differential display and Northern analyses that yielded complementary gene fragments for MT-1. Expression of MT-1 mRNA increased significantly by 24 h after hCG treatment and reached a peak at 144 h after hCG. In contrast, a disintegrin and metalloproteinase with thrombospondin motifs and a tissue inhibitor of metalloproteinase 1, which were also detected by the RT-PCR differential display procedure, reached a peak at 12 h after hCG and returned to control levels in the ovaries by 72 h after hCG. In situ hybridization indicated that most of the MT-1 mRNA was expressed in the vicinity of the theca interna of preovulatory follicles and in the lutein granulosa of postovulatory follicles. Thus, MT-1 mRNA expression is primarily in the vicinity of steroid-secreting areas of the ovary. The substantial increase in MT-1 mRNA expression might be important in protecting the ovarian tissues from oxidative stress generated by ovarian inflammatory events during the ovulatory process and luteinization.


Assuntos
Metalotioneína/biossíntese , Ovário/metabolismo , Ovulação/fisiologia , RNA Mensageiro/biossíntese , Esteroides/metabolismo , Proteínas ADAM , Proteína ADAMTS1 , Abortivos/farmacologia , Androstenóis/farmacologia , Animais , Northern Blotting , Corpo Lúteo/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , DNA Complementar/biossíntese , DNA Complementar/genética , Dinoprostona/metabolismo , Desintegrinas/biossíntese , Feminino , Hibridização In Situ , Indometacina/farmacologia , Metaloendopeptidases/biossíntese , Folículo Ovariano/fisiologia , Ovário/citologia , Progesterona/metabolismo , Ratos , Ratos Wistar , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Transcrição Gênica
4.
Artigo em Inglês | MEDLINE | ID: mdl-12402539

RESUMO

The multiplicity of genes regulated by the actions of LH in specific granulosa cell and cumulus cell microenvironments of the follicle during ovulation has now revealed how complex and finely tuned the ovulation process is. Although many proteases are expressed in the ovary and are hormonally regulated, the novel proteases ADAMTS-1 and cathepsin L, rather than the MMPs, have gained particular recognition. These may play critical roles in the eventual rupture of the follicle at the ovarian surface. Rupture is only one part of ovulation. Another important aspect is the actual release of the oocyte from the ovulation pore. Several recent studies have shown that the production of the matrix that is evidenced by cumulus expansion is somehow critical for extrusion to occur and for the oocyte to travel into the oviduct. At any point the oocyte may be trapped within a non-ovulating structure. Lastly, if the events of ovulation fail to occur before the events of luteinization are complete, oocytes are destined to degenerate within the non-ovulating structures.


Assuntos
Oócitos/fisiologia , Folículo Ovariano/fisiologia , Oviductos/fisiologia , Ovulação/fisiologia , Proteínas de Peixe-Zebra , Proteínas ADAM , Proteína ADAMTS1 , Animais , Catepsina L , Catepsinas/metabolismo , Cisteína Endopeptidases , Desintegrinas/metabolismo , Feminino , Receptores Frizzled , Regulação da Expressão Gênica , Hormônio Luteinizante/metabolismo , Metaloendopeptidases/metabolismo , Folículo Ovariano/citologia , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt
5.
J Endocrinol ; 174(3): 485-92, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12208669

RESUMO

The ovulatory process in mammals involves gross physiological events in the ovary that cause transient deterioration of the ovarian connective tissue and rupture of the apical walls of mature follicles. This gonadotropin-induced process has features similar to an acute inflammatory reaction that affects most of the ovary. The present study reveals that the ovulatory events include induction of mRNA for pancreatitis-associated protein-III (PAP-III). Immature Wistar rats were primed with 10 IU equine chorionic gonadotropin s.c., and 48 h later the 12-h ovulatory process was initiated by 10 IU human chorionic gonadotropin (hCG) s.c. Ovarian RNA was extracted at 0, 2, 4, 8, 12 and 24 h after the animals were injected with hCG. The RNA extracts were used for RT-PCR differential display to detect PAP-III gene expression in the stimulated ovarian tissue. Northern blotting showed that transcription was significantly greater at 4-12 h after the ovaries had been stimulated by hCG. In situ hybridization indicated that PAP-III mRNA expression was limited mainly to the hilar region of the ovarian stroma, with most of the signal emanating from endothelial cells that lined the inner walls of blood vessels, and from small secondary follicles. Treatment of the animals with ovulation-blocking doses of indomethacin (an inhibitor of prostanoid synthesis) or epostane (an inhibitor of progesterone synthesis) revealed that ovarian transcription of PAP-III mRNA was moderately dependent on ovarian progesterone synthesis. In conclusion, the present evidence of an increase in PAP-III gene expression in gonadotropin-stimulated ovaries provides further evidence that the ovulatory process is comparable to an inflammatory reaction.


Assuntos
Antígenos de Neoplasias/genética , Biomarcadores Tumorais/genética , Gonadotropinas Equinas/farmacologia , Lectinas Tipo C/genética , Ovário/metabolismo , Ovulação/fisiologia , RNA Mensageiro/análise , Androstenóis/farmacologia , Animais , Northern Blotting , Feminino , Expressão Gênica , Indometacina/farmacologia , Proteínas Associadas a Pancreatite , Progesterona/antagonistas & inibidores , Antagonistas de Prostaglandina/farmacologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estimulação Química , Fatores de Tempo
6.
Am J Phys Med Rehabil ; 80(5): 366-70; quiz 371-3, 387, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11327559

RESUMO

OBJECTIVE: To examine the impact of gunshot-caused spinal cord injury on acute and rehabilitative care outcome using a case control design. DESIGN: Two groups (i.e., gunshot- vs. nongunshot-caused spinal cord injury) of 212 individuals were matched case-for-case on age (i.e., within 10 yr), education, gender, race, marital status, primary occupation, impairment level, and Model System region. Outcome measures included length of hospital stay, functional status (FIM), treatment charges, and home discharge rates. RESULTS: The two groups did not differ in the length of stay during acute and rehabilitative care, charges during rehabilitative care, or postrehabilitation discharge placement. Several significant between-group differences in treatment procedures were noted (e.g., prevalence of spinal surgery), which may, in part, account for the higher acute-care charges among those persons with nongunshot-caused spinal cord injury. CONCLUSION: Once an individual is stabilized and admitted for rehabilitative care, gunshot etiology of spinal cord injury seems largely unrelated to the initial rehabilitation outcome.


Assuntos
Traumatismos da Medula Espinal/etiologia , Traumatismos da Medula Espinal/reabilitação , Resultado do Tratamento , Ferimentos por Arma de Fogo/reabilitação , Adulto , Estudos de Casos e Controles , Escolaridade , Emprego , Feminino , Humanos , Tempo de Internação , Masculino , Estado Civil , Ocupações , Centros de Reabilitação/economia
7.
Mol Endocrinol ; 15(2): 209-18, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11158328

RESUMO

The glycoprotein hormones, ACTH, TSH, FSH, and LH regulate diverse functions in endocrine cells. Although cAMP and PKA have long been shown to mediate specific intracellular signaling events including the transcription of specific genes via the CREB-CBP complex, recent observations have indicated that PKA does not account for all of the intracellular targets of cAMP. For example, TSH stimulation of thyroid cell proliferation is not completely blocked by PKA inhibitors. TSH and FSH can stimulate PKB phosphorylation by a PKAindependent but PI3-K/PDK1-dependent pathway. An FSH inducible kinase, Sgk, has recently been shown to be a close relative of PKB. Sgk is also a target of PI3-K-PDK1 pathway, indicating that some effects previously ascribed to PKB may be mediated by this inducible kinase. The identification of novel cAMP-binding proteins that exhibit guanine nucleotide exchange (GEF) activity (cAMP-GEFS; Epacs) has open new doors for cAMP action that include activation of small GTPases such as Rap1a, Rap2, and possibly Ras. These GTPases are known activators of downstream kinase cascades, including p38MAPK and Erk1/2 as well as PI3-K. Thus, FSH and TSH activation of PKB and Sgk may occur via this alternative cAMP pathway that involves cAMP-GEFs and the activation of the PI3-K/PDK1 pathway.


Assuntos
AMP Cíclico/fisiologia , Glândulas Endócrinas/fisiologia , Hormônios Hipofisários/fisiologia , Transdução de Sinais , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/fisiologia , Animais , Diferenciação Celular , Divisão Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Glândulas Endócrinas/citologia , Feminino , Humanos , Masculino , Ovário/citologia , Ovário/fisiologia , Testículo/citologia , Testículo/fisiologia , Glândula Tireoide/citologia , Glândula Tireoide/fisiologia
8.
Steroids ; 65(10-11): 559-70, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11108860

RESUMO

The luteinizing hormone (LH) surge initiates a cascade of proteolytic events that control ovulation. One of the genes induced by LH is the progesterone receptor (PR). Because mice with a mutant PR gene (PRKO) fail to ovulate and are infertile, we have used them as a model in which to determine PR target genes that might mediate the ovulatory process. The matrix metalloproteinases (MMPs: MMP2, MMP9, and MMP13) appear to be expressed in ovaries of PRKO mice in a manner similar to that in their wild-type littermates. However, the expression of two other types of proteases, cathepsin L (a member of the papain family) and ADAMTS-1 (A Disintegrin And Metalloproteinase with Thrombospondin-like motifs), are selectively induced in granulosa cells of preovulatory follicles by the LH surge. Maximal levels of these proteases are observed at 12-16 h after an LH surge, the time of ovulation. Furthermore, mRNAs encoding cathepsin L and ADAMTS-1 are reduced in the PRKO mice compared to their wild-type littermates. These novel observations indicate that these two proteases regulate some key step(s) controlling ovulation.


Assuntos
Endopeptidases , Ovulação/genética , Ovulação/fisiologia , Proteínas ADAM , Proteína ADAMTS1 , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Catepsina L , Catepsinas/genética , Catepsinas/farmacologia , Ciclo-Oxigenase 2 , Cisteína Endopeptidases , Desintegrinas/genética , Desintegrinas/farmacologia , Feminino , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , Isoenzimas/genética , Isoenzimas/farmacologia , Hormônio Luteinizante/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Metaloproteinases da Matriz/efeitos dos fármacos , Metaloproteinases da Matriz/farmacologia , Metaloendopeptidases/genética , Metaloendopeptidases/farmacologia , Camundongos , Camundongos Knockout , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Regiões Promotoras Genéticas , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/farmacologia , RNA Mensageiro/metabolismo , Ratos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Transfecção
9.
Endocrinology ; 141(11): 4114-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11089543

RESUMO

Current evidence supports the hypothesis that the biochemical events of mammalian ovulation are analogous to an acute inflammatory reaction. This study reveals that tumor necrosis factor-stimulated gene-6 (TSG-6), which encodes a member of the superfamily of hyaluronan-binding proteins that is specifically translated in inflammatory reactions, is expressed in ovarian follicles that have been induced to ovulate. Immature Wistar rats were primed with 10 IU equine CG s.c.; and 48 h later, the 12-h ovulatory process was initiated by 10 IU human CG (hCG), s.c.. Ovarian RNA was extracted at 0, 2, 4, 8, 12, and 24 h after the primed animals were injected with hCG. The RNA extracts were used for RT-PCR differential display of amplified complementary DNAs (cDNAs) that represented gene expression in the stimulated ovarian tissue. Northern analysis of one of the differentially amplified cDNAs confirmed that it was part of a gene that was substantially up-regulated at 4-8 h after the ovaries had been stimulated by hCG. Subcloning and sequence analysis revealed that the cDNA matched the gene for TSG-6. In situ hybridization indicated that the TSG-6 messenger RNA was primarily located in the cumulus mass and the antral granulosa cells of large ovarian follicles. In conclusion, the data show that expression of TSG-6 is an integral part of the cascade of inflammatory-like changes that occur in an ovulatory follicle in response to a trophic hormone that couples with luteinizing hormone/hCG receptors.


Assuntos
Moléculas de Adesão Celular/genética , Gonadotropina Coriônica/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Ovário/metabolismo , Ovulação , Androstenóis/farmacologia , Animais , DNA Complementar/análise , DNA Complementar/química , Feminino , Hibridização In Situ , Indometacina/farmacologia , RNA Mensageiro/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
10.
J Biol Chem ; 275(43): 33718-28, 2000 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-10934195

RESUMO

AP1 transcription factors control rapid responses of mammalian cells to stimuli that impact proliferation, differentiation, and transformation. To determine which AP1 factors are present in and regulated by hormones in ovarian cells during specific stages of proliferation and differentiation, we used both in vitro and in vivo models, Western blotting, immunohistochemistry, DNA binding assays, and transfections of AP1 promoter-reporter constructs. The expression patterns of Jun and Fos family members in response to hormones (follicle-stimulating hormone (FSH), luteinizing hormone (LH), and cAMP) were distinct. JunB, c-Jun, c-Fos, and Fra2 were rapidly but transiently induced by FSH in immature granulosa cells. JunD and Fra2 were induced by LH and maintained as granulosa cells terminally differentiated into luteal cells. Forskolin and phorbol myristate acetate acted synergistically to enhance transcription of an AP1(-73COL)-luciferase construct. JunD appears to be one mediator of this effect, since JunD was a major component of the AP1-DNA binding complex in granulosa cells, and menin, a selective inhibitor of JunD, blocked transcription of -73COL-luciferase. Thus, FSH and LH via cAMP induce specific AP1 factors, the AP1 expression patterns are distinct, and that of JunD and Fra2 correlates with the transition of proliferating granulosa cells to terminally differentiated, non-dividing luteal cells.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Células da Granulosa/metabolismo , Proteínas Proto-Oncogênicas c-jun/fisiologia , Fator de Transcrição AP-1/biossíntese , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Colforsina/farmacologia , DNA/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Antígeno 2 Relacionado a Fos , Imuno-Histoquímica , Hormônio Luteinizante/farmacologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Ratos , Ratos Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-1/análise , Transgenes
11.
Mol Endocrinol ; 14(8): 1283-300, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10935551

RESUMO

FSH stimulates in ovarian granulosa cells diverse, differentiation-dependent responses that implicate activation of specific cellular signaling cascades. In these studies three kinases were investigated to determine their relationship to FSH, cAMP, and A kinase signaling: protein kinase B (PKB/Akt), serum and glucocorticoid-induced kinase (Sgk), and p38 mitogen-activated protein kinase (p38MAPK). The phosphorylation (activation) of these kinases was analyzed by using selective agonists/inhibitors: forskolin/H89 for cAMP-dependent protein kinase (A kinase), insulin-like growth factor I (IGF-I)/LY294002 and wortmannin for phosphatidylinositol-dependent kinase (PI3-K), and phorbol myristate (PMA)/GF109203X for diacylglycerol and Ca++-dependent kinases (C kinases). An inhibitor (PD98059) of MEK1, which regulates extracellular regulated kinases (ERKs), and SB203580, which inhibits p38MAPK, were also used. In addition, we analyzed the expression of the recently described, cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFI and GEFII) that impact Ras-related GTPases and Raf kinases, known regulators of various protein kinase cascades. We provide evidence that FSH, forskolin, and 8-bromo-cAMP stimulate phosphorylation of PKB by mechanisms involving PI3-K (LY294002/wortmannin sensitive) not A kinase (H89 insensitive), a pattern of response mimicking that of IGF-I. In contrast, FSH induction and phosphorylation of Sgk protein requires A kinase (H89 sensitive) but also involves PI3-K (LY294002 sensitive) as well as p38MAPK (SB203580 sensitive) pathways. PMA (C kinase) abolished FSH-mediated (but not IGF-I-mediated) phosphorylation of PKB at a step(s) upstream of PI3-K and independent of A kinase. Lastly, FSH-mediated phosphorylation of p38MAPK is negatively affected by A kinase and PI3-K, suggesting that it may be downstream of specific members of the cAMP-GEF/Rap/Raf pathway. We propose that cAMP activation of A kinase is obligatory for transcription of Sgk in granulosa cells whereas cAMP (IGF-I-like)-mediated phosphorylation (activation) of PKB and Sgk (via PI3-K), as well as p38MAPK, involves other cellular events. These results provide new and exciting evidence that cAMP acts in granulosa cells by A kinase-dependent and -independent mechanisms, each of which controls specific kinase cascades.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Feminino , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Imediatamente Precoces , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas de Protozoários/efeitos dos fármacos , Proteínas de Protozoários/metabolismo , Ratos , Ratos Sprague-Dawley , Testosterona/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
12.
Proc Natl Acad Sci U S A ; 97(9): 4689-94, 2000 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-10781075

RESUMO

Ovulation is a precisely timed process by which a mature oocyte is released from an ovarian follicle. This process is initiated by the pituitary surge of luteinizing hormone (LH), is temporally associated with transcriptional regulation of numerous genes, and is presumed to involve the synthesis and/or activation of specific proteases that degrade the follicle wall. The progesterone receptor (PR), a nuclear receptor transcription factor, is induced in granulosa cells of preovulatory follicles in response to the LH surge and has been shown to be essential for ovulation, because mice lacking PR fail to ovulate and are infertile. Using these mice as a model in which to elucidate PR-regulated genes in the ovulation process, we show that the matrix metalloproteinases MMP-2 and MMP-9 are not targets of PR during ovulation. In contrast, two other proteases, ADAMTS-1 (A disintegrin and metalloproteinase with thrombospondin-like motifs) and cathepsin L (a lysosomal cysteine protease), are transcriptional targets of PR action. ADAMTS-1 is induced after LH stimulation in granulosa cells of preovulatory follicles and depends on PR. Cathepsin L is induced in granulosa cells of growing follicles by follicle-stimulating hormone, but the highest levels of cathepsin L mRNA occur in preovulatory follicles in response to LH in a PR-dependent manner. The identification of two regulated proteases in the ovary, together with their abnormal expression in anovulatory PR knockout mice, suggests that each plays a critical role in follicular rupture and represents a major advance in our understanding of the proteolytic events that control ovulation.


Assuntos
Catepsinas/genética , Desintegrinas/genética , Endopeptidases , Regulação Enzimológica da Expressão Gênica , Metaloendopeptidases/genética , Ovário/fisiologia , Ovulação/genética , Progesterona/fisiologia , Receptores de Progesterona/fisiologia , Proteínas ADAM , Proteína ADAMTS1 , Animais , Catepsina L , Cisteína Endopeptidases , Feminino , Células da Granulosa/fisiologia , Hormônio Luteinizante/fisiologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Fenótipo , Receptores de Progesterona/deficiência , Receptores de Progesterona/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Mol Endocrinol ; 13(12): 2049-64, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10598581

RESUMO

PRL activates an important cytokine signaling cascade that is obligatory for maintaining luteal cell function in the rat ovary. To determine when specific components of this cascade are expressed and can be activated by PRL, we analyzed the expression of receptor subtypes (short, PRL-R(s), and long, PRL-R(L)), the presence and kinetics of Stat (signal transducer and activator of transcription) activation using the PRL-response element (PRL-RE) of the alpha2M (alpha2-macroglobulin) gene, and the content and hormonal regulation of three specific modulators of cytokine signaling; the tyrosine phosphatases (SHP-1 and SHP-2), and the protein inhibitor of activated Stat3 (PIAS-3). These components were analyzed in differentiating granulosa/ luteal cells of hypophysectomized (H) rats and in corpora lutea of pregnant rats. Levels of PRL-R mRNAs increased as granulosa cells differentiated and reached maximal levels in luteal cells of pregnant rats where levels of PRL-R(s) approached those of PRL-R(L). The relative concentrations shifted from a 27-fold excess of PRL-R(L) in preovulatory granulosa cells to a 3.7-fold difference in luteal cells during midgestation. Despite the increased PRL-R(L) expression in differentiated granulosa cells, PRL did not stimulate detectable activation of Stats. Rather PRL activation of Stat5, principally Stat5b, occurred in association with luteinization. In contrast, granulosa cells of untreated immature and H rats contained a high level of DNA binding activity, which was shown to be comprised entirely of activated, phosphorylated Stat3. Treatment with estrogen and FSH reduced the amount of phosphorylated Stat3 and abolished its ability to bind DNA, an effect temporally related to increased PIAS-3. Expression of SHP-1 (but not SHP-2) was also hormonally regulated; SHP-1 mRNA and protein were high in granulosa cells of H rats, decreased by estrogen and FSH, and subsequently increased dramatically with luteinization. Of particular note, SHP-1 was localized in cytoplasm of granulosa cells in atretic follicles but was distinctly nuclear in luteal cells, indicative of different functional roles. Collectively, these results indicate that Stat3 and Stat5 are activated by distinct cytokine-signaling pathways modulated through differentiation-dependent transcriptional regulation of signaling pathway components and mediate distinct functional processes in the rat ovary: early follicle growth and atresia vs. luteinization.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Proteínas do Leite , Prolactina/farmacologia , Transdução de Sinais , Transativadores/metabolismo , Animais , DNA/metabolismo , Proteínas de Ligação a DNA/análise , Feminino , Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Células Lúteas/citologia , Células Lúteas/efeitos dos fármacos , Ovário/química , Ovário/citologia , Fosforilação , Gravidez , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/análise , RNA Mensageiro/análise , Ratos , Receptores da Prolactina/genética , Receptores da Prolactina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3 , Fator de Transcrição STAT5 , Transativadores/análise
14.
Mol Endocrinol ; 13(8): 1318-37, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10446906

RESUMO

The responsiveness of granulosa cells to FSH (cAMP) changes as these cells switch from the proliferative stage in growing follicles to the terminally differentiated, nonproliferating stage after LH-induced luteinization. To analyze this transition, two well characterized culture systems were used. 1) Granulosa cells isolated from immature rats were cultured in serum-free medium, a system that permits analysis of dynamic, short-term responses to hormones/cAMP. 2) Granulosa cells from preovulatory (PO) follicles that have been exposed in vivo to surge concentrations of hCG (PO/ hCG) were cultured in medium containing 1% FBS, a system that permits analyses of cells that have undergo irreversible, long-term changes associated with luteinization. To analyze the biochemical basis for the switch in cAMP responsiveness, the localization of A-kinase pathway components was related to the expression of two cAMP target genes, aromatase (CYP19) and serum-and glucocorticoid-induced kinase (Sgk). Components of the A-kinase pathway were analyzed by Western blotting and indirect immunofluorescence using specific antibodies to the C subunit, RIIalpha/beta subunits, CREB (cAMP-regulatory element binding protein), phospho-CREB, CBP (CREB binding protein), and Sgk. Cellular levels of C subunit and CREB were similar in all cell types and hormone treatments. CREB and CBP were nuclear; RIIalpha/beta was restricted to a cytoplasmic basket-like structure. Addition of FSH to immature granulosa cells caused rapid nuclear import of C subunit within 1 h. Nuclear C subunit decreased by 6 h after FSH but could be rapidly reimported to the nucleus by the addition of forskolin at 6, 24, or 48 h. Nuclear C subunit was associated with the rapid but transient increases in phospho-CREB. FSH induced Sgk in a biphasic manner in which the protein was nuclear at 1 h and cytoplasmic at 48 h. Aromatase mRNA was only expressed at 24-48 h after FSH, a pattern that was not altered by phosphodiesterases or phosphatases. In the luteinized (PO/hCG) granulosa cells, immunoreactive C subunit was localized in a punctate pattern in the nucleus as well as to a cytoplasmic basket-like structure, a distribution pattern not altered by forskolin. Aromatase, Sgk, and phospho-CREB were expressed at elevated levels in a non-forskolin-responsive manner. Most notable, both phospho-CREB and Sgk were preferentially localized in a punctate pattern within the cytoplasm and not altered by forskolin. Collectively, these data indicate that when granulosa cells differentiate to luteal cells the subcellular localization (nuclear vs. cytoplasmic) of A-kinase pathway components changes markedly. Thus, either the mechanisms of nuclear import and export or the presence of distinct docking sites (and functions ?) dictate where A-kinase, phospho-CREB and Sgk are localized in granulosa cells compared with the terminally differentiated luteal cells.


Assuntos
Aromatase/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células da Granulosa/enzimologia , Células Lúteas/enzimologia , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Animais , Diferenciação Celular , Núcleo Celular/metabolismo , Colforsina/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Hormônio Foliculoestimulante/farmacologia , Expressão Gênica , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/ultraestrutura , Proteínas Imediatamente Precoces , Células Lúteas/ultraestrutura , Fragmentos de Peptídeos/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
15.
Endocrinology ; 140(9): 4320-34, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10465306

RESUMO

The expression and function of estrogen receptor ERalpha/beta subtypes and ERbeta variants in granulosa cells have been determined using several integrated approaches:, Western blotting, indirect immunofluorescence, RT-PCR, and transient transfection assays. Each of these approaches has provided specific details concerning the dynamics of ER expression, ER functional activity, and estradiol (E) regulation of target genes in granulosa cells. Specifically, the studies presented herein document that messenger RNAs (mRNAs) encoding ERbeta and its splice variants, as well as mRNA encoding ERalpha, are expressed in granulosa cells of immature rats before and during culture in serum-free medium. The results also provide the first documentation that functional (DNA binding and transcriptionally active) ER is present in cultured granulosa cells and that its ability to bind consensus estrogen response element (ERE) oligonucleotide and to transactivate an ERE promoter-reporter construct is associated with the level (type?) of receptor protein as well as the stage of granulosa cell differentiation. Using a labeled ERE consensus oligonucleotide and antibodies specific for ERbeta and ERalpha, we show that ERbeta but not ERalpha was detected (supershifted in electrophoretic mobility shift assays) in extracts of granulosa cells cultured overnight (0 h) in defined medium alone. When the cells were cultured with FSH and testosterone (T) to stimulate their differentiation, ERbeta binding activity, as well as immunoreactive ERbeta as determined by Western blot analyses, decreased progressively from 24 to 48 h and was undetectable by 72 h. ERbeta mRNA was low, and ERbeta binding activity was not observed in luteinized granulosa cells. ERalpha DNA binding activity was not observed in any of the granulosa cell cultures, although low levels of immunoreactive ERalpha were detected by Western blot analyses. Immunofluorescent analyses documented that ERbeta, as well as ERalpha, were localized to granulosa cell nuclei and that the intensity of nuclear staining was related to agonist stimulation and differentiation: forskolin increased, whereas E decreased immunostaining for ERbeta and ERalpha at 48 h. When an ERE-E1b-luciferase vector was transfected into granulosa cells of unprimed rats, basal luciferase activity was low but increased by forskolin (3-4x) and by E (2x), responses to both agonists being blocked by the ER antagonist, ICI. When the same vector was transfected into differentiated granulosa cells (cultured for 48 h with FSH/T), forskolin alone increased activity. Collectively, these results show that ERbeta protein is preferentially expressed in immature granulosa cells, is functionally active (binds DNA), can transactivate (either as a homodimer or heterodimer with ERalpha) ERE-containing promoter constructs, and might be associated with increased expression of the endogenous gene encoding c-Jun.


Assuntos
Células da Granulosa/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Células Cultivadas , Colforsina/farmacologia , DNA/metabolismo , Estradiol/farmacologia , Feminino , Imunofluorescência , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
16.
Mol Endocrinol ; 12(8): 1201-14, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9717846

RESUMO

Expression of progesterone receptor (PR) mRNA in granulosa cells of ovarian preovulatory follicles is induced by LH (1, 2) and is essential for ovulation (3). Although 17beta-estradiol (E) can induce PR mRNA and activate PR promoter-reporter constructs in other cell types, the effects of E in granulosa cells appear to be indirect. We show herein that E alone does not induce the expression of PR mRNA in preovulatory granulosa cells. Rather, induction of PR mRNA depends on the differentiation of granulosa cells in response to E and a physiological amount of FSH followed by exposure to agonists (elevated levels of LH, FSH, and forskolin) that markedly increase cAMP. Induction of PR mRNA by forskolin is blocked by the A-kinase inhibitor H89 and cycloheximide but not by the E antagonist, ICI 164,384. These results indicate that phosphorylation and synthesis of some regulatory factor(s) other than or in addition to the estrogen receptor (ER) are essential for transactivation of the PR gene. When distal and proximal PR promoter-reporter constructs that are responsive to E in other cell types were transiently transfected into differentiated granulosa cells, forskolin, but not E, induced activity. Likewise, when a vector containing the consensus vitellogenin B1 gene estrogen response element (ERE) was transfected into differentiated granulosa cells, forskolin, but not E, induced activity. Using electrophoretic mobility shift assays, the consensus ERE was shown to bind ERbeta, the predominant subtype present in rat granulosa cells, and ERalpha, the predominant subtype present in luteal cells, whereas the putative ERE-like region (ERE3) of the proximal PR promoter did not bind either ER subtype. Although the identity of the specific factors binding to the ERE3 site remain to be determined, mutation of this region abolished forskolin-induced activity of ERE3-PR-CAT constructs. The GC-rich region of the distal PR promoter bound Sp1 and Sp3 but not C/EBPalpha/beta, indicating that factors binding to ERE3 interact synergistically with Sp1/Sp3 to confer increased responsiveness of the distal promoter to forskolin. Taken together, these results indicate that activation of the A-kinase pathway leads to the phosphorylation of some transcription factor(s) other than or in addition to ER that is (are) critical for the transactivation of the PR gene and that this mechanism is selectively activated in differentiated granulosa cells possessing a preovulatory phenotype.


Assuntos
Monofosfato de Adenosina/metabolismo , Estradiol/farmacologia , Células da Granulosa/metabolismo , Receptores de Progesterona/genética , Sulfonamidas , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estradiol/análogos & derivados , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Isoquinolinas/farmacologia , Hormônio Luteinizante/metabolismo , Hormônio Luteinizante/farmacologia , Dados de Sequência Molecular , Ovulação/efeitos dos fármacos , Ovulação/fisiologia , Alcamidas Poli-Insaturadas , Regiões Promotoras Genéticas , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sequências Reguladoras de Ácido Nucleico
18.
Mol Endocrinol ; 12(7): 924-40, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9658398

RESUMO

The proliferation and terminal differentiation of granulosa cells are critical for normal follicular growth, ovulation, and luteinization. Therefore, the in situ localization and hormonal regulation of cell cycle activators (cyclin D1, D2, and D3) and cell cycle inhibitors (p27Kip1 and p21Cip1) were analyzed in ovaries of mice and rats at defined stages of follicular growth and differentiation. Cyclin D2 mRNA was specifically localized to granulosa cells of growing follicles, while cyclin D1 and cyclin D3 were restricted to theca cells. In hypophysectomized (H) rats, cyclin D2 mRNA and protein were increased in granulosa cells by treatment with estradiol or FSH and were increased maximally by treatment with both hormones. In serum-free cultures of rat granulosa cells, cyclin D2 mRNA was rapidly elevated in response to FSH, forskolin, and estradiol, indicating that estradiol as well as cAMP can act directly and independently to increase cyclin D2 expression. The levels of p27Kip1 protein were not increased in response to estradiol or FSH. In contrast, when ovulatory doses of human CG (LH) were administered to hormonally primed H rats to stimulate luteinization, cyclin D2 mRNA and protein were rapidly decreased and undetectable within 4 h, specifically in granulosa cells of large follicles. Also in response to LH, the expression of the cell cycle inhibitor p27Kip1 was induced between 12 and 24 h (p21Cip1 was induced within 4 h) and remained elevated specifically in luteal tissue. A critical role for cyclin D2 in the hormone-dependent phase of follicular growth is illustrated by the ovarian follicles of cyclin D2-/- mice, which do not undergo rapid growth in response to hormones, but do express markers of FSH/LH action, cell cycle exit, and terminal differentiation. Collectively, these data indicate that FSH and estradiol regulate granulosa cell proliferation during the development of preovulatory follicles by increasing levels of cyclin D2 relative to p27Kip1 and that LH terminates follicular growth by down-regulating cyclin D2 concurrent with up-regulation of p27Kip1 and p21Cip1.


Assuntos
Proteínas de Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ciclinas/fisiologia , Células da Granulosa/citologia , Hormônios/farmacologia , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Supressoras de Tumor , Animais , Ciclina D2 , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Ciclinas/genética , Inibidores Enzimáticos , Estradiol/farmacologia , Feminino , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Folículo Ovariano/fisiologia , Ovário/química , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
19.
Can J Public Health ; 88(5): 320-4, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9401166

RESUMO

PURPOSE: We sought to test the effectiveness and application of a system for prioritizing corneal disease patients for corneal transplantation. METHODS: All patients wait-listed for corneal transplantation in British Columbia in April 1995 were followed for 10 months to determine whether they received surgery and to assess the application of recently introduced priority criteria. RESULTS: The factors that determined whether a patient had surgery were as follows: having vision in one eye only, being female, having progressive disease, and experiencing pain. The surgeon involved was also a factor. Overall, the priority system did not adequately predict who had surgery and who did not have surgery. CONCLUSIONS: The priority system needs to be re-structured to reduce the contribution of months waited and to more adequately take into account patient need. Furthermore, its application by individual surgeons needs to be strengthened.


Assuntos
Transplante de Córnea/normas , Listas de Espera , Canadá , Ética Médica , Feminino , Alocação de Recursos para a Atenção à Saúde , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Seleção de Pacientes , Estatísticas não Paramétricas
20.
Mol Cell Endocrinol ; 129(1): 101-14, 1997 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-9175634

RESUMO

Activation of cyclic AMP-dependent protein kinases (protein kinase A, PKA) by gonadotropins and cyclic AMP (cAMP) plays an important role in the regulation of testicular functions. A regulatory subunit, RIIbeta, of PKA is transcriptionally induced in rat Sertoli cells in response to treatment with cAMP. The present study addresses regulatory mechanisms leading to increased transcription of the rat RIIbeta gene. We have localized a footprint which overlaps one of the major transcription initiation sites in the basal promoter (-293 to -123). One of the proteins binding this sequence belongs to the NF-1 family of transcription factors. We also observed binding to a basic helix-loop-helix (bHLH) response element. Furthermore, transfection studies of various 5'-deletions of the rat RIIbeta gene in primary cultures of rat Sertoli cells and in peritubular cells revealed the presence of an upstream region (-723 to -395, cAMP-responsive region) inhibiting basal expression from the rat RIIbeta gene only in Sertoli cells. This region was found to enhance cAMP responsiveness in Sertoli cells but not in peritubular cells. Interactions with downstream elements seemed to be important for the function of the cAMP-responsive region. Although some short stretches reveal homology to the cAMP-responsive regions of other slowly cAMP-responding genes, and an AP-1-like element is present, no strong resemblance to any known regulatory element responsive to cAMP is found.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/genética , Células de Sertoli/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Células Cultivadas , AMP Cíclico/metabolismo , Subunidade RIIbeta da Proteína Quinase Dependente de AMP Cíclico , DNA/genética , DNA/metabolismo , Pegada de DNA , Masculino , Sondas de Oligonucleotídeos/genética , Regiões Promotoras Genéticas , Ratos , Deleção de Sequência , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA