Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 35(39): 5191-201, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-26996671

RESUMO

Progestins have long been used clinically for the treatment of endometrial cancers; however, the response rates to progestin therapy vary and the molecular mechanisms behind progestin insensitivity are poorly understood. We hypothesized that in PTEN-mutated endometrial cancers, hyperactive Akt signaling downregulates progesterone receptor B (PRB) transcriptional activity, leading to overall impaired progestin responses. We report that inhibition of Akt with the Akt inhibitor, MK-2206 (MK), in conjunction with progestin (R5020) treatment, is sufficient to upregulate a subset of PRB target genes in Ishikawa cells stably expressing PRB (PRB-Ishikawa). Through gene ontology analysis of Akt-regulated PRB target genes, angiogenesis was found to be the principle process regulated by Akt-PRB. To further interrogate the mechanism by which Akt modulates PRB transcriptional activity, ChIP-Mass spectrometry was performed to identify potential cofactors that differentially interact with PRB in the presence of R5020 and MK+R5020. 14-3-3σ was identified as a protein enriched in the MK+R5020 data set, and it was demonstrated that 14-3-3σ is required for the upregulation in PRB target gene expression following inhibition of Akt. To determine the ramifications of MK+R5020 treatment on angiogenesis, in vitro assays were performed and combinatorial MK+R5020 treatment significantly decreased endothelial cell invasion and tube formation more than MK or R5020 treatment alone. Furthermore, we found that combinatorial MK-2206+progesterone treatments decreased angiogenesis and proliferation in the Pten(d/d) conditional mouse model of endometrial cancer. Taken together, these findings suggest that a combinatorial therapeutic approach utilizing Akt inhibitors with progestins may improve the efficacy of progestin therapy for the treatment of endometrial cancer.


Assuntos
Neoplasias do Endométrio/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Progesterona/genética , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Humanos , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Progestinas/administração & dosagem , Promegestona/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores
2.
Cell Death Differ ; 23(1): 169-81, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26184908

RESUMO

Successful embryo implantation requires functional luminal epithelia to establish uterine receptivity and blastocyst-uterine adhesion. During the configuration of uterine receptivity from prereceptive phase, the luminal epithelium undergoes dynamic membrane reorganization and depolarization. This timely regulated epithelial membrane maturation and precisely maintained epithelial integrity are critical for embryo implantation in both humans and mice. However, it remained largely unexplored with respect to potential signaling cascades governing this functional epithelial transformation prior to implantation. Using multiple genetic and cellular approaches combined with uterine conditional Rac1 deletion mouse model, we demonstrated herein that Rac1, a small GTPase, is spatiotemporally expressed in the periimplantation uterus, and uterine depletion of Rac1 induces premature decrease of epithelial apical-basal polarity and defective junction remodeling, leading to disrupted uterine receptivity and implantation failure. Further investigations identified Pak1-ERM as a downstream signaling cascade upon Rac1 activation in the luminal epithelium necessary for uterine receptivity. In addition, we also demonstrated that Rac1 via P38 MAPK signaling ensures timely epithelial apoptotic death at postimplantation. Besides uncovering a potentially important molecule machinery governing uterine luminal integrity for embryo implantation, our finding has high clinical relevance, because Rac1 is essential for normal endometrial functions in women.


Assuntos
Proteínas de Ligação a DNA/genética , Implantação do Embrião/genética , Neuropeptídeos/genética , Fatores de Transcrição/genética , Quinases Ativadas por p21/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas rac1 de Ligação ao GTP/genética , Animais , Blastocisto/metabolismo , Proteínas de Ligação a DNA/biossíntese , Implantação do Embrião/fisiologia , Endométrio/crescimento & desenvolvimento , Endométrio/metabolismo , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Humanos , Camundongos , Neuropeptídeos/biossíntese , Transdução de Sinais/genética , Fatores de Transcrição/biossíntese , Útero/metabolismo , Útero/fisiologia , Quinases Ativadas por p21/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas rac1 de Ligação ao GTP/biossíntese
3.
Oncogene ; 34(19): 2471-82, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24998851

RESUMO

Type II endometrial carcinomas (ECs) are estrogen independent, poorly differentiated tumors that behave in an aggressive manner. As TP53 mutation and CDH1 inactivation occur in 80% of human endometrial type II carcinomas, we hypothesized that mouse uteri lacking both Trp53 and Cdh1 would exhibit a phenotype indicative of neoplastic transformation. Mice with conditional ablation of Cdh1 and Trp53 (Cdh1(d/d)Trp53(d/d)) clearly demonstrate architectural features characteristic of type II ECs, including focal areas of papillary differentiation, protruding cytoplasm into the lumen (hobnailing) and severe nuclear atypia at 6 months of age. Further, Cdh1(d/d)Trp53(d/d) tumors in 12-month-old mice were highly aggressive, and metastasized to nearby and distant organs within the peritoneal cavity, such as abdominal lymph nodes, mesentery and peri-intestinal adipose tissues, demonstrating that tumorigenesis in this model proceeds through the universally recognized morphological intermediates associated with type II endometrial neoplasia. We also observed abundant cell proliferation and complex angiogenesis in the uteri of Cdh1(d/d)Trp53(d/d) mice. Our microarray analysis found that most of the genes differentially regulated in the uteri of Cdh1(d/d)Trp53(d/d) mice were involved in inflammatory responses. CD163 and Arg1, markers for tumor-associated macrophages, were also detected and increased in the uteri of Cdh1(d/d)Trp53(d/d) mice, suggesting that an inflammatory tumor microenvironment with immune cell recruitment is augmenting tumor development in Cdh1(d/d)Trp53(d/d) uteri. Further, inflammatory mediators secreted from CDH1-negative, TP53 mutant endometrial cancer cells induced normal macrophages to express inflammatory-related genes through activation of nuclear factor-κB signaling. These results indicate that absence of CDH1 and TP53 in endometrial cells initiates chronic inflammation, promotes tumor microenvironment development following the recruitment of macrophages and promotes aggressive ECs.


Assuntos
Proteínas Cdh1/genética , Neoplasias do Endométrio/genética , Inflamação/genética , Macrófagos/imunologia , Proteína Supressora de Tumor p53/genética , Animais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Arginase/genética , Linhagem Celular , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias do Endométrio/patologia , Feminino , Humanos , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Neovascularização Patológica/genética , Receptores de Superfície Celular/genética , Microambiente Tumoral/imunologia , Útero/citologia , Útero/patologia
4.
Oncogene ; 29(26): 3770-80, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20418913

RESUMO

Ablation of Mig-6 in the murine uterus leads to the development of endometrial hyperplasia and estrogen-induced endometrial cancer. An additional endometrial cancer mouse model is generated by the ablation of phosphatase and tensin homolog deleted from chromosome 10 (Pten) (either as heterozygotes or by conditional uterine ablation). To determine the interplay between Mig-6 and the PTEN/phosphoinositide 3-kinase signaling pathway during endometrial tumorigenesis, we generated mice with Mig-6 and Pten conditionally ablated in progesterone receptor-positive cells (PR(cre/+)Mig-6(f/f)Pten(f/f); Mig-6(d/d)Pten(d/d)). The ablation of both Mig-6 and Pten dramatically accelerated the development of endometrial cancer compared with the single ablation of either gene. The epithelium of Mig-6(d/d)Pten(d/d) mice showed a significant decrease in the number of apoptotic cells compared with Pten(d/d) mice. The expression of the estrogen-induced apoptotic inhibitors Birc1 was significantly increased in Mig-6(d/d)Pten(d/d) mice. We identified extracellular signal-regulated kinase 2 (ERK2) as an MIG-6 interacting protein by coimmunoprecipitation and demonstrated that the level of ERK2 phosphorylation was increased upon Mig-6 ablation either singly or in combination with Pten ablation. These results suggest that Mig-6 exerts a tumor-suppressor function in endometrial cancer by promoting epithelial cell apoptosis through the downregulation of the estrogen-induced apoptosis inhibitors Birc1 and the inhibition of ERK2 phosphorylation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias do Endométrio/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Progressão da Doença , Neoplasias do Endométrio/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Receptores de Progesterona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Supressoras de Tumor/metabolismo , Útero/metabolismo
5.
Neuroscience ; 166(1): 94-106, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20025939

RESUMO

The recent molecular cloning of membrane receptors for progesterone (mPRs) has tremendous implications for understanding the multiple actions of the hormone in the nervous system. The three isoforms which have been cloned from several species, mPRalpha, mPRbeta and mPRgamma, have seven-transmembrane domains, are G protein-coupled and may thus account for the rapid modulation of many intracellular signaling cascades by progesterone. However, in order to elucidate the precise functions of mPRs within the nervous system it is first necessary to determine their expression patterns and also to develop new pharmacological and molecular tools. The aim of the present study was to profile mPR expression in the mouse spinal cord, where progesterone has been shown to exert pleiotropic actions on neurons and glial cells, and where the hormone can also be locally synthesized. Our results show a wide distribution of mPRalpha, which is expressed in most neurons, astrocytes, oligodendrocytes, and also in a large proportion of NG2(+) progenitor cells. This mPR isoform is thus likely to play a major role in the neuroprotective and promyelinating effects of progesterone. On the contrary, mPRbeta showed a more restricted distribution, and was mainly present in ventral horn motoneurons and in neurites, consistent with an important role in neuronal transmission and plasticity. Interestingly, mPRbeta was not present in glial cells. These observations suggest that the two mPR isoforms mediate distinct and specific functions of progesterone in the spinal cord. A significant observation was their very stable expression, which was similar in both sexes and not influenced by the presence or absence of the classical progesterone receptors. Although mPRgamma mRNA could be detected in spinal cord tissue by reverse transcriptase-polymerase chain reaction (RT-PCR), in situ hybridization analysis did not allow us to verify and to map its presence, probably due to its relatively low expression. The present study is the first precise map of the regional and cellular distribution of mPR expression in the nervous system, a prior requirement for in vivo molecular and pharmacological strategies aimed to elucidate their precise functions. It thus represents a first important step towards a new understanding of progesterone actions in the nervous system within a precise neuroanatomical context.


Assuntos
Membrana Celular/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Progesterona/metabolismo , Receptores de Progesterona/genética , Medula Espinal/metabolismo , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Membrana Celular/genética , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neuroglia/citologia , Neurônios/citologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Reação em Cadeia da Polimerase , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Receptores de Progesterona/metabolismo , Medula Espinal/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo
6.
Oncogene ; 28(1): 31-40, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18806829

RESUMO

Endometrioid adenocarcinoma is the most frequent form of endometrial cancer, usually developing in pre- and peri-menopausal women. beta-catenin abnormalities are common in endometrioid type endometrial carcinomas with squamous differentiation. To investigate the role of beta-catenin (Ctnnb1) in uterine development and tumorigenesis, mice were generated which expressed a dominant stabilized beta-catenin or had beta-catenin conditionally ablated in the uterus by crossing the PR(Cre) mouse with the Ctnnb1(f(ex3)/+) mouse or Ctnnb1(f/f) mouse, respectively. Both of the beta-catenin mutant mice have fertility defects and the ability of the uterus to undergo a hormonally induced decidual reaction was lost. Expression of the dominant stabilized beta-catenin, PR(cre/+)Ctnnb1(f(ex3)/+), resulted in endometrial glandular hyperplasia, whereas ablation of beta-catenin, PR(cre/+)Ctnnb1(f/f), induced squamous cell metaplasia in the murine uterus. Therefore, we have demonstrated that correct regulation of beta-catenin is important for uterine function as well as in the regulation of endometrial epithelial differentiation.


Assuntos
Transformação Celular Neoplásica/genética , Hiperplasia Endometrial/genética , Endométrio/crescimento & desenvolvimento , beta Catenina/fisiologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Hiperplasia Endometrial/patologia , Endométrio/patologia , Feminino , Infertilidade Feminina/genética , Camundongos , Camundongos Mutantes , beta Catenina/genética
7.
Oncogene ; 26(54): 7526-34, 2007 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-17546050

RESUMO

Branching morphogenesis within the peripubertal mouse mammary gland is directed by progesterone (P). A role for the homeobox-containing transcription factor, Msx2, during branching morphogenesis is suggested from its ontogenic expression profile and hormonal regulation. Herein, we define the spatio-temporal control of Msx2 expression, the regulation of its expression by P and its direct role in ductal branching morphogenesis. P induces Msx2 in the presence of estrogen (E) both in vitro and in vivo while absence of the P receptor (PR) decreased Msx2 expression. Stable transfection of PR into mouse mammary epithelial cells increased the endogenous expression of Msx2 and their ability to undergo branching morphogenesis in vitro. Furthermore, normal mammary cells stably-transfected with Msx2 demonstrated increased branching morphogenesis in vitro while transgenic mice expressing Msx2 in their mammary glands demonstrated enhanced lateral branching during early development. The action of P on branching morphogenesis appears to involve Bmp2/4. Together, these data demonstrate that P, acting through PR-A and the Bmp2/4 pathway, induces Msx2 to enhance ductal branching in the mammary glands.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Glândulas Mamárias Animais/fisiologia , Morfogênese/fisiologia , Progesterona/farmacologia , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Morfogênese/efeitos dos fármacos , Ovariectomia , Reação em Cadeia da Polimerase , RNA/genética , RNA/isolamento & purificação , Receptores de Progesterona/fisiologia , Transdução de Sinais
8.
Artigo em Inglês | MEDLINE | ID: mdl-18540568

RESUMO

The importance of the progesterone receptor (PR) in transducing the progestin signal is firmly established in female reproductive and mammary gland biology; however, the coregulators preferentially recruited by PR in these systems have yet to be comprehensively investigated. Using an innovative genetic approach, which ablates gene function specifically in murine cell-lineages that express PR, steroid receptor coactivator 2 (SRC-2, also known as TIF-2 or GRIP-1) was shown to exert potent coregulator properties in progestin-dependent responses in the uterus and mammary gland. Uterine cells positive for PR (but devoid of SRC-2) led to an early block in embryo implantation, a phenotype not shared by knockouts for SRC-1 or SRC-3. In the case of the mammary gland, progestin-dependent branching morphogenesis and alveologenesis failed to occur in the absence of SRC-2, thereby establishing a critical coactivator role for SRC-2 in cellular proliferative programs initiated by progestins in this tissue. Importantly, the recent detection of SRC-2 in both human endometrium and breast suggests that this coregulator may provide a new clinical target for the future management of female reproductive health and/or breast cancer.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Coativador 2 de Receptor Nuclear/fisiologia , Progestinas/fisiologia , Útero/crescimento & desenvolvimento , Animais , Feminino , Humanos , Morfogênese
9.
J Steroid Biochem Mol Biol ; 102(1-5): 41-50, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17067792

RESUMO

The ovarian steroid hormone progesterone is a major regulator of uterine function. The actions of this hormone is mediated through its cognate receptor, the progesterone receptor, Pgr. Ablation of the Pgr has shown that this receptor is critical for all female reproductive functions including the ability of the uterus to support and maintain the development of the implanting mouse embryo. High density DNA microarray analysis has identified direct and indirect targets of Pgr action. One of the targets of Pgr action is a member of the Hedgehog morphogen Indian Hedgehog, Ihh. Ihh and members of the Hh signaling cascade show a coordinate expression pattern in the mouse uterus during the preimplantation period of pregnancy. The expression of Ihh and its receptor Patched-1, Ptc1, as well as, down stream targets of Ihh-Ptch1 signaling, such as the orphan nuclear receptor COUP-TF II show that this morphogen pathway mediates communication between the uterine epithelial and stromal compartments. The members of the Ihh signaling axis may function to coordinate the proliferation, vascularization and differentiation of the uterine stroma during pregnancy. This analysis demonstrates that progesterone regulates uterine function in the mouse by coordinating the signals from the uterine epithelium to stroma in the preimplantation mouse uterus.


Assuntos
Receptores de Progesterona/metabolismo , Útero/fisiologia , Animais , Feminino , Proteínas Hedgehog/metabolismo , Humanos , Transdução de Sinais , Útero/citologia
10.
J Endocrinol ; 189(3): 473-84, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731779

RESUMO

Progesterone (P4) and its cognate receptor, the progesterone receptor (PGR), have important roles in the establishment and maintenance of pregnancy in the murine uterus. In previous studies, using high-density DNA microarray analysis, we identified a subset of genes whose expression is repressed by chronic P4-PGR activation in the uterus. The Clca3 gene is one of the genes whose expression is the most significantly downregulated by P4 and PGR. In the present study, we performed real-time RT-PCR and in situ hybridization to investigate the regulation of Clca3 by P4 and determine the pattern of expression of Clca3 in the uterus during early pregnancy. This analysis shows that Clca3 mRNA transcripts were detected in the luminal and glandular epithelium of the pseudopregnant uterus at day 0.5 and that the expression of Clca3 was not detected after day 3.5. P4 represses Clca3 mRNA synthesis in the luminal epithelial and glandular epithelial cells of the uterus in ovariectomized wild-type mice, but not in Pgr knockout (PRKO) mice. Conversely, estrogen (E2) induces Clca3 expression in the luminal epithelium and glandular epithelium, and this induction was repressed by P4 in the murine uterus. Analysis of the promoter region of Clca3 by in silico and transient transfection analysis in HEC-1A cells identified the regulation of Clca3 by estrogen receptor-alpha (ESR1) within the first 528 bp of 5'-flanking region of the Clca3 gene. Our studies identified Clca3 as a novel downregulated gene of PGR that is a direct target of E2 regulation.


Assuntos
Canais de Cloreto/genética , Regulação da Expressão Gênica , Progesterona/metabolismo , Regiões Promotoras Genéticas , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animais , Linhagem Celular , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Hibridização In Situ/métodos , Camundongos , Camundongos Knockout , Ovariectomia , Gravidez , Progesterona/genética , Pseudogravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção/métodos
11.
Psychopharmacology (Berl) ; 186(3): 312-22, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16538472

RESUMO

RATIONALE: Aging is associated with reduced secretion of, and down-regulation of receptors for, progesterone (P); yet, P's effects when administered to younger and older animals have not been systematically investigated. Some of P's antianxiety effects may be due to its conversion to 3alpha-hydroxy-5alpha-pregnan-20-one (3alpha,5alpha-THP) and its subsequent actions as a positive modulator at GABAA receptor complexes (GBRs). OBJECTIVES: We investigated whether P administration can decrease anxiety behavior of progestin receptor (PR) knockout (PRKO) or wild-type control mice. METHODS: P (10 mg/kg) or vehicle (propylene glycol) were administered subcutaneously to intact, female or male wild-type or PRKO mice that were either 9-12 or 18-24 months of age. Behavior in tasks that assess spontaneous activity (activity monitor and roto-rod), free exploration of a novel environment (open field, elevated plus maze, and elevated zero maze), and conflict behavior (mirror chamber, dark-light transition, and punished drinking) were examined 1 h after injection. RESULTS: P significantly decreased anxiety behavior of both PRKO and wild-type mice. P did not alter motor behavior but increased central entries in the open field, time in the open quadrants of the elevated zero maze, time in the mirrored chamber, time in the light compartment of the dark-light transition, and punished drinking in young and old mice. P-administered mice had higher levels of hippocampal 3alpha,5alpha-THP and GABA-stimulated chloride flux than did vehicle-administered PRKO or wild-type mice. CONCLUSIONS: The effects of P to decrease anxiety behavior of younger and older mice do not require classic PRs and may involve actions of 3alpha,5alpha-THP at GBRs.


Assuntos
Envelhecimento/fisiologia , Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Progesterona/farmacologia , Receptores de Progesterona/fisiologia , Animais , Ansiedade/fisiopatologia , Conflito Psicológico , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Destreza Motora/efeitos dos fármacos , Pregnanolona/metabolismo , Receptores de Progesterona/deficiência , Receptores de Progesterona/genética , Ácido gama-Aminobutírico/farmacologia
12.
Psychopharmacology (Berl) ; 185(4): 423-32, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16544163

RESUMO

RATIONALE: Progesterone (P) and its 5alpha-reduced metabolite, 3alpha-hydroxy-5alpha-pregnan-20-one (3alpha,5alpha-THP), facilitate sexual behavior of rodents via agonist-like actions at intracellular progestin receptors (PRs) and membrane GABA(A)/benzodiazepine receptor complexes (GBRs), respectively. OBJECTIVE: Given that ovarian secretion of progestins declines with aging, whether or not senescent mice are responsive to progestins was of interest. METHODS: Homozygous PR knockout (PRKO) or wild-type mice that were between 10-12 (mid-aged) or 20-24 (aged) months of age were administered P or 3alpha,5alpha-THP, and the effect on lordosis were examined. Effects of a progestin-priming regimen that enhances PR-mediated (experiment 1) or more rapid, PR-independent effects of progestins (experiments 2 and 3) on sexual behavior were examined. Levels of P, 3alpha,5alpha-THP, and muscimol binding were examined in tissues from aged mice (experiment 4). RESULTS: Wild-type, but not PRKO, mice were responsive when primed with 17beta-estradiol (E(2); 0.5 microg) and administered P (500 microg, subcutaneously). Mid-aged wild-type mice demonstrated greater increases in lordosis 6 h later compared to their pre-P, baseline test than did aged wild-type mice (experiment 1). Lordosis of younger and older wild-type, but not PRKO, mice was significantly increased within 5 min of intravenous (IV) administration of P (100 ng), compared with E(2)-priming alone (experiment 2). However, wild-type and PRKO mice demonstrated significant increases in lordosis 5 min after IV administration of 3alpha,5alpha-THP, an effect which was more pronounced in mid-aged than in aged animals (100 ng-experiment 3). In tissues from aged wild-type and PRKO mice, levels of P, 3alpha,5alpha-THP, and muscimol binding were increased by P administration (experiment 4). PR binding was lower in the cortex of PRKO than that of wild-type mice. CONCLUSIONS: Mid-aged and aged PRKO and wild-type mice demonstrated rapid P or 3alpha,5alpha-THP-facilitated lordosis that may be, in part, independent of activity at PRs.


Assuntos
Envelhecimento/fisiologia , Desoxicorticosterona/análogos & derivados , Progesterona/farmacologia , Receptores de Progesterona/fisiologia , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Desoxicorticosterona/administração & dosagem , Desoxicorticosterona/farmacologia , Estradiol/farmacologia , Feminino , Agonistas GABAérgicos/metabolismo , Injeções Intravenosas , Injeções Subcutâneas , Camundongos , Camundongos Knockout , Muscimol/metabolismo , Postura , Progesterona/administração & dosagem , Progesterona/metabolismo , Radioimunoensaio , Receptores de GABA/efeitos dos fármacos , Receptores de Progesterona/genética
13.
J Endocrinol ; 180(2): 287-95, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14765981

RESUMO

Progesterone, via its nuclear receptor, is mandatory not only for the induction and specification of mammary gland ductal side-branching and lobuloalveologenesis but also for carcinogen-induced mammary tumorigenesis. Notwithstanding these recent advances, a more comprehensive molecular explanation of progesterone-induced mammary morphogenesis is contingent upon the identification and characterization of mammary molecular targets that are responsive to the progesterone signal. Toward this goal, we report that calcitonin, a 32 amino acid peptide hormone involved in calcium homeostasis, is exclusively expressed in, and secreted from, luminal epithelial cells within the mammary gland of the pregnant mouse, and, importantly, its expression is progesterone-dependent. Conversely, the calcitonin receptor is present during all stages of post-natal mammary development examined, is localized to the myoepithelial cell lineage, and is not regulated by progesterone. Because calcitonin induction spatiotemporally correlates with increases in progesterone-induced mammary gland proliferation and structural remodeling, we posit that calcitonin - through its receptor - may be involved in one or both of these progesterone-dependent processes.


Assuntos
Calcitonina/metabolismo , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Prenhez/metabolismo , Progesterona/metabolismo , Animais , Calcitonina/análise , Calcitonina/genética , Divisão Celular , Estrogênios/farmacologia , Feminino , Imuno-Histoquímica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Gravidez , Progesterona/genética , Progesterona/farmacologia , RNA/análise , Receptores da Calcitonina/análise , Receptores da Calcitonina/metabolismo
14.
J Clin Invest ; 108(4): 611-8, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11518735

RESUMO

Clinical studies of hormone replacement therapy to prevent cardiovascular diseases have heightened interest in the cardiovascular effects of progestins. However, the role of the progesterone receptor (PR) in vascular biology has not been studied in vivo. We studied ovariectomized female PR knockout (PRKO) mice and their wild-type (WT) littermates using the mouse carotid artery injury model. Placebo-treated PRKO mice showed significantly greater vascular medial hypertrophy and vascular smooth muscle cell (VSMC) proliferation in response to vascular injury than did WT mice. Progesterone had no significant effect in the PRKO mice, but worsened the response to injury in WT mice. VSMCs cultured from PRKO mouse aortae were markedly hyperproliferative, and their growth was not affected by progesterone. In contrast to the in vivo findings, progesterone inhibited proliferation of WT-derived VSMCs. Furthermore, reintroduction of PR into PRKO-derived VSMCs using adenoviral methods restored progesterone-mediated inhibition of proliferation to these cells. This effect was reversed by the PR antagonist, RU 486. Thus, the effects of PR and progesterone differ markedly between cultured VSMCs and intact blood vessels. These data demonstrate a direct role for the PR in regulating the response to vascular injury and VSMC proliferation.


Assuntos
Lesões das Artérias Carótidas , Receptores de Progesterona/fisiologia , Animais , Artéria Carótida Primitiva/patologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Feminino , Antagonistas de Hormônios/farmacologia , Hiperplasia , Camundongos , Camundongos Knockout , Mifepristona/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Ovariectomia , Progesterona/antagonistas & inibidores , Progesterona/farmacologia , Receptores de Progesterona/deficiência , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/genética , Proteínas Recombinantes de Fusão/fisiologia , Transfecção
15.
Mol Cell Endocrinol ; 179(1-2): 97-103, 2001 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-11420134

RESUMO

Progesterone plays a central coordinate role in diverse reproductive events associated with establishment and maintenance of pregnancy. In humans and other vertebrates, the biological activities of progesterone are mediated by two proteins, A (PR-A) and B (PR-B) that arise from the same gene and function as progesterone activated transcription factors that exhibit different transcription regulatory activities in vitro. Mice lacking both PR isoforms (PRKO mice) exhibit pleiotropic reproductive abnormalities. To address the physiological role of the individual isoforms, we have selectively ablated PR-A expression in mice (PRAKO). We have demonstrated that PR-B mediates a subset of the reproductive functions of P. Ablation of PR-A does not affect responses of the mammary gland or thymus to P but results in severe abnormalities in ovarian and uterine function. Analysis of urine function of PRAKP mice reveals an unexpected P-dependent proliferative activity of PR-B in the epithelium and provides evidence that the tissue-specific reproductive effects of this isoform are due to specificity of target gene transactivation rather than differences in tissue-specific expression relative to PR-A. Taken together, our data indicate that PR-A and PR-B act in vivo as two functionally distinct transcription factors.


Assuntos
Mama/fisiologia , Ovário/fisiologia , Receptores de Progesterona/fisiologia , Útero/fisiologia , Animais , Mama/citologia , Epitélio/crescimento & desenvolvimento , Epitélio/fisiologia , Feminino , Camundongos , Camundongos Knockout , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Receptores de Progesterona/química , Útero/transplante
16.
Cell Death Differ ; 8(2): 192-200, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11313721

RESUMO

In males, androgens are essential in maintaining the integrity of the prostate. Androgen-ablation induces apoptosis of the prostatic epithelium. In females, ovariectomy induces apoptosis in uterine epithelium while progesterone inhibits this process. The objective of this study was to determine whether androgen and progesterone inhibit apoptosis, respectively, in mouse prostatic and uterine epithelia via steroid receptors in the epithelium or in the stroma. To address this question, prostatic tissue recombinants were prepared with rat urogenital sinus mesenchyme plus bladder epithelium from wild-type or testicular feminization mutant (Tfm) mice. Thus, prostatic tissue was generated having androgen receptor (AR) in both epithelium and stroma or in the stroma only. Castration of hosts induced apoptosis in the AR-negative Tfm prostatic epithelium with an epithelial apoptotic index virtually identical to prostatic tissue recombinants containing wild-type epithelium. Moreover, this castration-induced prostatic epithelial apoptosis was blocked by testosterone and dihydrotestosterone in both wild-type and Tfm prostatic tissue recombinants. Likewise, uterine tissue recombinants were prepared in which epithelium and/or stroma was devoid of progesterone receptor (PR) by using uterine epithelium and stroma of wild-type and PR knockout mice. Progesterone inhibited uterine epithelial apoptosis only in tissue recombinants prepared with PR-positive stroma. The PR status of the epithelium did not affect epithelial apoptotic index. Therefore, the apoptosis in prostatic and uterine epithelia is regulated by androgen and progesterone via stromal AR and PR, respectively. In both cases, epithelial AR or PR is not required for hormonal regulation of epithelial apoptosis in prostatic and uterine epithelium.


Assuntos
Apoptose/fisiologia , Comunicação Parácrina/fisiologia , Próstata/metabolismo , Esteroides/metabolismo , Útero/metabolismo , Androgênios/metabolismo , Androgênios/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Masculino , Camundongos , Camundongos Nus , Progesterona/metabolismo , Progesterona/farmacologia , Próstata/citologia , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/metabolismo , Esteroides/farmacologia , Útero/citologia
17.
Steroids ; 65(10-11): 571-7, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11108861

RESUMO

Progesterone (P) is a key regulator of female reproductive activity. The effects of P are mediated by two progesterone receptor (PR) proteins, termed A and B, that arise from a single gene and act as ligand-activated transcription factors to regulate the expression of reproductive target genes. Null mutation of the PR gene in mice (PRKO) leads to pleiotropic reproductive abnormalities. This paper will review the reproductive functions of PRs delineated using the PRKO mouse. Further, we will summarize the structure and functional properties of PRs and discuss how functional differences between the PR-A and PR-B isoforms are likely to impact on the overall physiological role of the receptor in reproductive systems.


Assuntos
Receptores de Progesterona/fisiologia , Reprodução/efeitos dos fármacos , Animais , Feminino , Humanos , Camundongos , Camundongos Knockout , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Receptores de Progesterona/química , Receptores de Progesterona/genética
18.
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
19.
Proc Natl Acad Sci U S A ; 97(23): 12816-21, 2000 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-11070092

RESUMO

The ventromedial hypothalamus (VMH) plays a central role in the regulation of the female reproductive behavior lordosis, a behavior dependent upon the sequential activation of receptors for the ovarian steroid hormones estradiol (E) and progesterone (P). These receptors function as transcription factors to alter the expression of target genes. To discover behaviorally relevant genes targeted by E and P in the VMH, we used the differential display PCR to identify messenger RNAs that are differentially expressed in the hypothalamus of ovariectomized (ovx) rats treated with E alone compared with ovariectomized rats treated with E and P. We show here that one interesting mRNA within the hypothalamus that is repressed by P after E priming encodes the protein 25-Dx, the rat homolog of the human membrane-associated P-binding protein Hpr6.6. Neurons in the brain containing the highest levels of 25-Dx are located in several nuclei of the basal forebrain, including the VMH. 25-Dx expression is also higher in the hypothalamus of female P receptor "knockout" mice than in their wild-type littermates. These findings suggest a mechanism in which the activation of nuclear P receptor represses expression of a membrane P receptor, 25-Dx, during lordosis facilitation.


Assuntos
Hipotálamo/metabolismo , Proteínas de Membrana/metabolismo , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Comportamento Sexual Animal , Animais , Sequência de Bases , Encéfalo/metabolismo , Encéfalo/patologia , Membrana Celular/metabolismo , DNA Complementar , Estradiol/administração & dosagem , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Expressão Gênica , Hipotálamo/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/metabolismo , Sistemas Neurossecretores/metabolismo , Postura/fisiologia , Progesterona/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/genética , Reprodução/fisiologia , Caracteres Sexuais
20.
Science ; 289(5485): 1751-4, 2000 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-10976068

RESUMO

Progesterone regulates reproductive function through two intracellular receptors, progesterone receptor-A (PR-A) and progesterone receptor-B (PR-B), that arise from a single gene and function as transcriptional regulators of progesterone-responsive genes. Although in vitro studies show that PR isoforms can display different transcriptional regulatory activities, their physiological significance is unknown. By selective ablation of PR-A in mice, we show that the PR-B isoform modulates a subset of reproductive functions of progesterone by regulation of a subset of progesterone-responsive target genes. Thus, PR-A and PR-B are functionally distinct mediators of progesterone action in vivo and should provide suitable targets for generation of tissue-selective progestins.


Assuntos
Implantação do Embrião , Progesterona/fisiologia , Receptores de Progesterona/fisiologia , Reprodução , Útero/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Cruzamentos Genéticos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica , Masculino , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Knockout , Ovariectomia , Ovulação , Progesterona/farmacologia , Isoformas de Proteínas , Receptores de Progesterona/genética , Útero/citologia , Útero/efeitos dos fármacos , Útero/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...