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











Base de dados
Intervalo de ano de publicação
1.
Res Rep Urol ; 13: 705-713, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34549035

RESUMO

INTRODUCTION: Although the ability of androgens to promote prostate cancer development has been known for decades, the molecular mechanisms of androgen receptor (AR) signaling in the tumorigenesis remain unclear. Enhancer RNAs (eRNAs) transcribed from strong enhancers, or super-enhancers (SEs), have recently emerged as a novel class of regulatory non-coding RNAs (ncRNAs) that facilitate transcription, including that of androgen target genes, through chromatin looping to position enhancers proximate to the promoters. The aim of this study was to assess androgen-dependent transcription in prostate tumors of eRNAs (designated as KLK3eRNAs) from the SE of the KLK3 gene encoding the prostate-specific antigen (PSA) protein, a clinical marker of prostate carcinogenesis. MATERIALS AND METHODS: The androgen-induced KLK3eRNAs were identified in the LNCaP human prostate cancer cell line. The expressions of these KLK3eRNAs together with KLK3 and AR mRNA transcripts were assessed by qRT-PCR in prostate tumor samples from five prostate cancer patients. RESULTS: Androgen-induced KLK3eRNAs have been identified in the LNCaP cells, and their expression was further analyzed in tumors of prostate cancer patients. Transcripts of the tested KLK3eRNAs have been detected in all clinical samples, but their expression patterns differed between individual tumor specimens. We found a statistically significant correlation between the levels of the KLK3 and AR mRNAs with those of the previously reported KLK3eRNAs, while such correlation was not observed for novel KLK3eRNAs described in our recent report. CONCLUSION: Presented data suggest that prostate tumor development may associate with epigenetic reorganization in the KLK3 genomic regulatory elements reflected by changes of the KLK3eRNA expression. Our findings support a potential of eRNAs profiling to be used as diagnostic marker.

2.
Biosci Biotechnol Biochem ; 85(10): 2121-2130, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34297060

RESUMO

Androgen induces the binding of its receptor (AR) to androgen-responsive elements (AREs), while genome-wide studies showed that most androgen-induced AR binding sites on chromatin were unrelated to AREs. Enhancer RNAs (eRNAs), a class of noncoding RNAs (ncRNAs), are transcribed from superenhancers (SEs) and trigger the formation of large ribonucleoprotein condensates of transcription factors. By in silico search, an SE is found to be located on the locus of KLK3 that encodes prostate specific antigen. On the KLK3 SE, androgen-induced expression of ncRNAs was detected and designated as KLK3eRNAs in LNCaP cells, and androgen-induced association of AR and FOXA1 on the KLK3eRNA coding regions was detected. Such androgen-induced association of an AR mutant lacking DNA binding activity on the KLK3eRNA coding regions was undetectable on an exogenous ARE. Thus, the present findings suggest a molecular basis of androgen-induced association of AR with chromatin on ARE-unrelated sequences.


Assuntos
Receptores Androgênicos , Androgênios , Fator 3-alfa Nuclear de Hepatócito , Humanos , Masculino , Antígeno Prostático Específico , Neoplasias da Próstata
3.
Res Rep Urol ; 9: 225-232, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29264358

RESUMO

INTRODUCTION: Therapeutically induced androgen deficiency (AD) is a standard treatment for patients with prostate cancer, but it is often associated with various adverse effects (AEs) that may lead to discontinuation. Some AEs may depend on the patient's health condition, while others may be due to complications of the drug delivery method. Degarelix is a gonadotropin-releasing hormone (GnRH) antagonist widely used for the treatment of androgen-dependent prostate cancer. This study aimed to ascertain the following: 1) the compatibility of degarelix treatment with diabetes and 2) any specific causal associations of degarelix injections with increased blood clotting and antithrombotic therapy requirements. PATIENTS AND METHODS: The medical records of 162 patients with prostate cancer who had undergone degarelix treatment were retrospectively examined. The association of a medical history of diabetes and anticoagulant co-treatment with degarelix treatment discontinuation was analyzed statistically. RESULTS: Rapid and significant decreases in prostate-specific antigen (PSA) levels during the course of degarelix treatment were detected for patients with prostate cancer regardless of clinical state. During the 27 months of treatment, 68 subjects (48%) ceased degarelix treatment, owing to several reasons, mainly financial issues. Among these subjects, 19 had diabetes, while 35 were treated with antithrombotics. Extensive statistical analysis indicated that there were no causal associations between degarelix treatment discontinuation and preexisting diabetes or antithrombotic therapy. CONCLUSION: Our study suggests that preexisting diabetes and antithrombotic therapy were not significant factors for the discontinuation of degarelix treatment in patients with prostate cancer.

4.
Bonekey Rep ; 4: 715, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26157578

RESUMO

Coordinated post-translational modifications (PTMs) of nucleosomal histones emerge as a key mechanism of gene regulation by defining chromatin configuration. Patterns of histone modifications vary in different cells and constitute core elements of cell-specific epigenomes. Recently, in addition to canonical histone proteins produced during the S phase of cell cycle, several non-canonical histone variants have been identified and shown to express in a DNA replication-independent manner. These histone variants generate diversity in nucleosomal structures and add further complexity to mechanisms of epigenetic regulation. Cell-specific functions of histone variants remain to be determined. Several recent studies reported an association between some point mutations in the non-canonical histone H3.3 and particular types of brain and bone tumors. This suggests a possibility of differential physiological effects of histone variants in different cells and tissues, including bone. In this review, we outline the roles of histone variants and their PTMs in the epigenetic regulation of chromatin structure and discuss possible mechanisms of biological effects of the non-canonical histone mutations found in bone tumors on tumorigenesis in differentiating bone stem cells.

5.
Physiol Rev ; 93(2): 481-523, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23589826

RESUMO

During the last decade, our view on the skeleton as a mere solid physical support structure has been transformed, as bone emerged as a dynamic, constantly remodeling tissue with systemic regulatory functions including those of an endocrine organ. Reflecting this remarkable functional complexity, distinct classes of humoral and intracellular regulatory factors have been shown to control vital processes in the bone. Among these regulators, nuclear receptors (NRs) play fundamental roles in bone development, growth, and maintenance. NRs are DNA-binding transcription factors that act as intracellular transducers of the respective ligand signaling pathways through modulation of expression of specific sets of cognate target genes. Aberrant NR signaling caused by receptor or ligand deficiency may profoundly affect bone health and compromise skeletal functions. Ligand dependency of NR action underlies a major strategy of therapeutic intervention to correct aberrant NR signaling, and significant efforts have been made to design novel synthetic NR ligands with enhanced beneficial properties and reduced potential negative side effects. As an example, estrogen deficiency causes bone loss and leads to development of osteoporosis, the most prevalent skeletal disorder in postmenopausal women. Since administration of natural estrogens for the treatment of osteoporosis often associates with undesirable side effects, several synthetic estrogen receptor ligands have been developed with higher therapeutic efficacy and specificity. This review presents current progress in our understanding of the roles of various nuclear receptor-mediated signaling pathways in bone physiology and disease, and in development of advanced NR ligands for treatment of common skeletal disorders.


Assuntos
Desenvolvimento Ósseo/genética , Osso e Ossos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Animais , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/citologia , Osso e Ossos/fisiopatologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Ligantes
6.
Annu Rev Physiol ; 75: 201-24, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23157556

RESUMO

Androgens play pivotal roles in the regulation of male development and physiological processes, particularly in the male reproductive system. Most biological effects of androgens are mediated by the action of nuclear androgen receptor (AR). AR acts as a master regulator of downstream androgen-dependent signaling pathway networks. This ligand-dependent transcriptional factor modulates gene expression through the recruitment of various coregulator complexes, the induction of chromatin reorganization, and epigenetic histone modifications at target genomic loci. Dysregulation of androgen/AR signaling perturbs normal reproductive development and accounts for a wide range of pathological conditions such as androgen-insensitive syndrome, prostate cancer, and spinal bulbar muscular atrophy. In this review we summarize recent advances in understanding of the epigenetic mechanisms of AR action as well as newly recognized aspects of AR-mediated androgen signaling in both men and women. In addition, we offer a perspective on the use of animal genetic model systems aimed at eventually developing novel therapeutic AR ligands.


Assuntos
Androgênios/fisiologia , Receptores Androgênicos/fisiologia , Transdução de Sinais/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Doenças dos Genitais Femininos/fisiopatologia , Doenças dos Genitais Masculinos/fisiopatologia , Humanos , Masculino , Camundongos , Saúde Reprodutiva
7.
Expert Opin Ther Targets ; 15(10): 1143-5, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21736533

RESUMO

The treatment of osteoporosis has been a critical issue in today's medical situation. Various therapeutic agents and strategies have been investigated and applied, and have proven successful in the treatment of osteoporosis. However, some concerns still remain, such as the adverse effects of such treatments. From this point of view, a search for novel therapeutic targets, such as Fas signaling, remains important.


Assuntos
Osso e Ossos/fisiologia , Proteína Ligante Fas/metabolismo , Osteoporose/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Receptor fas/metabolismo , Osso e Ossos/metabolismo , Feminino , Humanos , Masculino
8.
Epigenomics ; 2(6): 765-74, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22122081

RESUMO

Methylation and demethylation of cytosine residues in the genomic DNA play key roles in a wide range of fundamental biological processes such as differentiation and development, genome stability, imprinting, X chromosome inactivation, carcinogenesis and aging. DNA methylation is considered to be a stable modification associated with the epigenetic silencing of genomic loci and maintained through cellular division. Recent studies however, suggest that DNA methylation and demethylation are considerably more dynamic than previously thought and may be involved in repression and derepression of gene activity during the lifespan of a cell. This article is focused on epigenetic mechanisms in the hormonal regulation of the cytochrome p450 27B1 or CYP27B1 gene activity that involve reversible epigenetic modifications to chromatin and DNA methylation profiles.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Cromatina/genética , Metilação de DNA , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/fisiologia , Modelos Biológicos , Hormônio Paratireóideo/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Cromatina/metabolismo , Ilhas de CpG/genética , Regulação da Expressão Gênica/genética , Humanos , Regiões Promotoras Genéticas/genética , Receptores de Calcitriol/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
9.
Mol Endocrinol ; 24(5): 877-85, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19910454

RESUMO

The osteoprotective action of estrogen in women has drawn considerable attention because estrogen deficiency-induced osteoporosis became one of the most widely spread diseases in developed countries. In men, the significance of estrogen action for bone health maintenance is also apparent from the osteoporotic phenotype seen in male patients with genetically impaired estrogen signaling. Severe bone loss and high bone turnover, including typical osteofeatures seen in postmenopausal women, can also be recapitulated in rodents after ovariectomy. However, the expected osteoporotic phenotype is not observed in female mice deficient in estrogen receptor (ER)-alpha or -beta or both, even though the degenerative defects are clearly seen in other estrogen target tissues together with up-regulated levels of circulating testosterone. It has also been reported that estrogens may attenuate bone remodeling by cell autonomous suppressive effects on osteoblastogenesis and osteoclastogenesis. Hence, the effects of estrogens in bone appear to be complex, and the molecular role of bone estrogen receptors in osteoprotective estrogen action remains unclear. Instead, it has been proposed that estrogens indirectly control bone remodeling. For example, the enhanced production of cytokines under estrogen deficiency induces bone resorption through stimulation of osteoclastogenesis. However, the osteoporotic phenotype without systemic defects has been recapitulated in female (but not in male) mice by osteoclast-specific ablation of the ERalpha, proving that bone cells represent direct targets for estrogen action. An aberrant accumulation of mature osteoclasts in these female mutants indicates that in females, the inhibitory action of estrogens on bone resorption is mediated by the osteoclastic ERalpha through the shortened lifespan of osteoclasts.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Osteoclastos/metabolismo , Animais , Remodelação Óssea/genética , Remodelação Óssea/fisiologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Humanos , Modelos Biológicos
10.
Ann N Y Acad Sci ; 1173 Suppl 1: E31-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19751412

RESUMO

Estrogens play a key role in regulation of bone mass and strength by controlling activity of bone-forming osteoblasts and bone-resorbing osteoclasts. Cellular effects of estrogens are mediated predominantly by the action of estrogen receptor alpha (ERalpha). In earlier studies, ablation of the ERalpha gene in mice did not result in osteoporotic phenotypes due to systemic endocrine disturbance and compensatory effects of elevated levels of testosterone. Despite the relatively well-established effects in osteoblasts, little is known about the direct action of estrogen in osteoclasts. Development in the last decade of more sophisticated genetic manipulation approaches opened new possibilities to explore cell-specific roles of nuclear receptors in bone tissue. Recently, we have generated osteoclast-specific ERalpha gene knockout mice and shown that in vivo estrogens directly regulate the life span of mature osteoclasts by inducing the expression of pro-apoptotic Fas ligand (FasL). Inhibitory effects of estrogens on osteoclast function were further studied in vitro. We observed sufficiently detectable ERalpha expression in osteoclasts differentiating from primary bone marrow cells or RAW264 cells, although levels of ERalpha were decreasing during progression of the differentiation into mature osteoclasts. Treatment with estrogens led to reduction in expression of osteoclast-specific genes controlling bone resorption activity. However, estrogens did not affect the size of multinucleated osteoclasts or number of nuclei in a mature osteoclast. In conclusion, in osteoclasts, estrogens function to inhibit bone resorption activity and vitality rather than differentiation.


Assuntos
Osso e Ossos/anatomia & histologia , Estrogênios/fisiologia , Regulação da Expressão Gênica , Osteoclastos/fisiologia , Idoso , Animais , Densidade Óssea , Osso e Ossos/fisiologia , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Feminino , Fraturas Ósseas/epidemiologia , Fraturas Ósseas/prevenção & controle , Hormônios Esteroides Gonadais/fisiologia , Hematopoese , Humanos , Estilo de Vida , Locomoção , Longevidade/genética , Camundongos , Camundongos Knockout , Ovariectomia , Pós-Menopausa
11.
Nat Rev Rheumatol ; 5(8): 442-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19581903

RESUMO

Osteoblasts and adipocytes differentiate from a common pluripotent precursor, the mesenchymal stem cell (MSC). Studies have identified numerous transcription factors, and multiple extracellular and intracellular signaling pathways that regulate the closely linked processes of adipogenesis and osteoblastogenesis. Interestingly, inducers of differentiation along one lineage often inhibit differentiation along the other; for example, the transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma) is a prime inducer of adipogenesis that inhibits osteoblastogenesis. The latest research has shown that inducers of osteoblastogenesis (such as bone morphogenetic protein 2 and Wnt ligands) use different mechanisms to suppress the transactivation function of PPARgamma during osteoblastogenesis from MSCs. Signaling via the canonical Wnt-beta-catenin pathway inhibits PPARgamma mRNA expression, whereas signaling via the noncanonical Wnt pathway results in activation of a histone methyltransferase SETDB1 that represses PPARgamma transactivation through histone H3K9 methylation of target genes. This article summarizes Wnt and PPARgamma signaling in MSCs and the crosstalk between these pathways, and speculates on future clinical application of this knowledge as the basis of novel approaches for regeneration therapy.


Assuntos
Adipócitos/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/fisiologia , Osteoblastos/citologia , PPAR gama/metabolismo , Proteínas Wnt/metabolismo , Animais , Osso e Ossos/metabolismo , Histona-Lisina N-Metiltransferase , Humanos , Proteínas Metiltransferases/metabolismo , Transdução de Sinais
12.
Expert Opin Ther Targets ; 13(5): 593-603, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19397478

RESUMO

Osteoblasts and adipocytes differentiate from a common precursor, the pluripotent mesenchymal stem cell (MSC) found in bone marrow (BMSC) and adipose tissue (AD-MSC). Numerous transcription factors and multiple extracellular and intracellular signals regulating adipogenesis and osteoblastogenesis have been identified and analyzed. Significantly, inducers of differentiation towards one lineage may inhibit cell differentiation into an alternative lineage. For example, the canonical Wnt/beta-catenin pathway induces osteoblastogenesis and inhibits adipogenesis, whereas the peroxisome proliferator activated receptor-gamma (PPAR-gamma) is a prime inducer of adipogenesis and, as shown in recent studies, inhibits osteoblastogenesis. We have identified two signaling pathways that switch the cell fate decision from adipocytes to osteoblasts by suppressing the transactivation function of PPAR-gamma. In the first pathway, the TNF-alpha- or IL-1-induced TAK1/TAB1/NIK signaling cascade attenuates PPAR-gamma-mediated adipogenesis by inhibiting the binding of PPAR-gamma to the DNA response element. The second is the noncanonical Wnt pathway through the CaMKII-TAK1/TAB2-NLK (nemo-like kinase) signaling cascade. Specifically, Wnt-5a-induced phosphorylation of NLK triggers formation of a complex with the histone methyltransferase SETDB1 (SET domain, bifurcated 1) that represses PPAR-gamma transactivation through histone H3-K9 methylation at the target genes. Thus, two signaling cascades promote osteoblastic differentiation from MSC through two distinct modes of PPAR-gamma transrepression.


Assuntos
Adipogenia/fisiologia , Sistemas de Liberação de Medicamentos , Regeneração Tecidual Guiada , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , PPAR gama/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Citocinas/farmacologia , Citocinas/fisiologia , Epigênese Genética , Regulação da Expressão Gênica/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Neoplasias/induzido quimicamente , Osteoblastos/citologia , Osteogênese/efeitos dos fármacos , PPAR gama/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia
13.
Proc Natl Acad Sci U S A ; 106(10): 3818-22, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19237573

RESUMO

Spinal and bulbar muscular atrophy (SBMA) is a neurodegenerative disorder caused by a polyglutamine repeat (polyQ) expansion within the human androgen receptor (AR). Unlike other neurodegenerative diseases caused by abnormal polyQ expansion, the onset of SBMA depends on androgen binding to mutant human polyQ-AR proteins. This is also observed in Drosophila eyes ectopically expressing the polyQ-AR mutants. We have genetically screened mediators of androgen-induced neurodegeneration caused by polyQ-AR mutants in Drosophila eyes. We identified Rbf (Retinoblastoma-family protein), the Drosophila homologue of human Rb (Retinoblastoma protein), as a neuroprotective factor. Androgen-dependent association of Rbf or Rb with AR was remarkably potentiated by aberrant polyQ expansion. Such potentiated Rb association appeared to attenuate recruitment of histone deacetyltransferase 1 (HDAC1), a corepressor of E2F function. Either overexpression of Rbf or E2F deficiency in fly eyes reduced the neurotoxicity of the polyQ-AR mutants. Induction of E2F function by polyQ-AR-bound androgen was suppressed by Rb in human neuroblastoma cells. We conclude that abnormal expansion of polyQ may potentiate innate androgen-dependent association of AR with Rb. This appears to lead to androgen-dependent onset of SBMA through aberrant E2F transactivation caused by suppressed histone deacetylation.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fatores de Transcrição E2F/metabolismo , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patologia , Degeneração Neural/patologia , Peptídeos/metabolismo , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Animais , Proteínas de Drosophila/genética , Fatores de Transcrição E2F/genética , Humanos , Ligantes , Proteínas Mutantes/metabolismo , Degeneração Neural/metabolismo , Ligação Proteica , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional
14.
Mol Cell Endocrinol ; 310(1-2): 3-10, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-18789372

RESUMO

Bone tissue protects and supports soft organs and maintains calcium homeostasis. Steroid sex hormones and fat-soluble vitamins play a pivotal role in regulation of bone homeostasis, turnover and remodeling. These molecules act as ligands of nuclear receptors, through which they control gene expression in bone cells, namely bone-forming osteoblasts, bone-resorptive osteoclasts and osteocytes. Significant advances in our understanding of nuclear receptor physiology have been achieved due to development of novel genetic manipulation approaches and generation of experimental animal models in which nuclear receptor genes were mutated in specific cell types. In this review, we summarized some aspects of recent progress in studies on molecular mechanisms of cell-specific action of nuclear hormone receptors in bone tissue.


Assuntos
Osso e Ossos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Estrogênios/deficiência , Humanos , Especificidade de Órgãos , Osteoclastos/patologia , Osteoporose/etiologia , Osteoporose/patologia
15.
Mol Cell Biol ; 29(4): 1017-34, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19075001

RESUMO

Ligand-bound nuclear receptors (NR) activate transcription of the target genes. This activation is coupled with histone modifications and chromatin remodeling through the function of various coregulators. However, the nature of the dependence of a NR coregulator action on the presence of the chromatin environment at the target genes is unclear. To address this issue, we have developed a modified position effect variegation experimental model system that includes an androgen-dependent reporter transgene inserted into either a pericentric heterochromatin region or a euchromatic region of Drosophila chromosome. Human androgen receptor (AR) and its constitutively active truncation mutant (AR AF-1) were transcriptionally functional in both chromosomal regions. Predictably, the level of AR-induced transactivation was lower in the pericentric heterochromatin. In genetic screening for AR AF-1 coregulators, Drosophila CREB binding protein (dCBP) was found to corepress AR transactivation at the pericentric region whereas it led to coactivation in the euchromatic area. Mutations of Sir2 acetylation sites or deletion of the CBP acetyltransferase domain abrogated dCBP corepressive action for AR at heterochromatic areas in vivo. Such a CBP corepressor function for AR was observed in the transcriptionally silent promoter of an AR target gene in cultured mammalian cells. Thus, our findings suggest that the action of NR coregulators may depend on the state of chromatin at the target loci.


Assuntos
Proteína de Ligação a CREB/metabolismo , Efeitos da Posição Cromossômica/genética , Drosophila melanogaster/genética , Heterocromatina/metabolismo , Modelos Genéticos , Receptores Androgênicos/genética , Ativação Transcricional/genética , Acetilação , Sequência de Aminoácidos , Animais , Proteína de Ligação a CREB/química , Domínio Catalítico , Linhagem Celular Tumoral , Sequência Conservada , Proteínas de Drosophila/química , Eucromatina/metabolismo , Histona Desacetilases/química , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Proteínas Repressoras/metabolismo , Sirtuínas/química
16.
Biosci Biotechnol Biochem ; 72(9): 2255-61, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18776683

RESUMO

Abnormal polyglutamine (polyQ) expansion in the N-terminal domain of the human androgen receptor (hAR) is known to cause spinobulbar muscular atrophy (SBMA), a hereditary human neurodegenerative disorder. To explore the molecular mechanisms of neurodegeneration in SBMA, we genetically screened modulators of neurodegeneration in a Drosophila SBMA experimental model system. We identified hoip as an accelerator of polyQ-induced neurodegeneration. We found that hoip forms a complex with 18s rRNA together nop56 and nop5 proteins, whose human homologs are known to form a snoRNP complex involved in ribosomal RNA processing. Significantly, the levels of mutant polyQ-hAR were up-regulated in a mutant line overexpressing hoip. Consistently, severe neurodegeneration phenotype (rough eye) was also observed in both nop56 and nop5 overexpression mutant lines. These findings suggest that the process of neurodegeneration induced by abnormal polyQ expansion in the hAR may be regulated by the activity of snoRNP complex.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Genes de Insetos , Degeneração Neural/metabolismo , Peptídeos/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Animais Geneticamente Modificados , Células Cultivadas , Modelos Animais de Doenças , Drosophila/citologia , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Olho/metabolismo , Olho/ultraestrutura , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/metabolismo , Mutação , Degeneração Neural/genética , Proteínas de Ligação a RNA/metabolismo , Receptores Androgênicos , Transfecção
17.
Genes Cells ; 13(7): 723-30, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18498351

RESUMO

Numerous independent clinical and experimental studies indicate that estrogens confer a protective effect against development of intestinal tumors, however the molecular mechanisms involved remain unclear. Physiological effects of estrogens are predominantly mediated by the action of nuclear estrogen receptors (ERs). A multifunctional protein adenomatous polyposis coli (APC) is a tumor suppressor and thought to act as a gatekeeper in colon tumorigenesis, as loss of function APC mutations trigger the development of colorectal cancer. Here we report that APC physically associates with ERa in the ligand-dependent manner. We have shown in the endogenous setting that the ligand-activated ERa recruits APC to the promoters in ER target genes and that increased levels of ER-dependent recruitment of APC enhances the ER transactivation through stimulation of histone acetylation. Found in majority of human colon tumors APC truncation mutants lost the ability to interact with ER. Thus, here we present the first evidence of a functional interaction between APC and ER that may be accounted for a tumor protective action of estrogens.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Sequência de Aminoácidos , Estrogênios/fisiologia , Genes APC , Células HCT116 , Humanos , Ligantes , Elementos de Resposta/fisiologia , Deleção de Sequência , Ativação Transcricional/fisiologia
18.
Nature ; 446(7135): 562-6, 2007 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-17392787

RESUMO

Fat-soluble ligands, including sex steroid hormones and environmental toxins, activate ligand-dependent DNA-sequence-specific transcriptional factors that transduce signals through target-gene-selective transcriptional regulation. However, the mechanisms of cellular perception of fat-soluble ligand signals through other target-selective systems remain unclear. The ubiquitin-proteasome system regulates selective protein degradation, in which the E3 ubiquitin ligases determine target specificity. Here we characterize a fat-soluble ligand-dependent ubiquitin ligase complex in human cell lines, in which dioxin receptor (AhR) is integrated as a component of a novel cullin 4B ubiquitin ligase complex, CUL4B(AhR). Complex assembly and ubiquitin ligase activity of CUL4B(AhR) in vitro and in vivo are dependent on the AhR ligand. In the CUL4B(AhR) complex, ligand-activated AhR acts as a substrate-specific adaptor component that targets sex steroid receptors for degradation. Thus, our findings uncover a function for AhR as an atypical component of the ubiquitin ligase complex and demonstrate a non-genomic signalling pathway in which fat-soluble ligands regulate target-protein-selective degradation through a ubiquitin ligase complex.


Assuntos
Proteínas Culina/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Linhagem Celular , Proteínas Culina/química , Proteínas Culina/genética , Receptor alfa de Estrogênio/metabolismo , Humanos , Ligantes , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ligação Proteica , Receptores de Hidrocarboneto Arílico/genética , Especificidade por Substrato , Ativação Transcricional
19.
Genes Cells ; 9(10): 983-92, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15461668

RESUMO

Phosphorylation of the Ser(118) residue in the N-terminal A/B domain of the human oestrogen receptor alpha (hERalpha) by mitogen-activated protein kinase (MAPK), stimulated via growth factor signalling pathways, is known to potentiate ERalpha ligand-induced transactivation function. Besides MAPK, cyclin dependent kinase 7 (Cdk7) in the TFIIH complex has also been found to potentiate hERalpha transactivation in vitro through Ser(118) phosphorylation. To investigate an impact of Cdk7 on hERalpha transactivation in vivo, we assessed activity of hERalpha in a wild-type and cdk7 inactive mutant Drosophila that ectopically expressed hERalpha in the eye disc. Ectopic expression of the wild-type or mutant receptors, together with a green fluorescent protein (GFP) reporter gene, allowed us to demonstrate that hERalpha expressed in the fly tissues was transcriptionally functional and adequately responded to hERalpha ligands in the patterns similar to those observed in mammalian cells. Replacement of Ser(118) with alanine in hERalpha (S118A mutant) significantly reduced the ligand-induced hERalpha transactivation function. Importantly, while in cdk7 inactive mutant Drosophila the wild-type hERalpha exhibited reduced response to the ligand; levels of transactivation by the hERalpha S118A mutant were not affected in these inactive cdk7 mutant flies. Furthermore, phosphorylation of hERalpha at Ser(118) has been observed in vitro by both human and Drosophila Cdk7. Our findings demonstrate that Cdk7 is involved in regulation of the ligand-induced transactivation function of hERalphain vivo via Ser(118) phosphorylation.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Drosophila melanogaster/genética , Receptor alfa de Estrogênio/metabolismo , Acetiltransferases/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/metabolismo , Receptor alfa de Estrogênio/genética , Genes Reporter , Histona Acetiltransferases , Humanos , Ligantes , Mutação , Fosforilação , Serina/genética , Serina/metabolismo , Quinase Ativadora de Quinase Dependente de Ciclina
20.
J Biol Chem ; 279(39): 40255-8, 2004 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-15304487

RESUMO

Wnt and estrogen signaling represent important regulatory pathways, each controlling a wide range of biological processes. While an increasing number of observations suggest potential convergence between these pathways, no direct evidence of their functional interaction has been reported. Using human colon and breast cancer cells, we found that estrogen receptor (ER) alpha- and beta-catenin precipitated within the same immunocomplexes, reciprocally enhanced the transactivation of cognate reporter genes, and were reciprocally recruited to cognate response elements in the promoters of endogenous target genes. Using transgenic Drosophila that ectopically expressed human ERalpha alone or together with metabolically stable beta-catenin/Armadillo mutants, we demonstrated genetic interaction between these signal transducers in vivo. Thus, we present here the first direct evidence of cross-talk between Wnt and estrogen signaling pathways via functional interaction between beta-catenin and ERalpha.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Estrogênios/metabolismo , Transativadores/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas do Citoesqueleto/genética , Drosophila , Receptor alfa de Estrogênio , Genes Reporter , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Mutação , Testes de Precipitina , Regiões Promotoras Genéticas , Ligação Proteica , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Transativadores/genética , Transcrição Gênica , Ativação Transcricional , Transfecção , Transgenes , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA