Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Biosci Rep ; 37(4)2017 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-28607032

RESUMO

We have investigated and characterized a novel ornithine decarboxylase (ODC) related protein (ODCrp) also annotated as gm853. ODCrp shows 41% amino acid sequence identity with ODC and 38% with ODC antizyme inhibitor 1 (AZIN1). The Odcrp gene is selectively expressed in the epithelium of proximal tubuli of mouse kidney with higher expression in males than in females. Like Odc in mouse kidney, Odcrp is also androgen responsive with androgen receptor (AR)-binding loci within its regulatory region. ODCrp forms homodimers but does not heterodimerize with ODC. Although ODCrp contains 20 amino acid residues known to be necessary for the catalytic activity of ODC, no decarboxylase activity could be found with ornithine, lysine or arginine as substrates. ODCrp does not function as an AZIN, as it neither binds ODC antizyme 1 (OAZ1) nor prevents OAZ-mediated inactivation and degradation of ODC. ODCrp itself is degraded via ubiquination and mutation of Cys363 (corresponding to Cys360 of ODC) appears to destabilize the protein. Evidence for a function of ODCrp was found in ODC assays on lysates from transfected Cos-7 cells where ODCrp repressed the activity of endogenous ODC while Cys363Ala mutated ODCrp increased the enzymatic activity of endogenous ODC.


Assuntos
Regulação da Expressão Gênica/fisiologia , Túbulos Renais Proximais/metabolismo , Proteólise , Ubiquitinação/fisiologia , Animais , Células COS , Chlorocebus aethiops , Masculino , Camundongos , Camundongos Endogâmicos ICR
2.
Mol Cell Endocrinol ; 414: 91-8, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26219822

RESUMO

We have analyzed androgen receptor (AR) chromatin binding sites (ARBs) and androgen-regulated transcriptome in estrogen receptor negative molecular apocrine breast cancer cells. These analyses revealed that 42% of ARBs and 39% androgen-regulated transcripts in MDA-MB453 cells have counterparts in VCaP prostate cancer cells. Pathway analyses showed a similar enrichment of molecular and cellular functions among AR targets in both breast and prostate cancer cells, with cellular growth and proliferation being among the most enriched functions. Silencing of the coregulator SUMO ligase PIAS1 in MDA-MB453 cells influenced AR function in a target-selective fashion. An anti-apoptotic effect of the silencing suggests involvement of the PIAS1 in the regulation of cell death and survival pathways. In sum, apocrine breast cancer and prostate cancer cells share a core AR cistrome and target gene signature linked to cancer cell growth, and PIAS1 plays a similar coregulatory role for AR in both cancer cell types.


Assuntos
Neoplasias da Mama/genética , Cromatina/metabolismo , Perfilação da Expressão Gênica/métodos , Neoplasias da Próstata/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Receptores Androgênicos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sítios de Ligação , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/química
3.
Endocr Rev ; 36(4): 357-84, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26052734

RESUMO

The physiological androgens testosterone and 5α-dihydrotestosterone regulate the development and maintenance of primary and secondary male sexual characteristics through binding to the androgen receptor (AR), a ligand-dependent transcription factor. In addition, a number of nonreproductive tissues of both genders are subject to androgen regulation. AR is also a central target in the treatment of prostate cancer. A large number of studies over the last decade have characterized many regulatory aspects of the AR pathway, such as androgen-dependent transcription programs, AR cistromes, and coregulatory proteins, mostly in cultured cells of prostate cancer origin. Moreover, recent work has revealed the presence of pioneer/licensing factors and chromatin modifications that are important to guide receptor recruitment onto appropriate chromatin loci in cell lines and in tissues under physiological conditions. Despite these advances, current knowledge related to the mechanisms responsible for receptor- and tissue-specific actions of androgens is still relatively limited. Here, we review topics that pertain to these specificity issues at different levels, both in cultured cells and tissues in vivo, with a particular emphasis on the nature of the steroid, the response element sequence, the AR cistromes, pioneer/licensing factors, and coregulatory proteins. We conclude that liganded AR and its DNA-response elements are required but are not sufficient for establishment of tissue-specific transcription programs in vivo, and that AR-selective actions over other steroid receptors rely on relaxed rather than increased stringency of cis-elements on chromatin.


Assuntos
Androgênios/fisiologia , Receptores Androgênicos/fisiologia , Transdução de Sinais/fisiologia , Antagonistas de Androgênios , Células Cultivadas , Cromatina/metabolismo , DNA/metabolismo , Di-Hidrotestosterona/metabolismo , Humanos , Masculino , Especificidade de Órgãos , Progestinas , Neoplasias da Próstata , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores de Glucocorticoides/fisiologia , Elementos de Resposta , Testosterona/fisiologia
4.
Int J Cancer ; 137(10): 2374-83, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26014856

RESUMO

HOXB7 encodes a transcription factor that is overexpressed in a number of cancers and encompasses many oncogenic functions. Previous results have shown it to promote cell proliferation, angiogenesis, epithelial-mesenchymal transition, DNA repair and cell survival. Because of its role in many cancers and tumorigenic processes, HOXB7 has been suggested to be a potential drug target. However, HOXB7 binding sites on chromatin and its targets are poorly known. The aim of our study was to identify HOXB7 binding sites on breast cancer cell chromatin and to delineate direct target genes located nearby these binding sites. We found 1,504 HOXB7 chromatin binding sites in BT-474 breast cancer cell line that overexpresses HOXB7. Seventeen selected binding sites were validated by ChIP-qPCR in several breast cancer cell lines. Furthermore, we analyzed expression of a large number of genes located nearby HOXB7 binding sites and found several new direct targets, such as CTNND2 and SCGB1D2. Identification of HOXB7 chromatin binding sites and target genes is essential to understand better the role of HOXB7 in breast cancer and mechanisms by which it regulates tumorigenic processes.


Assuntos
Neoplasias da Mama/metabolismo , Cromatina/genética , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Sítios de Ligação , Cateninas/metabolismo , Linhagem Celular Tumoral , Cromatina/patologia , Imunoprecipitação da Cromatina/métodos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Secretoglobinas/metabolismo , delta Catenina
5.
Nucleic Acids Res ; 43(2): 848-61, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25552417

RESUMO

Androgen receptor (AR) is a ligand-activated transcription factor that plays a central role in the development and growth of prostate carcinoma. PIAS1 is an AR- and SUMO-interacting protein and a putative transcriptional coregulator overexpressed in prostate cancer. To study the importance of PIAS1 for the androgen-regulated transcriptome of VCaP prostate cancer cells, we silenced its expression by RNAi. Transcriptome analyses revealed that a subset of the AR-regulated genes is significantly influenced, either activated or repressed, by PIAS1 depletion. Interestingly, PIAS1 depletion also exposed a new set of genes to androgen regulation, suggesting that PIAS1 can mask distinct genomic loci from AR access. In keeping with gene expression data, silencing of PIAS1 attenuated VCaP cell proliferation. ChIP-seq analyses showed that PIAS1 interacts with AR at chromatin sites harboring also SUMO2/3 and surrounded by H3K4me2; androgen exposure increased the number of PIAS1-occupying sites, resulting in nearly complete overlap with AR chromatin binding events. PIAS1 interacted also with the pioneer factor FOXA1. Of note, PIAS1 depletion affected AR chromatin occupancy at binding sites enriched for HOXD13 and GATA motifs. Taken together, PIAS1 is a genuine chromatin-bound AR coregulator that functions in a target gene selective fashion to regulate prostate cancer cell growth.


Assuntos
Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , Proteínas Inibidoras de STAT Ativados/metabolismo , Receptores Androgênicos/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Humanos , Masculino , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ubiquitina-Proteína Ligases/metabolismo
6.
Nucleic Acids Res ; 42(7): 4230-40, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24459135

RESUMO

The DNA-binding domains (DBDs) of class I steroid receptors-androgen, glucocorticoid, progesterone and mineralocorticoid receptors-recognize a similar cis-element, an inverted repeat of 5'-AGAACA-3' with a 3-nt spacer. However, these receptors regulate transcription programs that are largely receptor-specific. To address the role of the DBD in and of itself in ensuring specificity of androgen receptor (AR) binding to chromatin in vivo, we used SPARKI knock-in mice whose AR DBD has the second zinc finger replaced by that of the glucocorticoid receptor. Comparison of AR-binding events in epididymides and prostates of wild-type (wt) and SPARKI mice revealed that AR achieves selective chromatin binding through a less stringent sequence requirement for the 3'-hexamer. In particular, a T at position 12 in the second hexamer is dispensable for wt AR but mandatory for SPARKI AR binding, and only a G at position 11 is highly conserved among wt AR-preferred response elements. Genome-wide AR-binding events agree with the respective transcriptome profiles, in that attenuated AR binding in SPARKI mouse epididymis correlates with blunted androgen response in vivo. Collectively, AR-selective actions in vivo rely on relaxed rather than increased stringency of cis-elements on chromatin. These elements are, in turn, poorly recognized by other class I steroid receptors.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Receptores Androgênicos/metabolismo , Elementos de Resposta , Transcrição Gênica , Animais , Elementos Facilitadores Genéticos , Epididimo/metabolismo , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Androgênicos/genética
7.
EMBO J ; 33(4): 312-26, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24451200

RESUMO

Androgen receptor (AR) binds male sex steroids and mediates physiological androgen actions in target tissues. ChIP-seq analyses of AR-binding events in murine prostate, kidney and epididymis show that in vivo AR cistromes and their respective androgen-dependent transcription programs are highly tissue specific mediating distinct biological pathways. This high order of tissue specificity is achieved by the use of exclusive collaborating factors in the three androgen-responsive tissues. We find two novel collaborating factors for AR signaling in vivo--Hnf4α (hepatocyte nuclear factor 4α) in mouse kidney and AP-2α (activating enhancer binding protein 2α) in mouse epididymis--that define tissue-specific AR recruitment. In mouse prostate, FoxA1 serves for the same purpose. FoxA1, Hnf4α and AP-2α motifs are over-represented within unique AR-binding loci, and the cistromes of these factors show substantial overlap with AR-binding events distinct to each tissue type. These licensing or pioneering factors are constitutively bound to chromatin and guide AR to specific genomic loci upon hormone exposure. Collectively, liganded receptor and its DNA-response elements are required but not sufficient for establishment of tissue-specific transcription programs.


Assuntos
Epididimo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 3-alfa Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Rim/metabolismo , Próstata/metabolismo , Receptores Androgênicos/metabolismo , Testosterona/farmacologia , Fator de Transcrição AP-2/fisiologia , Animais , Linhagem Celular , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Epididimo/citologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos ICR , Orquiectomia , Especificidade de Órgãos , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas de Ligação a Tacrolimo/biossíntese , Proteínas de Ligação a Tacrolimo/genética , Testosterona/fisiologia , Transcrição Gênica
8.
Artigo em Inglês | MEDLINE | ID: mdl-23702556

RESUMO

Computational analysis of data produced in deep sequencing (DS) experiments is challenging due to large data volumes and requirements for flexible analysis approaches. Here, we present a mathematical formalism based on set algebra for frequently performed operations in DS data analysis to facilitate translation of biomedical research questions to language amenable for computational analysis. With the help of this formalism, we implemented the Genomic Region Operation Kit (GROK), which supports various DS-related operations such as preprocessing, filtering, file conversion, and sample comparison. GROK provides high-level interfaces for R, Python, Lua, and command line, as well as an extension C++ API. It supports major genomic file formats and allows storing custom genomic regions in efficient data structures such as red-black trees and SQL databases. To demonstrate the utility of GROK, we have characterized the roles of two major transcription factors (TFs) in prostate cancer using data from 10 DS experiments. GROK is freely available with a user guide from >http://csbi.ltdk.helsinki.fi/grok/.


Assuntos
Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Modelos Genéticos , Software , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Cancer Res ; 73(5): 1570-80, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23269278

RESUMO

The forkhead protein FoxA1 has functions other than a pioneer factor, in that its depletion brings about a significant redistribution in the androgen receptor (AR) and glucocorticoid receptor (GR) cistromes. In this study, we found a novel function for FoxA1 in defining the cell-type specificity of AR- and GR-binding events in a distinct fashion, namely, for AR in LNCaP-1F5 cells and for GR in VCaP cells. We also found different, cell-type and receptor-specific compilations of cis-elements enriched adjacent to the AR- and GR-binding sites. The AR pathway is central in prostate cancer biology, but the role of GR is poorly known. We find that AR and GR cistromes and transcription programs exhibit significant overlap, and GR regulates a large number of genes considered to be AR pathway-specific. This raises questions about the role of GR in maintaining the AR pathway under androgen-deprived conditions in castration-resistant prostate cancer patients. However, in the presence of androgen, ligand-occupied GR acts as a partial antiandrogen and attenuates the AR-dependent transcription program. .


Assuntos
Fator 3-alfa Nuclear de Hepatócito/fisiologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Glucocorticoides/metabolismo , Castração , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Genes , Fator 3-alfa Nuclear de Hepatócito/genética , Humanos , Ligantes , Masculino , Especificidade de Órgãos , Neoplasias da Próstata/genética , Ligação Proteica , Receptores Androgênicos/genética , Receptores de Glucocorticoides/genética , Transdução de Sinais/genética
10.
Endocrinology ; 154(2): 698-708, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23270804

RESUMO

Properly functioning adipose tissue is essential for normal insulin sensitivity of the body. When mice are kept on high-fat diet (HFD), adipose tissue expands, adipocytes increase in size and number, and the mice become obese. Many of these changes are mediated by the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ), the activity of which is regulated by multiple posttranslational modifications, including SUMOylation. To address the role of small ubiquitin-like modifier-1 (SUMO-1) in PPARγ function in vivo, particularly in fat cell biology, we subjected Sumo1-knockout mice to HFD. Sumo1-null mice gained less weight and had smaller and fewer adipocytes in their gonadal fat tissue on HFD, but their glucose tolerance was similar to that of wild-type littermates. Adipogenesis was impaired in Sumo1-null cells, and expression of PPARγ target genes was attenuated. In addition, both Sumo1-null cells and Sumo1-null mice responded less efficiently to rosiglitazone, a PPARγ agonist. These findings indicate that SUMO-1 is important also for transcriptional activation by the PPARγ signaling pathway and not only for trans-repressive functions of PPARγ as previously reported.


Assuntos
Adipogenia/fisiologia , Peso Corporal/genética , PPAR gama/fisiologia , Proteína SUMO-1/genética , Tiazolidinedionas/farmacologia , Células 3T3-L1 , Tecido Adiposo Branco/efeitos dos fármacos , Animais , Dieta Hiperlipídica , Feminino , Masculino , Camundongos , Camundongos Knockout , PPAR gama/agonistas , Rosiglitazona , Proteína SUMO-1/fisiologia
11.
Eur J Endocrinol ; 168(1): K9-K18, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23076843

RESUMO

OBJECTIVE: Generalized glucocorticoid resistance is characterized by impaired cortisol signaling, resulting from mutations of the glucocorticoid receptor (GR) gene NR3C1. The objective of our study was to identify the causative mutation in a patient with clinical manifestations compatible with generalized glucocorticoid resistance and to determine the functional consequences of the mutation. The possible occurrence of NR3C1 mutations in a selected group of hypertensive subjects with low plasma renin and aldosterone levels was also explored. PATIENTS: The proband, a male athlete, was diagnosed with hypertension associated with low plasma renin activity and low serum aldosterone concentration at the age of 27 years. Liddle's syndrome was suspected and the patient was treated with amiloride with initial success. Subsequent examinations revealed elevated serum cortisol and ACTH levels, with resistance to suppression with low doses of dexamethasone. After identification of an NR3C1 mutation in the proband, the available family members and 51 nonrelated hypertensive subjects with low plasma renin and aldosterone concentrations were also studied. RESULTS: A two-nucleotide deletion in exon 9α, predicted to cause a frameshift mutation (p.L773VfsX25) in the hormone-binding domain of the GR, was identified in the patient in a heterozygous form. Affected brother and father died of premature coronary heart disease. Functional studies in COS-1 cells showed that this mutation eliminates both ligand-binding and transactivation ability of the receptor. No pathogenic NR3C1 mutations were identified in 51 unrelated hypertensive patients with low plasma renin and aldosterone levels. CONCLUSION: We identified a novel frameshift mutation in NR3C1 as the cause of glucocorticoid resistance. The mutation eliminates the functional activity of the GR, as studied by in vitro experiments. Mutations in NR3C1 do not seem to be common causes for hypertension with low renin and aldosterone levels.


Assuntos
Aldosterona/sangue , Hipertensão/genética , Erros Inatos do Metabolismo/genética , Receptores de Glucocorticoides/genética , Renina/sangue , Adulto , Animais , Sequência de Bases , Células COS , Doença da Artéria Coronariana/genética , Feminino , Mutação da Fase de Leitura , Humanos , Masculino , Receptores de Glucocorticoides/deficiência , Deleção de Sequência
12.
Prostate ; 72(11): 1223-32, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22212979

RESUMO

BACKGROUND: Castration-resistant prostate cancers (CRPCs) overexpress often androgen receptor (AR). Here, we investigated the effect of AR overexpression on the dynamics of AR loading and RNA polymerase II (RNA Pol II) recruitment to chromatin. Acetylation of histone 3 (AcH3) on lysines 9 and 14 (K9 and K14) was also studied. METHODS: We used an LNCaP-based AR overexpression cell line model that includes a control line and two sublines, LNCaP-ARmo and LNCaP-ARhi, which overexpress AR twofold to threefold and fourfold to fivefold, respectively. Cells were exposed to 1 or 100 nM of dihydrotestosterone (DHT). Chromatin immunoprecipitation (ChIP) on the promoters and enhancers of prostate specific antigen (PSA) and transmembrane protease, serine 2 (TMPRSS2) genes was performed. qRT-PCR was used to measure the levels of PSA and TMPRSS2 transcripts. RESULTS: Upon stimulation with 1 nM DHT, AR and RNA Pol II were recruited onto PSA and TMPRSS2 enhancer regions to a greater extent (P < 0.05) in AR-overexpressing cells compared to control cells. The difference in AR loading between the control and AR-overexpressing cells was abolished by a higher DHT concentration. The ratio of AcH3/H3 was increased in AR-overexpressing cells. The induction of transcription of PSA and TMPRSS2 occurred earlier in the AR-overexpressing cells. CONCLUSIONS: Our findings suggest that the levels of AR potentiate the recruitment of the AR, as well as components of the basic transcription machinery, to chromatin and affect the acetylation of histones in the presence of low levels of androgens. These changes result in enhanced gene transcription of AR target genes.


Assuntos
Cromatina/genética , Neoplasias da Próstata/genética , RNA Polimerase II/metabolismo , Receptores Androgênicos/metabolismo , Acetilação/efeitos dos fármacos , Androgênios/farmacologia , Linhagem Celular Tumoral , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Histonas/metabolismo , Humanos , Masculino , Orquiectomia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/cirurgia , Receptores Androgênicos/genética
13.
Endocrinology ; 152(11): 4395-405, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21878517

RESUMO

To enable studies of androgen signaling in different tissues in vivo, we generated an androgen receptor (AR) reporter mouse line by inserting a luciferase gene construct into the murine genome. The construct is driven by four copies of androgen-responsive elements from the mouse sex-limited protein gene (slp-HRE2) and a minimal thymidine kinase promoter. Luciferase activity was readily measurable in a number of murine tissues, including prostate, lung, testis, brain, and skeletal muscle, and testosterone administration elicited a significant increase in reporter gene activity in these tissues. Consumption of isoflavonoid genistein is linked to reduced risk of prostate cancer, but direct effects of genistein on the AR pathway are not well understood. To examine androgen-modulating activity of genistein in vivo, male mice received daily doses of genistein (10 mg/kg) for 5 d. In intact males, genistein was antiandrogenic in testis, prostate, and brain, and it attenuated reporter gene activity by 50-80%. In castrated males, genistein exhibited significant androgen agonistic activity in prostate and brain by increasing reporter gene activity over 2-fold in both tissues. No antiandrogenic action was seen in lung or skeletal muscle of intact males. Gene expression profiling of the murine prostate under the same experimental conditions revealed that genistein modulates androgen-dependent transcription program in prostate in a fashion similar to that observed in reporter mice by luciferase expression. In conclusion, genistein is a partial androgen agonist/antagonist in some but not in all mouse tissues and should be considered as a tissue-specific AR modulator.


Assuntos
Encéfalo/efeitos dos fármacos , Genisteína/farmacologia , Fitoestrógenos/farmacologia , Próstata/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Testículo/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Regiões Promotoras Genéticas/efeitos dos fármacos , Próstata/metabolismo , Transdução de Sinais/efeitos dos fármacos , Testículo/metabolismo
14.
EMBO J ; 30(19): 3962-76, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21915096

RESUMO

High androgen receptor (AR) level in primary tumour predicts increased prostate cancer-specific mortality. However, the mechanisms that regulate AR function in prostate cancer are poorly known. We report here a new paradigm for the forkhead protein FoxA1 action in androgen signalling. Besides pioneering the AR pathway, FoxA1 depletion elicited extensive redistribution of AR-binding sites (ARBs) on LNCaP-1F5 cell chromatin that was commensurate with changes in androgen-dependent gene expression signature. We identified three distinct classes of ARBs and androgen-responsive genes: (i) independent of FoxA1, (ii) pioneered by FoxA1 and (iii) masked by FoxA1 and functional upon FoxA1 depletion. FoxA1 depletion also reprogrammed AR binding in VCaP cells, and glucocorticoid receptor binding and glucocorticoid-dependent signalling in LNCaP-1F5 cells. Importantly, FoxA1 protein level in primary prostate tumour had significant association to disease outcome; high FoxA1 level was associated with poor prognosis, whereas low FoxA1 level, even in the presence of high AR expression, predicted good prognosis. The role of FoxA1 in androgen signalling and prostate cancer is distinctly different from that in oestrogen signalling and breast cancer.


Assuntos
Androgênios/metabolismo , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Neoplasias da Próstata/metabolismo , Motivos de Aminoácidos , Linhagem Celular Tumoral , Feminino , Glucocorticoides/metabolismo , Humanos , Masculino , Ligação Proteica , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais , Transcrição Gênica
15.
Mol Endocrinol ; 25(2): 269-81, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21193555

RESUMO

Human sex hormone-binding globulin (SHBG) accumulates within the cytoplasm of epithelial cells lining the proximal convoluted tubules of mice expressing human SHBG transgenes. The main ligands of SHBG, testosterone and its metabolite, 5α-dihydrotestosterone (DHT), alter expression of androgen-responsive genes in the kidney. To determine how intracellular SHBG might influence androgen action, we used a mouse proximal convoluted tubule (PCT) cell line with characteristics of S1/S2 epithelial cells in which human SHBG accumulates. Western blotting revealed that SHBG extracted from PCT cells expressing a human SHBG cDNA (PCT-SHBG) is 5-8 kDa smaller than the SHBG secreted by these cells, due to incomplete N-glycosylation and absence of O-linked oligosaccharides. PCT-SHBG cells sequester [(3)H]DHT more effectively from culture medium than parental PCT cells, and the presence of SHBG accentuates androgen-dependent activation of a luciferase reporter gene, as well as the endogenous kidney androgen-regulated protein (Kap) gene. After androgen withdrawal, androgen-induced Kap mRNA levels in PCT-SHBG cells are maintained for more than 2 wk vs 2 d in parental PCT cells. Transcriptome profiling after testosterone or DHT pretreatments, followed by 3 d of steroid withdrawal, also demonstrated that intracellular SHBG enhances androgen-dependent stimulation (e.g. Adh7, Vcam1, Areg, Tnfaip2) or repression (e.g. Cldn2 and Osr2) of many other genes in PCT cells. In addition, nuclear localization of the androgen receptor is enhanced and retained longer after steroid withdrawal in PCT cells containing functional SHBG. Thus, intracellular SHBG accentuates the uptake of androgens and sustains androgens access to the androgen receptor, especially under conditions of limited androgen supply.


Assuntos
Androgênios/metabolismo , Citoplasma/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Globulina de Ligação a Hormônio Sexual/metabolismo , Animais , Western Blotting , Linhagem Celular , Di-Hidrotestosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Glicosilação , Humanos , Camundongos , Oligossacarídeos/metabolismo , Isoformas de Proteínas , Proteínas/genética , RNA Mensageiro/análise , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Globulina de Ligação a Hormônio Sexual/genética , Testosterona/metabolismo , Testosterona/farmacologia
16.
Mol Cell Proteomics ; 9(6): 1352-67, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20308691

RESUMO

Estrogen receptor alpha (ERalpha) is a modular protein of the steroid/nuclear receptor family of transcriptional regulators that upon binding to the hormone undergoes structural changes, resulting in its nuclear translocation and docking to specific chromatin sites. In the nucleus, ERalpha assembles in multiprotein complexes that act as final effectors of estrogen signaling to the genome through chromatin remodeling and epigenetic modifications, leading to dynamic and coordinated regulation of hormone-responsive genes. Identification of the molecular partners of ERalpha and understanding their combinatory interactions within functional complexes is a prerequisite to define the molecular basis of estrogen control of cell functions. To this end, affinity purification was applied to map and characterize the ERalpha interactome in hormone-responsive human breast cancer cell nuclei. MCF-7 cell clones expressing human ERalpha fused to a tandem affinity purification tag were generated and used to purify native nuclear ER-containing complexes by IgG-Sepharose affinity chromatography and glycerol gradient centrifugation. Purified complexes were analyzed by two-dimensional DIGE and mass spectrometry, leading to the identification of a ligand-dependent multiprotein complex comprising beta-actin, myosins, and several proteins involved in actin filament organization and dynamics and/or known to participate in actin-mediated regulation of gene transcription, chromatin dynamics, and ribosome biogenesis. Time course analyses indicated that complexes containing ERalpha and actin are assembled in the nucleus early after receptor activation by ligands, and gene knockdown experiments showed that gelsolin and the nuclear isoform of myosin 1c are key determinants for assembly and/or stability of these complexes. Based on these results, we propose that the actin network plays a role in nuclear ERalpha actions in breast cancer cells, including coordinated regulation of target gene activity, spatial and functional reorganization of chromatin, and ribosome biogenesis.


Assuntos
Actinas/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Linhagem Celular Tumoral , Cromatografia de Afinidade , Cromatografia Líquida , Eletroforese em Gel Bidimensional , Receptor alfa de Estrogênio/isolamento & purificação , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lentivirus/genética , Espectrometria de Massas , Modelos Biológicos , Complexos Multiproteicos/isolamento & purificação , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo
17.
Genes Dev ; 24(4): 381-95, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20159957

RESUMO

The orphan receptor LRH-1 and the oxysterol receptors LXRalpha and LXRbeta are established transcriptional regulators of lipid metabolism that appear to control inflammatory processes. Here, we investigate the anti-inflammatory actions of these nuclear receptors in the hepatic acute phase response (APR). We report that selective synthetic agonists induce SUMOylation-dependent recruitment of either LRH-1 or LXR to hepatic APR promoters and prevent the clearance of the N-CoR corepressor complex upon cytokine stimulation. Investigations of the APR in vivo, using LXR knockout mice, indicate that the anti-inflammatory actions of LXR agonists are triggered selectively by the LXRbeta subtype. We further find that hepatic APR responses in small ubiquitin-like modifier-1 (SUMO-1) knockout mice are increased, which is due in part to diminished LRH-1 action at APR promoters. Finally, we provide evidence that the metabolically important coregulator GPS2 functions as a hitherto unrecognized transrepression mediator of interactions between SUMOylated nuclear receptors and the N-CoR corepressor complex. Our study extends the knowledge of anti-inflammatory mechanisms and pathways directed by metabolic nuclear receptor-corepressor networks to the control of the hepatic APR, and implies alternative pharmacological strategies for the treatment of human metabolic diseases associated with inflammation.


Assuntos
Reação de Fase Aguda/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Fígado/imunologia , Receptores Nucleares Órfãos/imunologia , Receptores Citoplasmáticos e Nucleares/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Animais , Anti-Inflamatórios/imunologia , Células COS , Chlorocebus aethiops , Feminino , Regulação da Expressão Gênica , Células HeLa , Humanos , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
Inflamm Bowel Dis ; 16(3): 469-73, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19714760

RESUMO

BACKGROUND: The pathophysiological basis for corticosteroid (CS) failure in ulcerative colitis (UC) is unknown. A transactivation glucocorticoid bioassay (GBA) was developed to measure the biological activity of CS by quantifying glucocorticoid response elements. This approach eliminates differences in bioavailability, chemistry, affinity, and other potential differences between the various steroids regarding their ability to activate the glucocorticoid receptor. In this multicenter prospective study, we aimed to evaluate whether CS bioavailability plays a role in CS refractoriness in severe pediatric UC. METHODS: GBA (using COS-1 transfected cells) was measured in the serum of 50 children (52% males, age 13.4 +/- 3.5 years) admitted for acute severe UC on the third day of CS treatment. Demographic, clinical, and laboratory data were prospectively recorded. RESULTS: Of the children enrolled, 16 (32%) failed CS therapy and required infliximab (n = 14) or colectomy (n = 2) within a median of 10 days (interquartile range [IQR] 6.5-14.5). Reflecting internal validity of the assay, GBA was highly correlated with the last CS dose and the time interval to bloodletting (r = -0.41 and r = -0.54, respectively; P < 0.001). There was no statistically significant difference in the GBA levels between responders and nonresponders (249 nM versus 200 nM cortisol equivalent, P = 0.18). In a multivariate regression model adjusted for time elapsed from CS and the administered dose, GBA did not predict response to CS (P = 0.34). CONCLUSIONS: The lack of correlation of GBA level and treatment outcome lends support to the hypothesis that the bioavailability, type, and dosing of intravenous CS are not associated with response or failure to the drug.


Assuntos
Bioensaio/métodos , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/imunologia , Monitoramento de Medicamentos/métodos , Glucocorticoides , Adolescente , Animais , Anti-Inflamatórios/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Bioensaio/normas , Células COS , Criança , Chlorocebus aethiops , Colectomia , Colite Ulcerativa/cirurgia , Feminino , Glucocorticoides/administração & dosagem , Glucocorticoides/sangue , Glucocorticoides/imunologia , Hospitalização , Humanos , Infliximab , Injeções Intravenosas , Masculino , Análise Multivariada , Valor Preditivo dos Testes , Estudos Prospectivos , Análise de Regressão , Reprodutibilidade dos Testes
19.
Mol Cell Endocrinol ; 317(1-2): 14-24, 2010 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-20035825

RESUMO

The androgen receptor (AR) mediates the effects of male sex steroids. There are major sex differences in lung development and pathologies, including lung cancer. In this report, we show that Ar is mainly expressed in type II pneumocytes and the bronchial epithelium of murine lung and that androgen treatment increases AR protein levels in lung cells. Androgen administration altered significantly murine lung gene expression profiles; for example, by up-regulating transcripts involved in oxygen transport and down-regulating those in DNA repair and DNA recombination. Androgen exposure also affected the gene expression profile in a human lung adenocarcinoma-derived cell line, A549, by up- or down-regulating significantly some 200 transcripts, including down-regulation of genes involved in cell respiration. Dexamethasone treatment of A549 cells augmented expression of transcript sets that overlapped in part with those up-regulated by androgen in these cells. Moreover, a human lung cancer tissue array revealed that different lung cancer types are all AR-positive. Our results indicate that adult lung is an AR target tissue and suggest that AR plays a role in lung cancer biology.


Assuntos
Androgênios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Receptores Androgênicos/genética , Animais , Linhagem Celular Tumoral , Dexametasona/farmacologia , Elementos Facilitadores Genéticos/genética , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Hemoglobinas/metabolismo , Humanos , Pulmão/citologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Testosterona/farmacologia , Regulação para Cima/efeitos dos fármacos
20.
Cancer Res ; 69(20): 8141-9, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19808968

RESUMO

Androgen receptor (AR) is known to be overexpressed in castration-resistant prostate cancer. To interrogate the functional significance of the AR level, we established two LNCaP cell sublines expressing in a stable fashion two to four times (LNCaP-ARmo) and four to six times (LNCaP-ARhi) higher level of AR than the parental cell line expressing the empty vector (LNCaP-pcDNA3.1). LNCaP-ARhi cell line grew faster than the control line in low concentrations, especially in 1 nmol/L 5alpha-dihydrotestosterone (DHT). Microarray-based transcript profiling and subsequent unsupervised hierarchical clustering showed that LNCaP-ARhi cells clustered together with VCaP cells, containing endogenous AR gene amplification and overexpression, indicating the central role of AR in the overall regulation of gene expression in prostate cancer cells. Two hundred forty genes showed >2-fold changes on DHT treatment in LNCaP-ARhi at 4 h time point, whereas only 164 and 52 showed changes in LNCaP-ARmo and LNCaP-pcDNA3.1, respectively. Many androgen-regulated genes were upregulated in LNCaP-ARhi at 10-fold lower concentration of DHT than in control cells. DHT (1 nmol/L) increased expression of several cell cycle-associated genes in LNCaP-ARhi cells. ChIP-on-chip assay revealed the presence of chromatin binding sites for AR within +/-200 kb of most of these genes. The growth of LNCaP-ARhi cells was also highly sensitive to cyclin-dependent kinase inhibitor, roscovitine, at 1nmol/L DHT. In conclusion, our results show that overexpression of AR sensitizes castration-resistant prostate cancer cells to the low levels of androgens. The activity of AR signaling pathway is regulated by the levels of both ligand and the receptor.


Assuntos
Androgênios/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Antineoplásicos/farmacologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Relação Dose-Resposta a Droga , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Masculino , Neoplasias Hormônio-Dependentes/genética , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/genética , Purinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Roscovitina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...