Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Br J Cancer ; 113(10): 1502-11, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26575822

RESUMEN

BACKGROUND: There is an acute need to uncover biomarkers that reflect the molecular pathologies, underpinning prostate cancer progression and poor patient outcome. We have previously demonstrated that in prostate cancer cell lines PDE4D7 is downregulated in advanced cases of the disease. To investigate further the prognostic power of PDE4D7 expression during prostate cancer progression and assess how downregulation of this PDE isoform may affect disease outcome, we have examined PDE4D7 expression in physiologically relevant primary human samples. METHODS: About 1405 patient samples across 8 publically available qPCR, Affymetrix Exon 1.0 ST arrays and RNA sequencing data sets were screened for PDE4D7 expression. The TMPRSS2-ERG gene rearrangement status of patient samples was determined by transformation of the exon array and RNA seq expression data to robust z-scores followed by the application of a threshold>3 to define a positive TMPRSS2-ERG gene fusion event in a tumour sample. RESULTS: We demonstrate that PDE4D7 expression positively correlates with primary tumour development. We also show a positive association with the highly prostate cancer-specific gene rearrangement between TMPRSS2 and the ETS transcription factor family member ERG. In addition, we find that in primary TMPRSS2-ERG-positive tumours PDE4D7 expression is significantly positively correlated with low-grade disease and a reduced likelihood of progression after primary treatment. Conversely, PDE4D7 transcript levels become significantly decreased in castration resistant prostate cancer (CRPC). CONCLUSIONS: We further characterise and add physiological relevance to PDE4D7 as a novel marker that is associated with the development and progression of prostate tumours. We propose that the assessment of PDE4D7 levels may provide a novel, independent predictor of post-surgical disease progression.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/cirugía , Progresión de la Enfermedad , Regulación hacia Abajo , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Análisis de Secuencia de ARN
2.
Adv Exp Med Biol ; 867: 93-114, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26530362

RESUMEN

In this chapter the use of prostate specific antigen (PSA) as a tumor marker for prostate cancer is discussed. The chapter provides an overview of biological and clinical aspects of PSA. The main drawback of total PSA (tPSA) is its lack of specificity for prostate cancer which leads to unnecessary biopsies. Moreover, PSA-testing poses a risk of overdiagnosis and subsequent overtreatment. Many PSA-based markers have been developed to improve the performance characteristics of tPSA. As well as different molecular subforms of tPSA, such as proPSA (pPSA) and free PSA (fPSA), and PSA derived kinetics as PSA-velocity (PSAV) and PSA-doubling time (PSADT). The prostate health index (phi), PSA-density (PSAD) and the contribution of non PSA-based markers such as the urinary transcripts of PCA3 and TMPRSS-ERG fusion are also discussed. To enable further risk stratification tumor markers are often combined with clinical data (e.g. outcome of DRE) in so-called nomograms. Currently the role of magnetic resonance imaging (MRI) in the detection and staging of prostate cancer is being explored.


Asunto(s)
Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/diagnóstico , Detección Precoz del Cáncer , Humanos , Imagen por Resonancia Magnética , Masculino , Neoplasias de la Próstata/sangre
3.
Br J Cancer ; 110(5): 1278-87, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24518597

RESUMEN

BACKGROUND: Isoforms of the PDE4 family of cAMP-specific phosphodiesterases (PDEs) are expressed in a cell type-dependent manner and contribute to underpinning the paradigm of intracellular cAMP signal compartmentalisation. Here we identify the differential regulation of the PDE4D7 isoform during prostate cancer progression and uncover a role in controlling prostate cancer cell proliferation. METHODS: PDE4 transcripts from 19 prostate cancer cell lines and xenografts were quantified by qPCR. PDE4D7 expression was further investigated because of its significant downregulation between androgen-sensitive (AS) and androgen-insensitive (AI) samples. Western blot analysis, PDE activity assay, immunofluorescent staining and cAMP responsive FRET assays were used to investigate the sub-plasma membrane localisation of a population of PDE4D7 in VCaP (AS) and PC3 (AI) cell lines. Disruption of this localisation pattern using dominant-negative protein expression and siRNA knockdown showed that PDE4D7 acts in opposition to proliferative signalling as assessed by electrical impedance-based proliferation assays. RESULTS: Here we identify the differential regulation of the PDE4D7 isoform during prostate cancer progression. PDE4D7 is highly expressed in AS cells and starkly downregulated in AI samples. The significance of this downregulation is underscored by our finding that PDE4D7 contributes a major fraction of cAMP degrading PDE activity tethered at the plasma membrane and that displacement of PDE4D7 from this compartment leads to an increase in the proliferation of prostate cancer cells. PDE4D7 mRNA expression is not, however, directly regulated by the androgen receptor signalling axis despite an overlapping genomic structure with the androgen responsive gene PART1. PDE4D7, which locates to the plasma membrane, acts to supress aberrant non-steroidal growth signals within the prostate or AS metastasis. CONCLUSIONS: PDE4D7 expression is significantly downregulated between AS and AI cell phenotypes. This change in expression potentially provides a novel androgen-independent biomarker and manipulation of its activity or its expression may provide therapeutic possibilities and insights into contributory aspects of the complex molecular pathology of prostate cancer.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , 3',5'-AMP Cíclico Fosfodiesterasas/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , AMP Cíclico/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Regulación hacia Abajo , Humanos , Isoenzimas , Masculino , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal
4.
Br J Cancer ; 105(6): 854-63, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21847120

RESUMEN

BACKGROUND: OCT3/4 (POU5F1) is an established diagnostic immunohistochemical marker for specific histological variants of human malignant germ cell tumours (GCTs), including the seminomatous types and the stem cell component of non-seminomas, known as embryonal carcinoma. OCT3/4 is crucial for the regulation of pluripotency and the self-renewal of normal embryonic stem- and germ cells. Detection of expression of this transcription factor is complicated by the existence of multiple pseudogenes and isoforms. Various claims have been made about OCT3/4 expression in non-GCTs, possibly related to using nonspecific detection methods. False-positive findings undermine the applicability of OCT3/4 as a specific diagnostic tool in a clinical setting. In addition, false-positive findings could result in misinterpretation of pluripotency regulation in solid somatic cancers and their stem cells. Of the three identified isoforms--OCT4A, OCT4B and OCT4B1--only OCT4A proved to regulate pluripotency. Up until now, no convincing nuclear OCT4A protein expression has been shown in somatic cancers or tissues. METHODS: This study investigates expression of the various OCT3/4 isoforms in GCTs (both differentiated and undifferentiated) and somatic (non-germ cell) cancers, including representative cell lines and xenografts. RESULTS: Using specific methods, OCT4A and OCT4B1 are shown to be preferentially expressed in undifferentiated GCTs. The OCT4B variant shows no difference in expression between GCTs (either differentiated or undifferentiated) and somatic cancers. In spite of the presence of OCT4A mRNA in somatic cancer-derived cell lines, no OCT3/4 protein is detected. Significant positive correlations between all isoforms of OCT3/4 were identified in both tumours with and without a known stem cell component, possibly indicating synergistic roles of these isoforms. CONCLUSION: This study confirms that OCT4A protein only appears in seminomatous GCTs, embryonal carcinoma and representative cell lines. Furthermore, it emphasises that in order to correctly assess the presence of functional OCT3/4, both isoform specific mRNA and protein detection are required.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Carcinoma Embrionario/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de Células Germinales y Embrionarias , Factor 3 de Transcripción de Unión a Octámeros/genética , Isoformas de Proteínas/metabolismo , Seudogenes , ARN Mensajero/metabolismo , Seminoma/metabolismo , Sensibilidad y Especificidad
5.
Sci Rep ; 11(1): 20760, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675268

RESUMEN

Urinary extracellular vesicles (EVs) have gained increased interest as a biomarker source. Clinical implementation on a daily basis requires protocols that inevitably includes short-term storage of the clinical samples, especially when collected at home. However, little is known about the effect of delayed processing on the urinary EVs concentration and proteome. We evaluated two storage protocols. First, urine stored at 4 °C. Secondly a protocol compatible with at-home collection, in which urine was stored with the preservative EDTA at room temperature (RT). EVs were isolated using the ME-kit (VN96-peptide). For both conditions we explored the effect of storage duration (0, 2, 4 and 8 days) on EV concentration and proteome using EVQuant and data-independent acquisition mass spectrometry, respectively. The urinary EV concentration and proteome was highly stable using both protocols, in terms of protein number and quantitative changes. Furthermore, EDTA does not affect the urinary EV concentration or global proteome. In conclusion, urine can be stored either at 4 °C or with EDTA at RT for up to 8 days without any significant decay in EV concentration or a notable effect on the EV-proteome. These findings open up biomarker studies in urine collected via self-sampling at home.


Asunto(s)
Vesículas Extracelulares/química , Proteoma/análisis , Humanos , Proteómica/métodos , Espectrometría de Masas en Tándem , Urinálisis/métodos , Toma de Muestras de Orina/métodos
6.
Front Oncol ; 10: 608, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582525

RESUMEN

Extracellular vesicles (EVs) have great potential as biomarkers since their composition and concentration in biofluids are disease state dependent and their cargo can contain disease-related information. Large tumor-derived EVs (tdEVs, >1 µm) in blood from cancer patients are associated with poor outcome, and changes in their number can be used to monitor therapy effectiveness. Whereas, small tumor-derived EVs (<1 µm) are likely to outnumber their larger counterparts, thereby offering better statistical significance, identification and quantification of small tdEVs are more challenging. In the blood of cancer patients, a subpopulation of EVs originate from tumor cells, but these EVs are outnumbered by non-EV particles and EVs from other origin. In the Dutch NWO Perspectief Cancer-ID program, we developed and evaluated detection and characterization techniques to distinguish EVs from non-EV particles and other EVs. Despite low signal amplitudes, we identified characteristics of these small tdEVs that may enable the enumeration of small tdEVs and extract relevant information. The insights obtained from Cancer-ID can help to explore the full potential of tdEVs in the clinic.

7.
Oncogene ; 5(4): 497-503, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1970152

RESUMEN

Amplification and overexpression of the neu gene have been found in several human adenocarcinomas. We have obtained monoclonal antibodies to the human neu protein by immunizing a Balb/c mouse with a Balb/c cell line expressing the human neu gene by transfection. The monoclonal antibodies reacted with neu protein on intact cells by immunofluorescence and immunoprecipitated neu in metabolically labeled cells, also in the presence of tunicamycin. We tested possible down-modulating effects of these monoclonal antibodies on SKBR-3 mammary tumor cells, which express high levels of wild-type human neu protein. We also used NIH3T3 cells transfected with either a normal or a mutated human neu gene, encoding a protein with a valine to glutamic acid substitution in the transmembrane domain. Down-modulation of the normal cell-surface neu protein was inefficient. In contrast, the antibodies induced 50-65% down-modulation in NIH3T3 cells expressing the mutated human neu protein and could inhibit these cells to form colonies in soft agar. We propose that these differences are due to changed aggregation properties of the point-mutated protein.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Regulación Neoplásica de la Expresión Génica , Mutación , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Animales , Adhesión Celular , División Celular , Línea Celular , Células Cultivadas , Células Clonales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias Mamarias Experimentales , Ratones , Ratones Endogámicos BALB C/inmunología , Plásmidos , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas/inmunología , Receptor ErbB-2
8.
Mol Endocrinol ; 14(5): 753-60, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10809237

RESUMEN

Previous studies have shown that androgen up-regulates expression of the p21 (WAF1, CIP1, SDI1, CAP20) gene, which contains a canonical androgen response element (ARE) in its proximal promoter region. We undertook the current studies to determine whether elements in the p21 promoter other than the ARE mediate androgen action. We found that deletion of the ARE did not completely abolish the promoter responsiveness to androgen, suggesting that additional cis-regulatory elements within the p21 core promoter may also be involved in androgen responsiveness. The p21 core promoter is GC-rich and contains six binding sites for transcription factor Sp1. We determined whether one or more of these Sp1 sites mediate androgen responsiveness of the p21 promoter. To do so, we used a transient transfection assay with p21 promoter-luciferase reporter constructs. The reporter activity of a construct lacking the ARE but containing all six Sp1 sites was induced approximately 3-fold by androgen. Mutation of Sp1-3 nearly eliminated basal promoter activity as well as androgen responsiveness, whereas deletion of Sp1-1 and Sp1-2 sites and mutation of Sp1-4, Sp1-5, and Sp1-6 sites had relatively little effect. We also used the mammalian one-hybrid assay and coimmunoprecipitation assay to show that androgen receptor (AR) and transcription factor Sp1 interact with one another. The current studies suggest a model in which AR and transcription factor Sp1 not only bind to their respective consensus sites within the p21 promoter, but also complex with one another, thereby recruiting coactivators and general transcription factors and inducing p21 transcription.


Asunto(s)
Andrógenos/farmacología , Ciclinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Receptores Androgénicos/fisiología , Factor de Transcripción Sp1/fisiología , Adenocarcinoma/patología , Andrógenos/fisiología , Secuencia de Bases , Secuencia de Consenso , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Genes Reporteros , Humanos , Luciferasas/genética , Sustancias Macromoleculares , Masculino , Metribolona/farmacología , Modelos Genéticos , Datos de Secuencia Molecular , Neoplasias Hormono-Dependientes/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Neoplasias de la Próstata/patología , Proteínas Recombinantes de Fusión/biosíntesis , Secuencias Reguladoras de Ácidos Nucleicos , Eliminación de Secuencia , Estimulación Química , Congéneres de la Testosterona/farmacología , Transfección , Células Tumorales Cultivadas
9.
Mol Endocrinol ; 5(10): 1396-404, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1775129

RESUMEN

A series of human androgen receptor (AR) deletion mutants was constructed to study the relationship between the structural domains and their different functions in the AR protein. Human AR mutants were expressed in COS-1 and HeLa cells to investigate hormone binding, transcriptional activation, and subcellular localization. The wild-type human AR (AR 1-910) was expressed as a 110- to 112-kDa doublet, as revealed on immunoblots. All mutant AR proteins also migrated as doublets, except for one. This AR has a deletion from amino acid residues 51-211 and migrated as a single protein band, possibly due to altered posttranslational modification. The AR steroid-binding domain is encoded by approximately 250 amino acid residues in the C-terminal end. Deletions in this domain as well as truncation of the last 12 C-terminal amino acid residues abolished hormone binding. Cotransfection studies in HeLa cells showed that transcriptional activation of an androgen-regulated reporter gene construct was induced by the wild-type human AR. Mutational analysis revealed two regions in the N-terminal part, encoded by amino acid residues 51-211 and 244-360, to be essential for this transcriptional activation. Deletion of the hormone-binding domain yielded a constitutively active AR protein, indicating that in the absence of hormone this domain displays an inhibitory function. In the presence of its ligand, the wild-type AR was located in the cell nucleus. In the absence of androgens the receptor was mainly nuclear, but cytoplasmic localization was observed as well.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Proteínas de Unión al ADN/genética , Metribolona/metabolismo , Receptores Androgénicos/genética , Transcripción Genética , Animales , Sitios de Unión , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Deleción Cromosómica , Proteínas de Unión al ADN/metabolismo , Vectores Genéticos , Células HeLa , Humanos , Inmunohistoquímica , Plásmidos , Receptores Androgénicos/metabolismo , Proteínas Recombinantes/metabolismo , Mapeo Restrictivo , Transfección
10.
Mol Endocrinol ; 5(10): 1562-9, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1775137

RESUMEN

We have identified two different single nucleotide alterations in codon 686 (GAC; aspartic acid) in exon 4 of the human androgen receptor gene in three unrelated families with the complete form of androgen insensitivity. One mutation (G----C) results in an aspartic acid----histidine substitution (with 15-20% of wild-type androgen-binding capacity), whereas the other mutation (G----A) leads to an aspartic acid----asparagine substitution (with normal androgen-binding capacity, but a rapidly dissociating ligand-receptor complex). The mutations eliminate a Hinfl restriction site. Screening for the loss of the Hinfl site in both families with the Asp----Asn mutation resulted in the recognition of heterozygous carriers in successive generations of each. Both mutant androgen receptors were generated in vitro and transiently expressed in COS and HeLa cells. The receptor proteins produced had the same altered binding characteristics as those measured in fibroblasts from the affected subjects. R1881-activated transcription of a GRE-tk-CAT reporter gene construct was strongly diminished by both mutant receptors and was only partially restored using a 100-fold higher concentration of ligand compared with wild-type receptor. Thus, aspartic acid-686 appears essential for normal androgen receptor function. Substitution of this amino acid residue, by either histidine or asparagine, results in androgen insensitivity and lack of androgen-dependent male sexual differentiation.


Asunto(s)
Ácido Aspártico , Mutagénesis Sitio-Dirigida , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Asparagina , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Codón/genética , Exones , Femenino , Fibroblastos/metabolismo , Células HeLa , Histidina , Humanos , Cinética , Masculino , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Linaje , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/metabolismo , Piel/metabolismo , Transfección
11.
Oncogene ; 34(5): 568-77, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24488012

RESUMEN

Gene fusions, mainly between TMPRSS2 and ERG, are frequent early genomic rearrangements in prostate cancer (PCa). In order to discover novel genomic fusion events, we applied whole-genome paired-end sequencing to identify structural alterations present in a primary PCa patient (G089) and in a PCa cell line (PC346C). Overall, we identified over 3800 genomic rearrangements in each of the two samples as compared with the reference genome. Correcting these structural variations for polymorphisms using whole-genome sequences of 46 normal samples, the numbers of cancer-related rearrangements were 674 and 387 for G089 and PC346C, respectively. From these, 192 in G089 and 106 in PC346C affected gene structures. Exclusion of small intronic deletions left 33 intergenic breaks in G089 and 14 in PC346C. Out of these, 12 and 9 reassembled genes with the same orientation, capable of generating a feasible fusion transcript. Using PCR we validated all the reliable predicted gene fusions. Two gene fusions were in-frame: MPP5-FAM71D in PC346C and ARHGEF3-C8ORF38 in G089. Downregulation of FAM71D and MPP5-FAM71D transcripts in PC346C cells decreased proliferation; however, no effect was observed in the RWPE-1-immortalized normal prostate epithelial cells. Together, our data showed that gene rearrangements frequently occur in PCa genomes but result in a limited number of fusion transcripts. Most of these fusion transcripts do not encode in-frame fusion proteins. The unique in-frame MPP5-FAM71D fusion product is important for proliferation of PC346C cells.


Asunto(s)
Proliferación Celular/genética , Proteínas de la Membrana/genética , Nucleósido-Fosfato Quinasa/genética , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/genética , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Nucleósido-Fosfato Quinasa/biosíntesis , Proteínas de Fusión Oncogénica/aislamiento & purificación , Neoplasias de la Próstata/patología , Factores de Intercambio de Guanina Nucleótido Rho/biosíntesis , Factores de Intercambio de Guanina Nucleótido Rho/genética
12.
Endocrinology ; 140(9): 4070-80, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10465278

RESUMEN

Prolonged exposure of the developing neonatal ovine uterus to a progestin from birth prevents uterine gland development and creates an adult endometrial phenotype characterized by the absence of glandular epithelium, the uterine gland knockout (UGKO) phenotype. This study used endometrium from normal and UGKO sheep to identify messenger RNAs (mRNAs) expressed differentially in the endometrial epithelium using the molecular techniques of mRNA differential display PCR (DD-PCR) and suppression subtractive complementary DNA (cDNA) hybridization (SSH). Sequence analyses of DD- and SSH-identified and cloned cDNAs indicated similarity of some to known mRNAs, including beta-lactoglobulin, alkaline phosphatase, type B and D endogenous sheep retroviruses, gp330/megalin, matrix Gla protein, and others. Other cDNAs were not similar to any known sequences and are considered novel, although some of these match human expressed sequence tags. In situ hybridization analyses of uteri from cyclic and pregnant ewes indicated that all DD-PCR- and SSH-identified mRNAs were expressed in either the endometrial lumenal and/or glandular epithelium, although some were also expressed in other uterine cell types. Northern and in situ hybridization analyses revealed that patterns of mRNA expression for most clones were affected by the day of the estrous cycle and pregnancy in a manner consistent with regulation by progesterone. Studies demonstrate the utility of the ovine UGKO model as a tool with which to identify known and novel uterine epithelial-specific genes. Cloned cDNAs identified here are expressed sequence tags useful for comparative and physical genetic mapping and may be used to reveal new factors and pathways regulating endometrial function.


Asunto(s)
Endometrio/metabolismo , ARN Mensajero/metabolismo , Ovinos/metabolismo , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Endometrio/efectos de los fármacos , Endometrio/crecimiento & desarrollo , Epitelio/efectos de los fármacos , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Estro/fisiología , Femenino , Biblioteca de Genes , Hibridación in Situ , Hibridación de Ácido Nucleico/métodos , Reacción en Cadena de la Polimerasa/métodos , Embarazo , Progestinas/farmacología , Factores de Tiempo
13.
Semin Oncol ; 26(4): 407-21, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10482183

RESUMEN

Androgens are essential for the development, growth, and maintenance of the prostate. They exert their effects via the intracellular androgen receptor (AR), which is a ligand-dependent transcription activator. As is the case with normal prostate development, primary prostatic cancers are largely dependent on androgens for growth and survival. Most patients respond favorably to androgen ablation and antiandrogen therapy, which has become a standard treatment of metastatic disease. However, virtually all patients will relapse with clinically defined androgen-independent cancer. This phenomenon raises the question of how cancer cells survive and grow in the low androgen environment? Two of the routes cells can take to adapt are (1) bypassing and (2) sensitizing the AR pathway. The vast numbers of AR abnormalities observed in prostate tumors from patients treated with hormonal therapy suggest that many cells sensitize or change the AR pathway. To continue to activate this pathway in a low androgen environment, cells can (1) mutate the AR to become promiscuously activated by different steroids, (2) amplify the AR, (3) activate the AR in a ligand-independent manner by growth factors and cytokines, or (4) amplify coactivators. Alternatively, prostate cancer cells that have lost AR expression must have bypassed the AR pathway. Activation of oncogenes and autocrine growth factor stimulation are two mechanisms that likely contribute to becoming completely androgen-independent. From all the studies on AR function in prostate cancer, it is clear that the AR plays an important role in cancer development and progression. Moreover, the AR pathway remains important in most cells from patients with clinically defined androgen-independent prostate cancer.


Asunto(s)
Neoplasias Hormono-Dependientes/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Andrógenos/fisiología , Progresión de la Enfermedad , Humanos , Masculino , Neoplasias Hormono-Dependientes/fisiopatología , Próstata/fisiología , Neoplasias de la Próstata/fisiopatología , Receptores Androgénicos/genética
14.
Mol Cell Endocrinol ; 143(1-2): 1-7, 1998 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-9806345

RESUMEN

The past 3 years have been an exciting time in the field of hormone receptor research because of the discovery and characterization of novel groups of proteins that mediate the transcriptional activity of steroid receptors. These classes of proteins, called coactivators and corepressors, have greatly enhanced our understanding of how steroid receptors activate or inhibit transcription of their target genes. Multiple coactivators have been identified that fit the definition of a protein that connects or bridges the DNA-bound receptor to proteins in the preinitiation complex and thereby enhance transcription. Besides this bridging function, some coactivators can modify chromatin by histone acetylation and make promoters more accessible for the binding of other transcription factors. This finding explains old data concerning steroid receptor-induced nucleosome displacement and indicates a dual role for coactivators as bridging factors and chromatin remodeling proteins. The opposites of coactivators are corepressors, which are recruited into the receptor-DNA-bound complex in the absence of ligand and actively inhibit transcription of the target gene. Although unliganded steroid receptors are associated with heat shock proteins and do not bind to their response elements, the binding of antagonists to these receptors can result in the recruitment of corepressors. The expression level and repertoire of coactivators and corepressors have become important determinants in the functional activity of steroid hormones and their receptors.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Esteroides/fisiología , Animales , Humanos , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología
15.
Mol Cell Endocrinol ; 184(1-2): 115-23, 2001 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-11694347

RESUMEN

Previous studies have established that the cell-cell adhesion molecule-1 (CEACAM1, previously known as C-CAM1) functions as a tumor suppressor in prostate cancer and is involved in the regulation of prostate growth and differentiation. However, the molecular mechanism that modulates CEACAM1 expression in the prostate is not well defined. Since the growth of prostate epithelial cells is androgen-regulated, we investigated the effects of androgen and the androgen receptor (AR) on CEACAM1 expression. Transient transfection experiments showed that the AR can enhance the Ceacam1 promoter activity in a ligand-dependent manner and that the regulatory element resides within a relatively short (-249 to -194 bp) segment of the 5'-flanking region of the Ceacam1 gene. This androgen regulation is likely through direct AR-promoter binding because a mutant AR defective in DNA binding failed to upregulate reporter gene expression. Furthermore, electrophoretic mobility shift assays demonstrated that the AR specifically binds to this sequence, and mutation analysis of the potential ARE sequences revealed a region within the sequence that was required for the AR to activate the Ceacam1 gene. Therefore, the regulation of Ceacam1 gene expression by androgen may be one of the mechanisms by which androgen regulates prostatic function.


Asunto(s)
Andrógenos/fisiología , Antígenos CD/genética , Antígenos de Diferenciación/genética , Regulación de la Expresión Génica/efectos de los fármacos , Animales , Antígenos CD/efectos de los fármacos , Antígenos de Diferenciación/efectos de los fármacos , Sitios de Unión/genética , Moléculas de Adhesión Celular , Proteínas de Unión al ADN/fisiología , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/efectos de los fármacos , Ratas , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/fisiología , Transfección
16.
Mol Cell Endocrinol ; 183(1-2): 19-28, 2001 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11604220

RESUMEN

The androgen receptor (AR) is a ligand-dependent transcription activator responsible for male sexual development. In order to specifically inhibit the AR pathway, dominant negative ARs were constructed by inactivation of the major transactivation domains of the wild type AR and fusing this mutant (AR122) to the Krüppel-associated box (KRAB) repressor domain and/or histone deacetylase (HDAC1). The HDAC1-KRAB-AR122 protein was the most successful dominant negative AR, capable of repressing the wild type AR ninefold when co-expressed at a 1:1 plasmid ratio. A maximal repression of 41-fold was achieved when HDAC1-KRAB-AR122 was cotransfected with the wild type AR at a 4:1 plasmid ratio. HDAC1-KRAB-AR122 repressed transcription in a ligand-dependent manner since it inhibited a constitutively active AR mutant (AR5) only in the presence of agonists. High concentrations of partial agonists such as RU486, cyproterone acetate, and estradiol were also capable of triggering repression by HDAC1-KRAB-AR122. The potent dominant negative AR proteins might prove useful tools to inhibit AR function in vitro and in vivo.


Asunto(s)
Regulación de la Expresión Génica , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Genes Reporteros , Células HeLa , Histona Desacetilasas/genética , Humanos , Ligandos , Masculino , Regiones Promotoras Genéticas , Receptores Androgénicos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras/genética , Timidina Quinasa/genética
17.
J Steroid Biochem Mol Biol ; 41(3-8): 671-5, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1562540

RESUMEN

A series of human androgen receptor (AR) deletion mutants was constructed to study the relationship between various structural domains and their different functions in the AR protein. Immunoblots of wild type AR and AR mutants expressed in COS-1 cells, revealed a doublet appearance of all AR proteins. One exception was an AR mutant lacking amino acid residues 51-211 that migrated as a single protein band, possibly due to altered post-translational modification. The steroid binding domain was found to be encoded by approx. 250 amino acid residues in the C-terminal end. Deletions and truncations in this part of the receptor abolished hormone binding. The N-terminal domain was observed to be essential for transcriptional activation. AR mutants lacking large parts of this domain were transcriptionally inactive. Deletion of the hormone binding domain yielded a constitutively active AR protein, indicating that in the absence of hormone this domain displays an inhibitory function. In the absence of ligand the wild type AR expressed in COS-1 cells was distributed over nucleus and cytoplasm. The addition of hormone directed all androgen receptors to the nucleus. In contrast, an AR mutant lacking part of the DNA binding domain and part of the hinge region, was almost exclusively cytoplasmic in the absence of hormone. This mutant lacks a conserved region, homologous to the SV40 large T- and nucleoplasmin nuclear localization signal. Hormone induced transfer of this AR mutant to the nucleus, indicating the presence of a second, hormone dependent nuclear targeting mechanism.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Receptores Androgénicos/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Deleción Cromosómica , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Receptores Androgénicos/genética , Homología de Secuencia de Ácido Nucleico , Fracciones Subcelulares , Transfección
18.
J Steroid Biochem Mol Biol ; 41(3-8): 665-9, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1562539

RESUMEN

The human prostate tumor cell line LNCaP contains an abnormal androgen receptor system with broad steroid binding specificity. Progestagens, estradiol and several antiandrogens compete with androgens for binding to the androgen receptor in the cells to a higher extent than in other androgen sensitive systems. Optimal growth of LNCaP cells is observed after addition of the synthetic androgen R1881 (0.1 nM). In addition, estrogens, progestagens and several antiandrogens do not inhibit androgen responsive growth, but have striking growth stimulatory effects and increase EGF receptor level and acid phosphatase secretion. We have found that the androgen receptor in the LNCaP cells contains a single point mutation changing the sense of codon 868 (Thr to Ala) in the ligand binding domain. Expression vectors containing the normal or mutated androgen receptor sequence were transfected into COS or HeLa cells. Androgens, progestagens, estrogens and several antiandrogens bind the mutated androgen receptor protein and activate the expression of an androgen-regulated reporter gene (GRE-tk-CAT), indicating that the mutation directly affects both binding specificity and the induction of gene expression. Interestingly, the antiandrogen casodex showed antiandrogenic properties in growth studies of LNCaP cells and did not induce reporter gene activity in Hela cells transfected with the mutant receptor. The mutated androgen receptor of LNCaP cells is therefore a useful tool in the elucidation of different levels of action of steroids and antisteroids.


Asunto(s)
Antagonistas de Estrógenos/farmacología , Hormonas/farmacología , Mutación , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Esteroides/farmacología , Sitios de Unión , División Celular/efectos de los fármacos , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Células HeLa , Humanos , Masculino , Neoplasias de la Próstata , Transfección
19.
J Steroid Biochem Mol Biol ; 53(1-6): 443-8, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7626493

RESUMEN

Male sexual differentiation and development proceed under direct control of androgens. Androgen action is mediated by the intracellular androgen receptor, which belongs to the superfamily of ligand-dependent transcription factors. At least three pathological situations are associated with abnormal androgen receptor structure and function: androgen insensitivity syndrome (AIS), spinal and bulbar muscular atrophy (SBMA) and prostate cancer. In the X-linked androgen insensitivity syndrome, defects in the androgen receptor gene have prevented the normal development of both internal and external male structures in 46,XY individuals. Complete or gross deletions of the androgen receptor gene have not been found frequently in persons with complete androgen insensitivity syndrome. Point mutations at several different sites in exons 2-8 encoding the DNA- and androgen-binding domain, have been reported for partial and complete forms of androgen insensitivity. A relatively high number of mutations were reported in two different clusters in exon 5 and in exon 7. The number of mutations in exon 1 is extremely low and no mutations have been reported in the hinge region, located between the DNA-binding domain and the ligand-binding domain and which is encoded by the first half of exon 4. Androgen receptor gene mutations in prostate cancer are very rare and are reported only in exons 4-8. The X-linked spinal and bulbar muscle atrophy (SBMA; Kennedy's disease) is associated with an expanded length (> 40 residues) of one of the polyglutamine stretches in the N-terminal domain of the androgen receptor.


Asunto(s)
Andrógenos/fisiología , Enfermedad de la Neurona Motora/fisiopatología , Neoplasias de la Próstata/fisiopatología , Receptores Androgénicos/genética , Secuencia de Aminoácidos , Andrógenos/metabolismo , Sitios de Unión , Proteínas de Unión al ADN/genética , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Puntual , Síndrome
20.
J Steroid Biochem Mol Biol ; 41(3-8): 361-8, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1562511

RESUMEN

Discrete functions have been attributed to precise regions of the human androgen receptor (hAR) by expression of deletion mutants in COS and HeLa cells. A large C-terminal domain constitutes the hormone-binding region and a central basis, cysteine-rich domain is responsible for DNA binding. In addition, separate domains responsible for transactivation and nuclear translocation have been identified. In LNCaP cells (a prostate tumor cell line) the hAR is a heterogeneous protein which is synthesized as a single 110 kDa protein, but becomes rapidly phosphorylated to a 112 kDa protein. Metabolic labeling experiments using radioactive orthophosphate also indicated that the hAR is a phosphoprotein. Structural analysis of the AR gene in LNCaP cells and in 46, XY-individuals displaying androgen insensitivity (AIS) has revealed several different point mutations. In LNCaP cells the mutation affects both binding specificity and transactivation by different steroids. In a person with complete AIS a point mutation was identified in the splice donor site of intron 4, which prevents normal splicing and activates a cryptic splice donor site in exon 4. The consequence is a functionally inactive AR protein due to an in-frame deletion in the steroid-binding domain. In two unrelated individuals with complete AIS, two different single nucleotide alterations in codon 686 (Asp) were found. Both mutations resulted in functionally inactive ARs due to rapidly dissociating hormone-AR complexes. It is concluded that the hAR is a heterogeneous phosphoprotein in which functional errors have a dramatic impact on phenotype and fertility of 46, XY-individuals.


Asunto(s)
Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Deleción Cromosómica , Células HeLa , Humanos , Masculino , Datos de Secuencia Molecular , Mutagénesis , Fosforilación , Neoplasias de la Próstata/metabolismo , Procesamiento Proteico-Postraduccional , Empalme del ARN , Receptores Androgénicos/aislamiento & purificación , Valores de Referencia , Secuencias Reguladoras de Ácidos Nucleicos , Mapeo Restrictivo , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda