Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Aspects Med ; 99: 101307, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39213722

RESUMO

This comprehensive review examines the complex interplay between endocrine disrupting chemicals (EDCs) and the development of testicular germ cell tumors (TGCTs). Despite the high cure rates of TGCTs, challenges in diagnosis and treatment remain, necessitating a deeper understanding of the etiology of the disease. Here, we emphasize current knowledge on the role of EDCs as potential risk factors for TGCTs, focusing on pesticides and perfluorinated and polyfluoroalkyl substances (PFAs/PFCs). Evidence suggests that EDCs disrupt endocrine pathways and induce epigenetic changes that contribute to the development of TGCTs. However, the direct link between EDCs and TGCTs remains elusive and requires further investigation of the molecular mechanisms. We also highlighted the importance of studying nuclear receptors as potential targets for understanding TGCT etiology. In addition, recent evidence implicates PFAs/PFCs in TGCT incidence, highlighting the need for further research into their impact on human health. Overall, this review provides valuable insights into the potential role of EDCs in TGCT development and suggests avenues for future research, while also highlighting how understanding their influence may pave the way for novel therapeutic approaches to improve disease management.


Assuntos
Disruptores Endócrinos , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Humanos , Disruptores Endócrinos/efeitos adversos , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/genética , Neoplasias Testiculares/etiologia , Neoplasias Testiculares/tratamento farmacológico , Masculino , Neoplasias Embrionárias de Células Germinativas/metabolismo , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Embrionárias de Células Germinativas/etiologia , Neoplasias Embrionárias de Células Germinativas/patologia , Praguicidas/efeitos adversos , Animais , Epigênese Genética , Exposição Ambiental/efeitos adversos , Fatores de Risco , Fluorocarbonos
2.
Adv Sci (Weinh) ; 9(17): e2200626, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35435331

RESUMO

Spermatogonial stem cells regenerate and maintain spermatogenesis throughout life, making testis a good model for studying stem cell biology. The effects of chemotherapy on fertility have been well-documented previously. This study investigates how busulfan, an alkylating agent that is often used for chemotherapeutic purposes, affects male fertility. Specifically, the role of the TGR5 pathway is investigated on spermatogonia homeostasis using in vivo, in vitro, and pharmacological methods. In vivo studies are performed using wild-type and Tgr5-deficient mouse models. The results clearly show that Tgr5 deficiency can facilitate restoration of the spermatogonia homeostasis and allow faster resurgence of germ cell lineage after exposure to busulfan. TGR5 modulates the expression of key genes of undifferentiated spermatogonia such as Gfra1 and Fgfr2. At the molecular level, the present data highlight molecular mechanisms underlying the interactions among the TGR5, GLIS2, and TP53 pathways in spermatogonia associated with germ cell apoptosis following busulfan exposure. This study makes a significant contribution to the literature because it shows that TGR5 plays key role on undifferentiated germ cell homeostasis and that modulating the TGR5 signaling pathway could be used as a potential therapeutic tool for fertility disorders.


Assuntos
Bussulfano , Resistencia a Medicamentos Antineoplásicos , Fatores de Transcrição Kruppel-Like , Proteínas do Tecido Nervoso , Receptores Acoplados a Proteínas G , Proteína Supressora de Tumor p53 , Animais , Bussulfano/metabolismo , Bussulfano/farmacologia , Homeostase , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Espermatogônias/metabolismo , Proteína Supressora de Tumor p53/genética
3.
Cells ; 10(9)2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34572036

RESUMO

Prostate cancer is the most common cancer in aging men. Despite recent progress, there are still few effective treatments to cure its aggressive and metastatic stages. A better understanding of the molecular mechanisms driving disease initiation and progression appears essential to support the development of more efficient therapies and improve patient care. To do so, multiple research models, such as cell culture and mouse models, have been developed over the years and have improved our comprehension of the biology of the disease. Recently, a new model has been added with the use of the Drosophila accessory gland. With a high level of conservation of major signaling pathways implicated in human disease, this functional equivalent of the prostate represents a powerful, inexpensive, and rapid in vivo model to study epithelial carcinogenesis. The purpose of this review is to quickly overview the existing prostate cancer models, including their strengths and limitations. In particular, we discuss how the Drosophila accessory gland can be integrated as a convenient complementary model by bringing new understanding in the mechanisms driving prostate epithelial tumorigenesis, from initiation to metastatic formation.


Assuntos
Modelos Animais de Doenças , Drosophila/fisiologia , Genitália Masculina/patologia , Neoplasias da Próstata/patologia , Animais , Humanos , Masculino
4.
Cells ; 10(9)2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34572051

RESUMO

Spermatogenesis is a process within the testis that leads to the production of spermatozoa. It is based on a population of spermatogonial stem cells, which have the capacity to self-renew and to differentiate throughout life to ensure the functions of reproduction are maintained. Male fertility disorders are responsible for half of the cases of infertility in couples worldwide. It is well known that cancer treatments are associated with reversible or irreversible fertility disorders. Busulfan (Bu) is an alkylating agent that significantly inhibits spermatogenesis. The present study relied on a combination of in vivo and in vitro approaches as well as RNAseq analysis to characterize the effects of Bu, in which mouse testes were used as a model. An in silico analysis revealed that many of the Bu-modulated genes are potentially regulated by the SIN3 Transcription Regulator Family Member A (SIN3A) and E2F Transcription Factor (E2F) families of transcription factors. The results demonstrate that the deregulated genes function in processes related to the cell cycle, DNA repair, and cell death mechanisms, including the Tumor Protein 53 (TP53) pathway. This reinforces the role of the TP53 signaling pathway as a major player in Bu effects. In addition, Bu altered the patterns of mRNA accumulation for various genes in undifferentiated spermatogonia. This work provides significant insight into the kinetics and impacts of busulfan, which could pave the way for developing strategies to minimize the impact of chemodrugs and, thus, could lead to germ cell lineage regeneration following anticancer treatments.


Assuntos
Bussulfano/farmacologia , Fertilidade/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Imunossupressores/farmacologia , Testículo/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA-Seq , Testículo/metabolismo
5.
Nat Commun ; 11(1): 2300, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32385236

RESUMO

One of the most important but less understood step of epithelial tumourigenesis occurs when cells acquire the ability to leave their epithelial compartment. This phenomenon, described as basal epithelial cell extrusion (basal extrusion), represents the first step of tumour invasion. However, due to lack of adequate in vivo model, implication of emblematic signalling pathways such as Ras/Mitogen-Activated Protein Kinase (MAPK) and phosphoinositide 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathways, is scarcely described in this phenomenon. We have developed a unique model of basal extrusion in the Drosophila accessory gland. There, we demonstrate that both Ras/MAPK and PI3K/AKT/mTOR pathways are necessary for basal extrusion. Furthermore, as in prostate cancer, we show that these pathways are co-activated. This occurs through set up of Epidermal Growth Factor Receptor (EGFR) and Insulin Receptor (InR) dependent autocrine loops, a phenomenon that, considering human data, could be relevant for prostate cancer.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Drosophila , Glândulas Exócrinas/metabolismo , Masculino , Neoplasias da Próstata/metabolismo , Transdução de Sinais/fisiologia
6.
Cell Mol Life Sci ; 76(24): 4849-4859, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31407019

RESUMO

The farnesoid-X-receptorα (FXRα; NR1H4) is one of the main bile acid (BA) receptors. During the last decades, through the use of pharmalogical approaches and transgenic mouse models, it has been demonstrated that the nuclear receptor FXRα controls numerous physiological functions such as glucose or energy metabolisms. It is also involved in the etiology or the development of several pathologies. Here, we will review the unexpected roles of FXRα on the male reproductive tract. FXRα has been demonstrated to play functions in the regulation of testicular and prostate homeostasis. Even though additional studies are needed to confirm these findings in humans, the reviewed reports open new field of research to better define the effects of bile acid-FXRα signaling pathways on fertility disorders and cancers.


Assuntos
Genitália Masculina/crescimento & desenvolvimento , Próstata/crescimento & desenvolvimento , Receptores Citoplasmáticos e Nucleares/genética , Testículo/crescimento & desenvolvimento , Animais , Ácidos e Sais Biliares/metabolismo , Genitália Masculina/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Próstata/metabolismo , Transdução de Sinais/genética , Testículo/metabolismo , Fatores de Transcrição/genética
7.
J Steroid Biochem Mol Biol ; 194: 105460, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31470110

RESUMO

The bile acid receptor Farnesoid-X-Receptor alpha (FXRα), a member of the nuclear receptor superfamily, is well known for its roles in the enterohepatic tract. In addition, FXRα regulates testicular physiology through the control of both endocrine and exocrine functions. The endocrine function of the Leydig cells is mainly controlled by the hypothalamo-pituitary axis viaLH/chorionic gonadotropin (CG). If FXRα was demonstrated to control the expression of the Lhcgr gene, encoding the LH receptor; the impact of the LH/CG signaling on the Fxrα expression has not been defined so far. Here, we demonstrate that hCG increases the Fxrα gene expression through the protein kinase-A signaling pathway. Fxrα is then involved in a negative feedback of steroid synthesis. These data improve our knowledge of the local control of the testicular steroidogenesis with the identification of the link between the hypothalamo-pituitary axis and the FXRα signaling pathway.


Assuntos
Gonadotropina Coriônica/farmacologia , Receptores Citoplasmáticos e Nucleares/genética , Testículo/efeitos dos fármacos , Animais , Linhagem Celular , Masculino , Camundongos Endogâmicos C57BL , Fosfoproteínas/genética , Progesterona/metabolismo , Receptores do LH/genética , Transdução de Sinais/efeitos dos fármacos , Testículo/metabolismo , Testosterona/sangue , Testosterona/metabolismo
8.
Int J Mol Sci ; 19(11)2018 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-30453651

RESUMO

Structural and functional studies have provided numerous insights over the past years on how members of the nuclear hormone receptor superfamily tightly regulate the expression of drug-metabolizing enzymes and transporters. Besides the role of the farnesoid X receptor (FXR) in the transcriptional control of bile acid transport and metabolism, this review provides an overview on how this metabolic sensor prevents the accumulation of toxic byproducts derived from endogenous metabolites, as well as of exogenous chemicals, in coordination with the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR). Decrypting this network should provide cues to better understand how these metabolic nuclear receptors participate in physiologic and pathologic processes with potential validation as therapeutic targets in human disabilities and cancers.


Assuntos
Ácidos e Sais Biliares/metabolismo , Doença , Saúde , Receptores Citoplasmáticos e Nucleares/metabolismo , Xenobióticos/metabolismo , Animais , Ácidos e Sais Biliares/química , Humanos , Inativação Metabólica
9.
Proc Natl Acad Sci U S A ; 114(15): 3909-3914, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28348226

RESUMO

Lysine Specific Demethylase 1 (LSD1) removes mono- and dimethyl groups from lysine 4 of histone H3 (H3K4) or H3K9, resulting in repressive or activating (respectively) transcriptional histone marks. The mechanisms that control the balance between these two antagonist activities are not understood. We here show that LSD1 and the orphan nuclear receptor estrogen-related receptor α (ERRα) display commonly activated genes. Transcriptional activation by LSD1 and ERRα involves H3K9 demethylation at the transcriptional start site (TSS). Strikingly, ERRα is sufficient to induce LSD1 to demethylate H3K9 in vitro. The relevance of this mechanism is highlighted by functional data. LSD1 and ERRα coregulate several target genes involved in cell migration, including the MMP1 matrix metallo-protease, also activated through H3K9 demethylation at the TSS. Depletion of LSD1 or ERRα reduces the cellular capacity to invade the extracellular matrix, a phenomenon that is rescued by MMP1 reexpression. Altogether our results identify a regulatory network involving a direct switch in the biochemical activities of a histone demethylase, leading to increased cell invasion.


Assuntos
Histona Desmetilases/metabolismo , Histonas/metabolismo , Receptores de Estrogênio/metabolismo , Movimento Celular , Regulação da Expressão Gênica , Células HEK293 , Histona Desmetilases/genética , Humanos , Lisina/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Metilação , Regiões Promotoras Genéticas , Receptores de Estrogênio/genética , Sítio de Iniciação de Transcrição , Receptor ERRalfa Relacionado ao Estrogênio
10.
Mol Aspects Med ; 56: 2-9, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28153453

RESUMO

Primary bile acids are synthetized from cholesterol within the liver and then transformed by the bacteria in the intestine to secondary bile acids. In addition to their involvement in digestion and fat solubilization, bile acids also act as signaling molecules. Several receptors are sensors of bile acids. Among these receptors, this review focuses on the nuclear receptor FXRα and the G-protein-coupled receptor TGR5. This review briefly presents the potential links between bile acids and cancers that are discussed in more details in the other articles of this special issue of Molecular Aspects of Medicine focused on "Bile acids, roles in integrative physiology and pathophysiology".


Assuntos
Ácidos e Sais Biliares/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Acoplados a Proteínas G/genética , Animais , Biotransformação , Colesterol/metabolismo , Microbioma Gastrointestinal/fisiologia , Homeostase/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Fígado/citologia , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Polimorfismo Genético , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
11.
Cell Cycle ; 15(10): 1352-62, 2016 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-27050906

RESUMO

The mammalian target of rapamycin (mTOR) plays essential roles in the regulation of growth-related processes such as protein synthesis, cell sizing and metabolism in both normal and pathological growing conditions. These functions of mTOR are thought to be largely a consequence of its cytoplasmic activity in regulating translation rate, but accumulating data highlight supplementary role(s) for this serine/threonine kinase within the nucleus. Indeed, the nuclear activities of mTOR are currently associated with the control of protein biosynthetic capacity through its ability to regulate the expression of gene products involved in the control of ribosomal biogenesis and proliferation. Using primary murine embryo fibroblasts (MEFs), we observed that cells with overactive mTOR signaling displayed higher abundance for the growth-associated Npm1 protein, in what represents a novel mechanism of Npm1 gene regulation. We show that Npm1 gene expression is dependent on mTOR as demonstrated by treatment of wild-type and Pten inactivated MEFs cultured with rapamycin or by transient transfections of small interfering RNA directed against mTOR. In accordance, the mTOR kinase localizes to the Npm1 promoter gene in vivo and it enhances the activity of a human NPM1-luciferase reporter gene providing an opportunity for direct control. Interestingly, rapamycin did not dislodge mTOR from the Npm1 promoter but rather strongly destabilized the Npm1 transcript by increasing its turnover. Using a prostate-specific Pten-deleted mouse model of cancer, Npm1 mRNA levels were found up-regulated and sensitive to rapamycin. Finally, we also showed that Npm1 is required to promote mTOR-dependent cell proliferation. We therefore proposed a model whereby mTOR is closely involved in the transcriptional and posttranscriptional regulation of Npm1 gene expression with implications in development and diseases including cancer.


Assuntos
Proteínas Nucleares/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Nucleofosmina , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Transplante Heterólogo , Regulação para Cima/efeitos dos fármacos
12.
Brain Pathol ; 26(1): 18-30, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25736961

RESUMO

Radiotherapy is an essential component of glioma standard treatment. Glioblastomas (GBM), however, display an important radioresistance leading to tumor recurrence. To improve patient prognosis, there is a need to radiosensitize GBM cells and to circumvent the mechanisms of resistance caused by interactions between tumor cells and their microenvironment. STAT3 has been identified as a therapeutic target in glioma because of its involvement in mechanisms sustaining tumor escape to both standard treatment and immune control. Here, we studied the role of STAT3 activation on tyrosine 705 (Y705) and serine 727 (S727) in glioma radioresistance. This study explored STAT3 phosphorylation on Y705 (pSTAT3-Y705) and S727 (pSTAT3-S727) in glioma cell lines and in clinical samples. Radiosensitizing effect of STAT3 activation down-modulation by Gö6976 was explored. In a panel of 15 human glioma cell lines, we found that the level of pSTAT3-S727 was correlated to intrinsic radioresistance. Moreover, treating GBM cells with Gö6976 resulted in a highly significant radiosensitization associated to a concomitant pSTAT3-S727 down-modulation only in GBM cell lines that exhibited no or weak pSTAT3-Y705. We report the constitutive activation of STAT3-S727 in all GBM clinical samples. Targeting pSTAT3-S727 mainly in pSTAT3-Y705-negative GBM could be a relevant approach to improve radiation therapy.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Glioblastoma/metabolismo , Glioblastoma/patologia , Fator de Transcrição STAT3/metabolismo , Serina/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Carbazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Humanos , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Radiação , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/efeitos da radiação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Espectrofotometria , Estatísticas não Paramétricas , Fatores de Tempo , Raios X
13.
PLoS One ; 9(5): e96293, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24796332

RESUMO

The chaperone nucleophosmin (NPM1) is over-expressed in the epithelial compartment of prostate tumours compared to adjacent healthy epithelium and may represent one of the key actors that support the neoplastic phenotype of prostate adenocarcinoma cells. Yet, the mechanisms that underlie NPM1 mediated phenotype remain elusive in the prostate. To better understand NPM1 functions in prostate cancer cells, we sought to characterize its impact on prostate cancer cells behaviour and decipher the mechanisms by which it may act. Here we show that NPM1 favors prostate tumour cell migration, invasion and colony forming. Furthermore, knockdown of NPM1 leads to a decrease in the growth of LNCaP-derived tumours grafted in Nude mice in vivo. Such oncogenic-like properties are found in conjunction with a positive regulation of NPM1 on the ERK1/2 (Extracellular signal-Regulated Kinases 1/2) kinase phosphorylation in response to EGF (Epidermal Growth Factor) stimulus, which is critical for prostate cancer progression following the setting of an autonomous production of the growth factor. NPM1 could then be a target to switch off specifically ERK1/2 pathway activation in order to decrease or inhibit cancer cell growth and migration.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas Nucleares/fisiologia , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos Nus , Invasividade Neoplásica/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina
15.
Dalton Trans ; (35): 6894-902, 2009 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-20449127

RESUMO

New weapons to fight cancer are constantly needed. Among chemotherapeutics, anti-cancer metal-drugs have enjoyed a long and successful history since the discovery of the benchmark cisplatin. Advances in metal-drug discovery have motivated chemists to build plethora of complex structures. Among them, a novel area is emerging. This article presents a survey of the metal-N-Heterocyclic Carbenes (Ag(I), Au(I), Pd(II) and Cu(I)-NHCs) as potential anti-cancer agents. Most of the metal-NHCs considered display higher cytotoxicities than the reference metallo-drug cisplatin. Some of them are even selective for particular cell lines. Their mechanisms of action at the cellular level are further discussed, showing that the nature of the metal is of great importance. All these promising results demonstrate that this approach deserves more attention and work.


Assuntos
Antineoplásicos/farmacologia , Compostos Heterocíclicos/farmacologia , Metano/análogos & derivados , Compostos Organometálicos/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Compostos Heterocíclicos/química , Humanos , Metano/química , Metano/farmacologia , Compostos Organometálicos/síntese química , Compostos Organometálicos/química
17.
J Mol Endocrinol ; 39(2): 151-62, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17693613

RESUMO

In the male, androgens promote growth and differentiation of sex reproductive organs through ligand activation of the androgen receptor (AR). Here, we show that androgens are not major actors of the cell cycle arrest associated with the differentiation process, and that the epidermal growth factor (EGF)-mediated signalling interferes with AR activities to regulate androgen response when epithelial cells are differentiated. Higher AR expression and enhanced androgen responsiveness correlate with reduction of phosphorylated ERK1/2 over differentiation. These modifications are associated with recruitment of cells in phase G(0)/G(1), up-regulation of p27(kip1), down-regulation of p21(Cip1) and p53 proteins, and accumulation of hypo-phosphorylated Rb. Exposure to EGF reduces AR expression levels and blocks androgen-dependent transcription in differentiated cells. It also restores p53 and p21(Cip1) levels, Rb hyper-phosphorylation, ERK1/2 activation and promotes cell cycle re-entry as p27(kip1) protein levels are decreased. Treatment with a MEK inhibitor reverses the EGF-mediated AR down-regulation in differentiated cells, thus suggesting the existence of an inverse correlation between EGF and androgen signalling in non-tumoural epithelia. Interestingly, when androgen signalling is set in differentiated cells, dihydrotestosterone exerts an inhibitory effect on ERK activity but paradoxically does not modify EGFR (ErbB1) phosphorylation, indicating that androgens are able to disrupt the EGFR-ERK cascade. Overall, our data demonstrate the existence of a balance between AR and mitogen-activated protein kinase activities that favours either the maintenance of differentiated conditions or the enhancement of cell proliferation capacities.


Assuntos
Receptores ErbB/metabolismo , Receptor Cross-Talk/fisiologia , Receptores Androgênicos/metabolismo , Transdução de Sinais/fisiologia , Androgênios , Animais , Northern Blotting , Western Blotting , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Di-Hidrotestosterona/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptores ErbB/agonistas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Imunoprecipitação , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptor Cross-Talk/efeitos dos fármacos , Receptores Androgênicos/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Ducto Deferente/citologia
18.
J Cell Biol ; 171(4): 651-61, 2005 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-16301331

RESUMO

The proto-oncoprotein Cbl is known to control several signaling processes. It is highly expressed in the testis, and because spermatogenesis is androgen dependent, we investigated the androgen dependency expression of Cbl through its testicular sub-localization and its expression levels in rats that were exposed to the antiandrogen flutamide or were hypophysectomized. We report the androgen dependency of Cbl as it localizes in pachytene spermatocytes during androgen-dependent stages, is down-regulated upon flutamide exposure, and is up-regulated with testosterone in hypophysectomized rats. Coculture experiments showed the key control exerted by the Sertoli cell on Cbl activity. As flutamide induces germ cell apoptosis, we investigate members of the Bcl-2 family upon flutamide exposure. We show that the proapoptotic Bcl-2 family member Bim mirrored Cbl expression through a posttranscriptional process. We also show that in Cbl knockout mouse testes, the imbalance between the high expression of Bim and Smac/Diablo and anti-apoptotic factors such as cellular inhibitor of apoptosis 2 favors a survival process, which makes these mice unresponsive to androgen withdrawal and could explain their hypofertility.


Assuntos
Androgênios/metabolismo , Apoptose , Regulação da Expressão Gênica , Células Germinativas/metabolismo , Proteínas Proto-Oncogênicas c-cbl/fisiologia , Antagonistas de Androgênios/farmacologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Proteínas de Transporte/metabolismo , Técnicas de Cocultura , Flutamida/farmacologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Imunoprecipitação , Proteínas Inibidoras de Apoptose/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Processamento Pós-Transcricional do RNA , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/metabolismo , Espermatócitos/metabolismo , Testículo/metabolismo , Testosterona/metabolismo , Timo/metabolismo , Fatores de Tempo , Transcrição Gênica , Regulação para Cima
19.
J Biol Chem ; 280(26): 24330-8, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15857832

RESUMO

A variety of transcription factors are post-translationally modified by SUMO, a 97-residue ubiquitin-like protein bound covalently to the targeted lysine. Here we describe SUMO modification of the Ets family member ERM at positions 89, 263, 293, and 350. To investigate how SUMO modification affects the function of ERM, Ets-responsive intercellular adhesion molecule 1 (ICAM-1) and E74 reporter plasmids were employed to demonstrate that SUMO modification causes inhibition of ERM-dependent transcription without affecting the subcellular localization, stability, or DNA-binding capacity of the protein. When the adenoviral protein Gam1 or the SUMO protease SENP1 was used to inhibit the SUMO modification pathway, ERM-dependent transcription was de-repressed. These results demonstrate that ERM is subject to SUMO modification and that this post-translational modification causes inhibition of transcription-enhancing activity.


Assuntos
Proteínas de Ligação a DNA/química , Proteína SUMO-1/fisiologia , Fatores de Transcrição/química , Transcrição Gênica , Adenoviridae/genética , Animais , Arginina/química , Sítios de Ligação , Western Blotting , Células COS , Cisteína Endopeptidases , DNA/química , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/genética , Endopeptidases/metabolismo , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Biblioteca Gênica , Genes Reporter , Células HeLa , Humanos , Imunoprecipitação , Molécula 1 de Adesão Intercelular/metabolismo , Lisina/química , Microscopia de Fluorescência , Modelos Genéticos , Plasmídeos/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Coelhos , Proteína SUMO-1/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina/química
20.
J Biol Chem ; 279(15): 14579-86, 2004 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-14668339

RESUMO

Androgens are known to modulate many cellular processes such as cell growth and survival by binding to the androgen receptor (AR) and activating the transcription of target genes. Recent data suggested that AR can also mediate non-transcriptional actions outside the nucleus in addition to its ligand-inducible transcription factor function. Here, we describe a transcription-independent activation of the phosphatidylinositol 3-OH kinase (PI3-K) signaling pathway by androgens. Using non-transformed androgen-sensitive epithelial cells, we show that androgens enhance the PI3-K activity by promoting accumulation of phosphoinositide-3-P phospholipids in vitro. This activation is found in conjunction with an increased time-dependent phosphorylation of the downstream kinase AKT/protein kinase B on both Ser(473) and Thr(308) residues. Hormone-stimulated phosphorylation of AKT requires AR since incubation with the anti-androgen bicalutamide completely abolishes the androgen-stimulated AKT phosphorylation. Accordingly, we show that androgens increase AKT phosphorylation level in prostatic carcinoma PC3 cells only once they have been transfected with AR. Downstream, androgens enhance phosphorylation of transcription factor FKHR (Forkhead in rhabdomyosarcoma)-L1 and proapoptotic Bad protein and promote cell survival as they can counteract an apoptotic process. We also report that non-genomic effects of androgens are based on direct interaction between AR and the p85alpha regulatory subunit of class I(A) PI3-K. Together, these novel findings point out an important and physiologically relevant link between androgens and the PI3-K/AKT signaling pathway in governing cell survival.


Assuntos
Células Epiteliais/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Receptores Androgênicos/fisiologia , Anilidas/farmacologia , Animais , Apoptose , Western Blotting , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Células Epiteliais/metabolismo , Escherichia coli/metabolismo , Glutationa Transferase/metabolismo , Humanos , Ligantes , Camundongos , Modelos Genéticos , Nitrilas , Fosforilação , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Serina/química , Transdução de Sinais , Treonina/química , Fatores de Tempo , Compostos de Tosil , Transcrição Gênica , Transfecção , Proteína de Morte Celular Associada a bcl
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA