Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Mol Life Sci ; 81(1): 75, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38315424

RESUMEN

Autosomal dominant variants in LRP10 have been identified in patients with Lewy body diseases (LBDs), including Parkinson's disease (PD), Parkinson's disease-dementia (PDD), and dementia with Lewy bodies (DLB). Nevertheless, there is little mechanistic insight into the role of LRP10 in disease pathogenesis. In the brains of control individuals, LRP10 is typically expressed in non-neuronal cells like astrocytes and neurovasculature, but in idiopathic and genetic cases of PD, PDD, and DLB, it is also present in α-synuclein-positive neuronal Lewy bodies. These observations raise the questions of what leads to the accumulation of LRP10 in Lewy bodies and whether a possible interaction between LRP10 and α-synuclein plays a role in disease pathogenesis. Here, we demonstrate that wild-type LRP10 is secreted via extracellular vesicles (EVs) and can be internalised via clathrin-dependent endocytosis. Additionally, we show that LRP10 secretion is highly sensitive to autophagy inhibition, which induces the formation of atypical LRP10 vesicular structures in neurons in human-induced pluripotent stem cells (iPSC)-derived brain organoids. Furthermore, we show that LRP10 overexpression leads to a strong induction of monomeric α-synuclein secretion, together with time-dependent, stress-sensitive changes in intracellular α-synuclein levels. Interestingly, patient-derived astrocytes carrying the c.1424 + 5G > A LRP10 variant secrete aberrant high-molecular-weight species of LRP10 in EV-free media fractions. Finally, we show that this truncated patient-derived LRP10 protein species (LRP10splice) binds to wild-type LRP10, reduces LRP10 wild-type levels, and antagonises the effect of LRP10 on α-synuclein levels and distribution. Together, this work provides initial evidence for a possible functional role of LRP10 in LBDs by modulating intra- and extracellular α-synuclein levels, and pathogenic mechanisms linked to the disease-associated c.1424 + 5G > A LRP10 variant, pointing towards potentially important disease mechanisms in LBDs.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/patología , Enfermedad por Cuerpos de Lewy/genética , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Cuerpos de Lewy/metabolismo , Encéfalo/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo
2.
Front Oncol ; 12: 877613, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35769712

RESUMEN

Treatment of prostate cancer (PCa) has changed considerably in the last decade due to the introduction of novel androgen receptor (AR)-targeted agents (ARTAs) for patients progressing on androgen deprivation therapy (ADT). Preclinical research however still relies heavily on AR-negative cell line models. In order to investigate potential differences in castration-resistant PCa (CRPC) growth, we set out to create a comprehensive panel of ARTA-progressive models from 4 androgen-responsive AR wild-type PCa cell lines and analyzed its androgen response as opposed to its ADT-progressive counterparts. Parallel cultures of VCaP, DuCaP, PC346C, and LAPC4 were established in their respective culture media with steroid-stripped fetal calf serum (FCS) [dextran-coated charcoal-stripped FCS (DCC)] without androgen (ADT) or in DCC plus 1 µM of the ARTAs bicalutamide, OH-flutamide, or RD162 (an enzalutamide/apalutamide analog). Cell growth was monitored and compared to those of parental cell lines. Short-term androgen response was measured using cell proliferation 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. qRT-PCR was performed to assess the mRNA expression of markers for AR signaling, steroidogenesis, glucocorticoid receptor (GR) signaling, epithelial-mesenchymal transition (EMT), and WNT signaling. Out of 35 parallel cultures per cell line, a total of 24, 15, 34, and 16 CRPC sublines emerged for VCaP, DuCaP, PC346C, and LAPC4, respectively. The addition of bicalutamide or OH-flutamide significantly increased CRPC growth compared to ADT or RD162. VCaP, DuCaP, and PC346C CRPC clones retained an AR-responsive phenotype. The expression of AR and subsequent androgen response were completely lost in all LAPC4 CRPC lines. Markers for EMT and WNT signaling were found to be elevated in the resilient PC346C model and CRPC derivatives of VCaP, DuCaP, and LAPC4. Although the resistant phenotype is pluriform between models, it seems consistent within models, regardless of type of ARTA. These data suggest that the progression to and the phenotype of the CRPC state might already be determined early in carcinogenesis.

3.
Clin Transl Gastroenterol ; 13(3): e00465, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35363220

RESUMEN

INTRODUCTION: Extracellular vesicles (EVs) and their cargo may provide promising biomarkers for the early detection of pancreatic ductal adenocarcinoma (PDAC). Although blood-borne EVs are most frequently studied as cancer biomarkers, pancreatic juice (PJ) may represent a better biomarker source because it is in close contact with the ductal cells from which PDAC arises. It is, as yet, unknown whether PDAC results in a distinct type or increased number of particles in PJ and whether this has diagnostic value. METHODS: Secretin-stimulated PJ was collected from the duodenum of 54 cases and 117 nonmalignant controls under surveillance for PDAC. Serum was available for a subset of these individuals. The vesicular composition of these biofluids was analyzed with nanoparticle tracking analysis. RESULTS: The concentration of EVs did not differ between controls and PDAC cases. However, a higher number of large vesicles were found in PJ (but not serum) for patients with PDAC compared with controls. DISCUSSION: The composition of isolated EVs from PJ, but not serum, is altered in patients with PDAC. This suggests that PJ may carry disease-specific markers not present in serum and provides a valuable biomarker source for PDAC diagnosis. The nature of the larger particles in EV isolates from PJ of PDAC cases requires further investigation.


Asunto(s)
Carcinoma Ductal Pancreático , Vesículas Extracelulares , Neoplasias Pancreáticas , Biomarcadores de Tumor , Carcinoma Ductal Pancreático/patología , Vesículas Extracelulares/patología , Humanos , Jugo Pancreático , Neoplasias Pancreáticas/patología
4.
Prostate ; 73(15): 1636-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23996639

RESUMEN

BACKGROUND: Despite an initial response to hormonal therapy, patients with advanced prostate cancer (PC) almost always progress to castration-resistant disease (CRPC). Although serum testosterone (T) is reduced by androgen deprivation therapy, intratumoral T levels in CRPC are comparable to those in prostate tissue of eugonadal men. These levels could originate from intratumoral conversion of adrenal androgens and/or from de novo steroid synthesis. However, the relative contribution of de novo steroidogenesis to AR-driven cell growth is unknown. METHODS: The relative contribution of androgen biosynthetic pathways to activate androgen receptor (AR)-regulated cell growth and expression of PSA, FKBP5, and TMPRSS2 was studied at physiologically relevant levels of adrenal androgen precursors and intermediates of de novo androgen biosynthesis in human prostate cancer cell lines, PC346C, VCaP, and LNCaP. RESULTS: In PC346C and VCaP, responses to pregnenolone and progesterone were absent or minimal, while large effects of adrenal androgen precursors were found. VCaP CRPC clones overexpressing CYP17A1 did not acquire an increased ability to use pregnenolone or progesterone to activate AR. In contrast, all precursors stimulated growth and gene expression in LNCaP cells, presumably resulting from the mutated AR in these cells. CONCLUSIONS: Our data indicate that at physiological levels of T precursors PC cells can generally convert adrenal androgens, while de novo steroidogenesis is not generally possible in PC cells and is not able to support AR transactivation and PC growth.


Asunto(s)
Andrógenos/biosíntesis , Proliferación Celular , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo
5.
Endocrinology ; 153(12): 5726-34, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23024260

RESUMEN

Local androgen synthesis in prostate cancer (PC) may contribute to the development of castration-resistant PC (CRPC), but pathways controlling intratumoral steroidogenic enzyme expression in PC are unknown. We investigated the effects of activin, a factor involved in the regulation of PC growth and steroidogenic enzyme expression in other steroidogenic tissues, on intratumoral steroidogenesis in PC. Activin A effects and regulation of the activin-signaling pathway molecules were studied in the PC cell lines LNCaP, VCaP, and PC-3 and in 13 individual PC xenograft models. Also, expression levels of inhibin ßA- and ßB-subunits (INHBA and INHBB) and of the activin antagonist follistatin were quantitated in patient PC tissues. Activin A induced the expression and enzyme activity of 17ß-hydroxysteroid dehydrogenase enzyme AKR1C3 in LNCaP and VCaP cells. Inhibition of endogenous activin A action in the PC-3 cell line decreased AKR1C3 levels and consequently testosterone synthesis. In return, androgens suppressed INHBA expression in both VCaP cells and the PC xenograft models. The antiproliferative effects of activin A were opposed by physiological concentrations of androstenedione in LNCaP cells. In patient PC tissues, expression levels of INHBA were increased in CRPC samples and correlated with AKR1C3 levels. Moreover, a high ratio of activin subunits to follistatin was associated with a worse metastasis-free survival in patients. In conclusion, activin A is controlled by androgens in PC models and regulates local androgen production. Activin A thus seems to mediate (residual) intratumoral androgen levels and could form a novel therapeutic target in CRPC.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/biosíntesis , Activinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Neoplasias de la Próstata/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Andrógenos/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Transducción de Señal , Testosterona/metabolismo
6.
Cancer Immunol Immunother ; 61(6): 905-10, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22113713

RESUMEN

BACKGROUND: Given the fact that prostate cancer incidence will increase in the coming years, new prognostic biomarkers are needed with regard to the biological aggressiveness of the prostate cancer diagnosed. Since cytokines have been associated with the biology of cancer and its prognosis, we determined whether transforming growth factor beta 1 (TGFß1), interleukin-7 (IL-7) receptor and IL-7 levels add additional prognostic information with regard to prostate cancer-specific survival. MATERIALS AND METHODS: Retrospective survival analysis of forty-four prostate cancer patients, that underwent radical prostatectomy, was performed (1989-2001). Age, Gleason score and pre-treatment PSA levels were collected. IL-7, IL-7 receptor and TGFß1 levels in prostate cancer tissue were determined by quantitative real-time RT-PCR and their additional prognostic value analyzed with regard to prostate cancer survival. Hazard ratios and their confidence intervals were estimated, and Akaike's information criterion was calculated for model comparison. RESULTS: The predictive ability of a model for prostate cancer survival more than doubled when TGFß1 and IL-7 were added to a model containing only the Gleason score and pre-treatment PSA (AIC: 18.1 and AIC: 6.5, respectively). CONCLUSION: IL-7 and TGFß1 are promising markers to indicate those at risk for poor prostate cancer survival. This additional information may be of interest with regard to the biological aggressiveness of the diagnosed prostate cancer, especially for those patients screened for prostate cancer and their considered therapy.


Asunto(s)
Interleucina-7/inmunología , Neoplasias de la Próstata/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Progresión de la Enfermedad , Humanos , Interleucina-7/genética , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Prostatectomía , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/cirugía , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/inmunología , Estudios Retrospectivos , Análisis de Supervivencia , Factor de Crecimiento Transformador beta1/genética
7.
PLoS One ; 6(8): e23144, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829708

RESUMEN

BACKGROUND: Prostate epithelial cells depend on androgens for survival and function. In (early) prostate cancer (PCa) androgens also regulate tumor growth, which is exploited by hormonal therapies in metastatic disease. The aim of the present study was to characterize the androgen receptor (AR) response in hormonal therapy-resistant PC346 cells and identify potential disease markers. METHODOLOGY/PRINCIPAL FINDINGS: Human 19K oligoarrays were used to establish the androgen-regulated expression profile of androgen-responsive PC346C cells and its derivative therapy-resistant sublines: PC346DCC (vestigial AR levels), PC346Flu1 (AR overexpression) and PC346Flu2 (T877A AR mutation). In total, 107 transcripts were differentially-expressed in PC346C and derivatives after R1881 or hydroxyflutamide stimulations. The AR-regulated expression profiles reflected the AR modifications of respective therapy-resistant sublines: AR overexpression resulted in stronger and broader transcriptional response to R1881 stimulation, AR down-regulation correlated with deficient response of AR-target genes and the T877A mutation resulted in transcriptional response to both R1881 and hydroxyflutamide. This AR-target signature was linked to multiple publicly available cell line and tumor derived PCa databases, revealing that distinct functional clusters were differentially modulated during PCa progression. Differentiation and secretory functions were up-regulated in primary PCa but repressed in metastasis, whereas proliferation, cytoskeletal remodeling and adhesion were overexpressed in metastasis. Finally, the androgen-regulated genes ENDOD1, MCCC2 and ACSL3 were selected as potential disease markers for RT-PCR quantification in a distinct set of human prostate specimens. ENDOD1 and ACSL3 showed down-regulation in high-grade and metastatic PCa, while MCCC2 was overexpressed in low-grade PCa. CONCLUSIONS/SIGNIFICANCE: AR modifications altered the transcriptional response to (anti)androgens in therapy-resistant cells. Furthermore, selective down-regulation of genes involved in differentiation and up-regulation of genes promoting proliferation and invasion suggest a disturbed balance between the growth and differentiation functions of the AR pathway during PCa progression. These findings may have implications in the current treatment and development of novel therapeutical approaches for metastatic PCa.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Receptores Androgénicos/metabolismo , Transducción de Señal , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Resistencia a Antineoplásicos , Flutamida/análogos & derivados , Flutamida/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Masculino , Metribolona/uso terapéutico , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
PLoS One ; 5(10): e13500, 2010 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-20976069

RESUMEN

BACKGROUND: Prostate cancer is initially dependent on androgens for survival and growth, making hormonal therapy the cornerstone treatment for late-stage tumors. However, despite initial remission, the cancer will inevitably recur. The present study was designed to investigate how androgen-dependent prostate cancer cells eventually survive and resume growth under androgen-deprived and antiandrogen supplemented conditions. As model system, we used the androgen-responsive PC346C cell line and its therapy-resistant sublines: PC346DCC, PC346Flu1 and PC346Flu2. METHODOLOGY/PRINCIPAL FINDINGS: Microarray technology was used to analyze differences in gene expression between the androgen-responsive and therapy-resistant PC346 cell lines. Microarray analysis revealed 487 transcripts differentially-expressed between the androgen-responsive and the therapy-resistant cell lines. Most of these genes were common to all three therapy-resistant sublines and only a minority (∼5%) was androgen-regulated. Pathway analysis revealed enrichment in functions involving cellular movement, cell growth and cell death, as well as association with cancer and reproductive system disease. PC346DCC expressed residual levels of androgen receptor (AR) and showed significant down-regulation of androgen-regulated genes (p-value = 10(-7)). Up-regulation of VAV3 and TWIST1 oncogenes and repression of the DKK3 tumor-suppressor was observed in PC346DCC, suggesting a potential AR bypass mechanism. Subsequent validation of these three genes in patient samples confirmed that expression was deregulated during prostate cancer progression. CONCLUSIONS/SIGNIFICANCE: Therapy-resistant growth may result from adaptations in the AR pathway, but androgen-independence may also be achieved by alternative survival mechanisms. Here we identified TWIST1, VAV3 and DKK3 as potential players in the bypassing of the AR pathway, making them good candidates as biomarkers and novel therapeutical targets.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Progresión de la Enfermedad , Regulación hacia Abajo , Perfilación de la Expresión Génica , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Cancer Res ; 70(3): 1256-64, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20086173

RESUMEN

Androgen-deprivation therapy for prostate cancer (PC) eventually leads to castration-resistant PC (CRPC). Intratumoral androgen production might contribute to tumor progression despite suppressed serum androgen concentrations. In the present study, we investigated whether PC or CRPC tissue may be capable of intratumoral androgen synthesis. Steroidogenic enzyme mRNAs were quantified in hormonally manipulated human PC cell lines and xenografts as well as in human samples of normal prostate, locally confined and advanced PC, local nonmetastatic CRPC, and lymph node metastases. Overall, the majority of samples showed low or absent mRNA expression of steroidogenic enzymes required for de novo steroid synthesis. Simultaneous but low expression of the enzymes CYP17A1 and HSD3B1, essential for the synthesis of androgens from pregnenolone, could be detected in 19 of 88 patient samples. Of 19 CRPC tissues examined, only 5 samples expressed both enzymes. Enzymes that convert androstenedione to testosterone (AKR1C3) and testosterone to dihydrotestosterone (DHT; SRD5A1) were abundantly expressed. AKR1C3 expression was negatively regulated by androgens in the experimental models and was increased in CRPC samples. Expression of SRD5A1 was upregulated in locally advanced cancer, CRPC, and lymph node metastases. We concluded that intratumoral steroid biosynthesis contributes less than circulating adrenal androgens, implying that blocking androgen production and its intraprostatic conversion into DHT, such as via CYP17A1 inhibition, may represent favorable therapeutic options in patients with CRPC.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Hidroxiprostaglandina Deshidrogenasas/genética , Progesterona Reductasa/genética , Neoplasias de la Próstata/genética , Esteroide 17-alfa-Hidroxilasa/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Anciano , Anciano de 80 o más Años , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Andrógenos/metabolismo , Androstenodiona/metabolismo , Animales , Línea Celular Tumoral , Dihidrotestosterona/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Orquiectomía , Pregnenolona/metabolismo , Progesterona Reductasa/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esteroide 17-alfa-Hidroxilasa/metabolismo , Testosterona/metabolismo , Trasplante Heterólogo
10.
BJU Int ; 103(11): 1574-80, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19220249

RESUMEN

OBJECTIVE To assess the expression of forkhead transcription factors (FOX) in normal prostate and prostate diseases, as since the first FOX was identified, its family members have been implicated in a variety of cellular processes, including embryonic development and disease. MATERIAL AND METHODS We analysed a set of 12 different FOX genes by quantitative reverse transcription-polymerase chain reaction in prostate zones, prostate cancer, lymph node metastases, benign prostatic hyperplasia (BPH), xenografts and several prostate cell lines. RESULTS There were striking differences among the expression of various FOX family members; most prominent were the high expression of FOXF1 and FOXF2 in the normal prostate transition zone and BPH, and their decreased expression in prostate cancer. Interestingly, although the FOXF genes are stroma-specific, some of the androgen-independent prostate cancer xenografts uniquely express these two genes. FOXD1 and FOXD2 were more highly expressed in prostate cancer and lymph node metastases. FOXA1 and FOXC1 have an opposite expression pattern for androgen-dependent growth of prostate cancer cell lines and xenografts. CONCLUSIONS Various members of the FOX family are differentially expressed in the zones of the normal prostate and in benign and malignant outgrowths. The expression profiles of FOXF1 and FOXF2 suggest a role in epithelial to mesenchymal transition, while FOXA1 and FOXC1 expression is linked to androgen-associated growth status of cancer.


Asunto(s)
Factores de Transcripción Forkhead/genética , Expresión Génica/genética , Próstata/metabolismo , Hiperplasia Prostática/genética , Neoplasias de la Próstata/genética , Anciano , Progresión de la Enfermedad , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Próstata/patología , Prostatectomía , Hiperplasia Prostática/patología , Hiperplasia Prostática/cirugía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/genética
11.
Cancer Res ; 68(18): 7541-9, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18794142

RESUMEN

In this study, we describe the properties of novel ETV1 fusion genes, encoding N-truncated ETV1 (dETV1), and of full-length ETV1, overexpressed in clinical prostate cancer. We detected overexpression of novel ETV1 fusion genes or of full-length ETV1 in 10% of prostate cancers. Novel ETV1 fusion partners included FOXP1, an EST (EST14), and an endogenous retroviral repeat sequence (HERVK17). Like TMPRSS2, EST14 and HERVK17 were prostate-specific and androgen-regulated expressed. This unique expression pattern of most ETV1 fusion partners seems an important determinant in prostate cancer development. In transient reporter assays, full-length ETV1 was a strong transactivator, whereas dETV1 was not. However, several of the biological properties of dETV1 and full-length ETV1 were identical. On stable overexpression, both induced migration and invasion of immortalized nontumorigenic PNT2C2 prostate epithelial cells. In contrast to dETV1, full-length ETV1 also induced anchorage-independent growth of these cells. PNT2C2 cells stably transfected with dETV1 or full-length ETV1 expression constructs showed small differences in induced expression of target genes. Many genes involved in tumor invasion/metastasis, including uPA/uPAR and MMPs, were up-regulated in both cell types. Integrin beta3 (ITGB3) was clearly up-regulated by full-length ETV1 but much less by dETV1. Based on the present data and on previous findings, a novel concept of the role of dETV1 and of full-length ETV1 overexpression in prostate cancer is proposed.


Asunto(s)
Proteínas de Unión al ADN/genética , Fusión Génica , Neoplasias de la Próstata/genética , Factores de Transcripción/genética , Animales , Adhesión Celular/genética , Movimiento Celular/genética , Proteínas de Unión al ADN/biosíntesis , Etiquetas de Secuencia Expresada , Factores de Transcripción Forkhead/genética , Expresión Génica , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/biosíntesis
12.
Cancer Res ; 68(9): 3094-8, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18451133

RESUMEN

Recently, fusion of ERG to the androgen-regulated, prostate-specific TMPRSS2 gene has been identified as the most frequent genetic alteration in prostate cancer. At low frequency, TMPRSS2-ETV1 and TMPRSS2-ETV4 fusion genes have been described. In this study, we report two novel ETV4 fusion genes in prostate cancer: KLK2-ETV4 and CANT1-ETV4. Both gene fusions have important unique aspects. KLK2 is a well-established androgen-induced and prostate-specific gene. Fusion of KLK2 to ETV4 results in the generation of an additional ETV4 exon, denoted exon 4a. This novel exon delivers an ATG for the longest open reading frame, in this way avoiding translation start in KLK2 exon 1. Although wild-type CANT1 has two alternative first exons (exons 1 and 1a), only exon 1a was detected in CANT1-ETV4 fusion transcripts. We show that CANT1 transcripts starting at exon 1a have an androgen-induced and prostate-specific expression pattern, whereas CANT1 transcripts starting at exon 1 are not prostate specific. So, the two novel ETV4 fusion partners possess as predominant common characteristics androgen-induction and prostate-specific expression.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Andrógenos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas/genética , Proteínas E1A de Adenovirus/metabolismo , Animales , Secuencia de Bases , Humanos , Masculino , Ratones , Ratones Desnudos , Modelos Biológicos , Datos de Secuencia Molecular , Neoplasias Hormono-Dependientes/genética , Nucleotidasas/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Especificidad de Órganos/genética , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Distribución Tisular , Calicreínas de Tejido/genética , Calicreínas de Tejido/metabolismo
13.
Cancer Res ; 66(10): 5012-20, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707422

RESUMEN

The present work focused on the potential involvement of selective adaptations of the androgen receptor pathway in the initiation and progression of prostate cancer. We defined the androgen receptor pathway by selecting 200 genes that were androgen responsive in prostate cancer cell lines and/or xenografts. This androgen receptor pathway gene signature was then used for profiling prostate cancer xenografts and patient-derived samples. Approximately half of the androgen receptor pathway genes were up-regulated in well-differentiated prostate cancer compared with normal prostate. Functionally distinct parts of the androgen receptor pathway were specifically down-regulated in high-grade cancers. Unexpectedly, metastases have down-regulated the vast majority of androgen receptor pathway genes. The significance of this progressive down-regulation of androgen receptor pathway genes was shown for a few androgen receptor-regulated genes. Lower mRNA expression of HERPUD1, STK39, DHCR24, and SOCS2 in primary prostate tumors was correlated with a higher incidence of metastases after radical prostatectomy. HERPUD1 mRNA expression predicted the occurrence of metastases almost perfectly. In vitro experiments showed that overexpression of the stress response gene HERPUD1 rapidly induces apoptosis. Based on the functions of the genes within the distinct subsets, we propose the following model. Enhanced androgen receptor activity is involved in the early stages of prostate cancer. In well-differentiated prostate cancer, the androgen receptor activates growth-promoting as well as growth-inhibiting and cell differentiation genes resulting in a low growth rate. The progression from low-grade to high-grade prostate carcinoma and metastases is mediated by a selective down-regulation of the androgen receptor target genes that inhibit proliferation, induce differentiation, or mediate apoptosis.


Asunto(s)
Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Metribolona/farmacología , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias Hormono-Dependientes/metabolismo , Orquiectomía , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...