Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Nucleic Acids Res ; 48(10): 5366-5382, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32324216

RESUMEN

Resistance to androgen receptor (AR) targeting therapeutics in prostate cancer (PC) is a significant clinical problem. Mechanisms by which this is accomplished include AR amplification and expression of AR splice variants, demonstrating that AR remains a key therapeutic target in advanced disease. For the first time we show that IKBKE drives AR signalling in advanced PC. Significant inhibition of AR regulated gene expression was observed upon siRNA-mediated IKBKE depletion or pharmacological inhibition due to inhibited AR gene expression in multiple cell line models including a LNCaP derivative cell line resistant to the anti-androgen, enzalutamide (LNCaP-EnzR). Phenotypically, this resulted in significant inhibition of proliferation, migration and colony forming ability suggesting that targeting IKBKE could circumvent resistance to AR targeting therapies. Indeed, pharmacological inhibition in the CWR22Rv1 xenograft mouse model reduced tumour size and enhanced survival. Critically, this was validated in patient-derived explants where enzymatic inactivation of IKBKE reduced cell proliferation and AR expression. Mechanistically, we provide evidence that IKBKE regulates AR levels via Hippo pathway inhibition to reduce c-MYC levels at cis-regulatory elements within the AR gene. Thus, IKBKE is a therapeutic target in advanced PC suggesting repurposing of clinically tested IKBKE inhibitors could be beneficial to castrate resistant PC patients.


Asunto(s)
Quinasa I-kappa B/fisiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Androgénicos/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Masculino , Ratones Desnudos , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas Señalizadoras YAP
2.
Nucleic Acids Res ; 47(11): 5634-5647, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31006810

RESUMEN

Resistance to androgen receptor (AR)-targeted therapies in prostate cancer (PC) is a major clinical problem. A key mechanism of treatment resistance in advanced PC is the generation of alternatively spliced forms of the AR termed AR variants (AR-Vs) that are refractory to targeted agents and drive tumour progression. Our understanding of how AR-Vs function is limited due to difficulties in distinguishing their discriminate activities from full-length AR (FL-AR). Here we report the development of a novel CRISPR-derived cell line which is a derivative of CWR22Rv1 cells, called CWR22Rv1-AR-EK, that has lost expression of FL-AR, but retains all endogenous AR-Vs. From this, we show that AR-Vs act unhindered by loss of FL-AR to drive cell growth and expression of androgenic genes. Global transcriptomics demonstrate that AR-Vs drive expression of a cohort of DNA damage response genes and depletion of AR-Vs sensitises cells to ionising radiation. Moreover, we demonstrate that AR-Vs interact with PARP1 and PARP2 and are dependent upon their catalytic function for transcriptional activation. Importantly, PARP blockade compromises expression of AR-V-target genes and reduces growth of CRPC cell lines suggesting a synthetic lethality relationship between AR-Vs and PARP, advocating the use of PARP inhibitors in AR-V positive PC.


Asunto(s)
Sistemas CRISPR-Cas , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/genética , Algoritmos , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , Reparación del ADN , Ensayos de Selección de Medicamentos Antitumorales , Técnicas Genéticas , Humanos , Lentivirus , Masculino , Receptores Androgénicos/biosíntesis , Análisis de Secuencia de ARN , Transcriptoma
3.
J Cell Sci ; 131(1)2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29222115

RESUMEN

Chronic kidney disease (CKD) is a global socioeconomic problem. It is characterised by the presence of differentiated myofibroblasts, which cause tissue fibrosis in response to TGFB1, leading to renal failure. Here, we define a novel interaction between the SET9 lysine methyltransferase (also known as SETD7) and SMAD3, the principal mediator of TGFB1 signalling in myofibroblasts. We show that SET9-deficient fibroblasts exhibit globally altered gene expression profiles in response to TGFB1, whilst overexpression of SET9 enhances SMAD3 transcriptional activity. We also show that SET9 facilitates nuclear import of SMAD3 and controls SMAD3 protein degradation via ubiquitylation. On a cellular level, we demonstrate that SET9 is broadly required for the effects of TGFB1 in diseased primary renal fibroblasts; SET9 promotes fibroblast migration into wounds, expression of extracellular matrix proteins, collagen contractility and myofibroblast differentiation. Finally, we demonstrate that SET9 is recruited to the α-smooth muscle actin gene in response to TGFB1, providing a mechanism by which SET9 regulates myofibroblast contractility and differentiation. Together with previous studies, we make the case for SET9 inhibition in the treatment of progressive CKD.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Miofibroblastos/metabolismo , Transducción de Señal/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Animales , Diferenciación Celular , Línea Celular , Movimiento Celular , Células Cultivadas , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibrosis , Regulación de la Expresión Génica , Humanos , Riñón/citología , Ratones , Ratones Noqueados , Miofibroblastos/citología
4.
Nucleic Acids Res ; 45(4): 1793-1804, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-27903893

RESUMEN

The androgen receptor (AR) is the main driver of prostate cancer (PC) development and progression, and the primary therapeutic target in PC. To date, two functional ubiquitination sites have been identified on AR, both located in its C-terminal ligand binding domain (LBD). Recent reports highlight the emergence of AR splice variants lacking the LBD that can arise during disease progression and contribute to castrate resistance. Here, we report a novel N-terminal ubiquitination site at lysine 311. Ubiquitination of this site plays a role in AR stability and is critical for its transcriptional activity. Inactivation of this site causes AR to accumulate on chromatin and inactivates its transcriptional function as a consequence of inability to bind to p300. Additionally, mutation at lysine 311 affects cellular transcriptome altering the expression of genes involved in chromatin organization, signaling, adhesion, motility, development and metabolism. Even though this site is present in clinically relevant AR-variants it can only be ubiquitinated in cells when AR retains LBD suggesting a role for AR C-terminus in E2/E3 substrate recognition. We report that as a consequence AR variants lacking the LBD cannot be ubiquitinated in the cellular environment and their protein turnover must be regulated via an alternate pathway.


Asunto(s)
Receptores Androgénicos/metabolismo , Activación Transcripcional , Ubiquitinación , Aminoácidos/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Análisis por Conglomerados , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Mutación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteoma , Proteómica/métodos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/genética , Transcripción Genética , Transcriptoma
6.
Nucleic Acids Res ; 43(1): 196-207, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25488809

RESUMEN

Endocrine therapy has successfully been used to treat estrogen receptor (ER)-positive breast cancer, but this invariably fails with cancers becoming refractory to treatment. Emerging evidence has suggested that fluctuations in ER co-regulatory protein expression may facilitate resistance to therapy and be involved in breast cancer progression. To date, a small number of enzymes that control methylation status of histones have been identified as co-regulators of ER signalling. We have identified the histone H3 lysine 9 mono- and di-methyl demethylase enzyme KDM3A as a positive regulator of ER activity. Here, we demonstrate that depletion of KDM3A by RNAi abrogates the recruitment of the ER to cis-regulatory elements within target gene promoters, thereby inhibiting estrogen-induced gene expression changes. Global gene expression analysis of KDM3A-depleted cells identified gene clusters associated with cell growth. Consistent with this, we show that knockdown of KDM3A reduces ER-positive cell proliferation and demonstrate that KDM3A is required for growth in a model of endocrine therapy-resistant disease. Crucially, we show that KDM3A catalytic activity is required for both ER-target gene expression and cell growth, demonstrating that developing compounds which target demethylase enzymatic activity may be efficacious in treating both ER-positive and endocrine therapy-resistant disease.


Asunto(s)
Neoplasias de la Mama/enzimología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Histona Demetilasas con Dominio de Jumonji/fisiología , Células MCF-7 , Elementos de Respuesta , Transducción de Señal
7.
J Pathol ; 235(4): 581-92, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25408555

RESUMEN

Biomechanical strain imposed by age-related thickening of the basal lamina and augmented tissue stiffness in the prostate gland coincides with increased cancer risk. Here we hypothesized that the structural alterations in the basal lamina associated with age can induce mechanotransduction pathways in prostate epithelial cells (PECs) to promote invasiveness and cancer progression. To demonstrate this, we developed a 3D model of PEC acini in which thickening and stiffening of basal lamina matrix was induced by advanced glycation end-product (AGE)-dependent non-enzymatic crosslinking of its major components, collagen IV and laminin. We used this model to demonstrate that antibody targeted blockade of CTLD2, the second of eight C-type lectin-like domains in Endo180 (CD280, CLEC13E, KIAA0709, MRC2, TEM9, uPARAP) that can recognize glycosylated collagens, reversed actinomyosin-based contractility [myosin-light chain-2 (MLC2) phosphorylation], loss of cell polarity, loss of cell-cell junctions, luminal infiltration and basal invasion induced by AGE-modified basal lamina matrix in PEC acini. Our in vitro results were concordant with luminal occlusion of acini in the prostate glands of adult Endo180(Δ) (Ex2-6/) (Δ) (Ex2-6) mice, with constitutively exposed CTLD2 and decreased survival of men with early (non-invasive) prostate cancer with high epithelial Endo180 expression and levels of AGE. These findings indicate that AGE-dependent modification of the basal lamina induces invasive behaviour in non-transformed PECs via a molecular mechanism linked to cancer progression. This study provides a rationale for targeting CTLD2 in Endo180 in prostate cancer and other pathologies in which increased basal lamina thickness and tissue stiffness are driving factors. © 2014 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Membrana Basal/metabolismo , Movimiento Celular , Células Epiteliales/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Lectinas de Unión a Manosa/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Mitogénicos/metabolismo , Animales , Membrana Basal/patología , Línea Celular Tumoral , Supervivencia Celular , Elasticidad , Humanos , Estimación de Kaplan-Meier , Lectinas Tipo C/metabolismo , Masculino , Mecanotransducción Celular , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones Noqueados , Invasividad Neoplásica , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Estructura Terciaria de Proteína , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Factores de Tiempo
8.
Nucleic Acids Res ; 41(14): 6892-904, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23723241

RESUMEN

The importance of the estrogen receptor (ER) in breast cancer (BCa) development makes it a prominent target for therapy. Current treatments, however, have limited effectiveness, and hence the definition of new therapeutic targets is vital. The ER is a member of the nuclear hormone receptor superfamily of transcription factors that requires co-regulator proteins for complete regulation. Emerging evidence has implicated a small number of histone methyltransferase (HMT) and histone demethylase (HDM) enzymes as regulators of ER signalling, including the histone H3 lysine 9 tri-/di-methyl HDM enzyme KDM4B. Two recent independent reports have demonstrated that KDM4B is required for ER-mediated transcription and depletion of the enzyme attenuates BCa growth in vitro and in vivo. Here we show that KDM4B has an overarching regulatory role in the ER signalling cascade by controlling expression of the ER and FOXA1 genes, two critical components for maintenance of the estrogen-dependent phenotype. KDM4B interacts with the transcription factor GATA-3 in BCa cell lines and directly co-activates GATA-3 activity in reporter-based experiments. Moreover, we reveal that KDM4B recruitment and demethylation of repressive H3K9me3 marks within upstream regulatory regions of the ER gene permits binding of GATA-3 to drive receptor expression. Ultimately, our findings confirm the importance of KDM4B within the ER signalling cascade and as a potential therapeutic target for BCa treatment.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Línea Celular , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/fisiología , Células MCF-7 , Receptores de Estrógenos/genética , Elementos Reguladores de la Transcripción
9.
Nucleic Acids Res ; 41(8): 4433-46, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23435229

RESUMEN

The androgen receptor (AR) is a key molecule involved in prostate cancer (PC) development and progression. Post-translational modification of the AR by co-regulator proteins can modulate its transcriptional activity. To identify which demethylases might be involved in AR regulation, an siRNA screen was performed to reveal that the demethylase, KDM4B, may be an important co-regulator protein. KDM4B enzymatic activity is required to enhance AR transcriptional activity; however, independently of this activity, KDM4B can enhance AR protein stability via inhibition of AR ubiquitination. Importantly, knockdown of KDM4B in multiple cell lines results in almost complete depletion of AR protein levels. For the first time, we have identified KDM4B to be an androgen-regulated demethylase enzyme, which can influence AR transcriptional activity not only via demethylation activity but also via modulation of ubiquitination. Together, these findings demonstrate the close functional relationship between AR and KDM4B, which work together to amplify the androgen response. Furthermore, KDM4B expression in clinical PC specimens positively correlates with increasing cancer grade (P < 0.001). Consequently, KDM4B is a viable therapeutic target in PC.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/metabolismo , Receptores Androgénicos/metabolismo , Andrógenos/farmacología , Animales , Línea Celular , Proliferación Celular , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Masculino , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Transducción de Señal , Transcripción Genética , Ubiquitinación
10.
J Biol Chem ; 288(45): 32641-32650, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24056413

RESUMEN

The androgen receptor (AR), a member of the nuclear receptor family, is a transcription factor involved in prostate cell growth, homeostasis, and transformation. AR is a key protein in growth and development of both normal and malignant prostate, making it a common therapeutic target in prostate cancer. AR is regulated by an interplay of multiple post-translational modifications including ubiquitination. We and others have shown that the AR is ubiquitinated by a number of E3 ubiquitin ligases, including MDM2, CHIP, and NEDD4, which can result in its proteosomal degradation or enhanced transcriptional activity. As ubiquitination of AR causes a change in AR activity or stability and impacts both survival and growth of prostate cancer cells, deubiquitination of these sites has an equally important role. Hence, deubiquitinating enzymes could offer novel therapeutic targets. We performed an siRNA screen to identify deubiquitinating enzymes that regulate AR; in that screen ubiquitin-specific protease 12 (Usp12) was identified as a novel positive regulator of AR. Usp12 is a poorly characterized protein with few known functions and requires the interaction with two cofactors, Uaf-1 and WDR20, for its enzymatic activity. In this report we demonstrate that Usp12, in complex with Uaf-1 and WDR20, deubiquitinates the AR to enhance receptor stability and transcriptional activity. Our data show that Usp12 acts in a pro-proliferative manner by stabilizing AR and enhancing its cellular function.


Asunto(s)
Proliferación Celular , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Células COS , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Masculino , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/genética , Estabilidad Proteica , Receptores Androgénicos/genética , Ubiquitina Tiolesterasa/genética , Ubiquitina-Proteína Ligasas/genética
11.
Commun Biol ; 7(1): 276, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38448753

RESUMEN

Immune checkpoint blockade has yet to produce robust anti-cancer responses for prostate cancer. Sialyltransferases have been shown across several solid tumours, including breast, melanoma, colorectal and prostate to promote immune suppression by synthesising sialoglycans, which act as ligands for Siglec receptors. We report that ST3 beta-galactoside alpha-2,3-sialyltransferase 1 (ST3Gal1) levels negatively correlate with androgen signalling in prostate tumours. We demonstrate that ST3Gal1 plays an important role in modulating tumour immune evasion through the synthesises of sialoglycans with the capacity to engage the Siglec-7 and Siglec-9 immunoreceptors preventing immune clearance of cancer cells. Here, we provide evidence of the expression of Siglec-7/9 ligands and their respective immunoreceptors in prostate tumours. These interactions can be modulated by enzalutamide and may maintain immune suppression in enzalutamide treated tumours. We conclude that the activity of ST3Gal1 is critical to prostate cancer anti-tumour immunity and provide rationale for the use of glyco-immune checkpoint targeting therapies in advanced prostate cancer.


Asunto(s)
Feniltiohidantoína , Neoplasias de la Próstata , beta-Galactosida alfa-2,3-Sialiltransferasa , Masculino , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Benzamidas/farmacología , Nitrilos , Ligandos
12.
Nucleic Acids Res ; 39(4): 1266-79, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20959290

RESUMEN

The androgen receptor (AR) is a member of the nuclear hormone receptor family of transcription factors that plays a critical role in regulating expression of genes involved in prostate development and transformation. Upon hormone binding, the AR associates with numerous co-regulator proteins that regulate the activation status of target genes via flux to the post-translational modification status of histones and the receptor. Here we show that the AR interacts with and is directly methylated by the histone methyltransferase enzyme SET9. Methylation of the AR on lysine 632 is necessary for enhancing transcriptional activity of the receptor by facilitating both inter-domain communication between the N- and C-termini and recruitment to androgen-target genes. We also show that SET9 is pro-proliferative and anti-apoptotic in prostate cancer cells and demonstrates up-regulated nuclear expression in prostate cancer tissue. In all, our date indicate a new mechanism of AR regulation that may be therapeutically exploitable for prostate cancer treatment.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Neoplasias de la Próstata/enzimología , Receptores Androgénicos/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Histonas/metabolismo , Humanos , Lisina/metabolismo , Masculino , Metilación , Antígeno Prostático Específico/genética , Receptores Androgénicos/química , Activación Transcripcional
13.
Oncogene ; 42(32): 2417-2427, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37438470

RESUMEN

A key challenge in the clinical management and cause of treatment failure of prostate cancer (PCa) is its molecular, cellular and clinical heterogeneity. Modelling systems that fully recapitulate clinical diversity and resistant phenotypes are urgently required for the development of successful personalised PCa therapies. The advent of the three-dimensional (3D) organoid model has revolutionised preclinical cancer research through reflecting heterogeneity and offering genomic and environmental manipulation that has opened up unparalleled opportunities for applications in disease modelling, high-throughput drug screening and precision medicine. Despite these remarkable achievements of organoid technology, several shortcomings in emulating the complex tumor microenvironment and dynamic process of metastasis as well as the epigenome profile limit organoids achieving true in vivo functionality. Technological advances in tissue engineering have enabled the development of innovative tools to facilitate the design of improved 3D cancer models. In this review, we highlight the current in vitro 3D PCa models with a special focus on organoids and discuss engineering approaches to create more physiologically relevant PCa organoid models and maximise their translational relevance that ultimately will help to realise the transformational power of precision medicine.


Asunto(s)
Neoplasias de la Próstata , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Organoides/patología , Medicina de Precisión , Microambiente Tumoral/genética
14.
Nat Commun ; 14(1): 7243, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37945563

RESUMEN

Histone modifications influence the recruitment of reader proteins to chromosomes to regulate events including transcription and cell division. The idea of a histone code, where combinations of modifications specify unique downstream functions, is widely accepted and can be demonstrated in vitro. For example, on synthetic peptides, phosphorylation of Histone H3 at threonine-3 (H3T3ph) prevents the binding of reader proteins that recognize trimethylation of the adjacent lysine-4 (H3K4me3), including the TAF3 component of TFIID. To study these combinatorial effects in cells, we analyzed the genome-wide distribution of H3T3ph and H3K4me2/3 during mitosis. We find that H3T3ph anti-correlates with adjacent H3K4me2/3 in cells, and that the PHD domain of TAF3 can bind H3K4me2/3 in isolated mitotic chromatin despite the presence of H3T3ph. Unlike in vitro, H3K4 readers are still displaced from chromosomes in mitosis in Haspin-depleted cells lacking H3T3ph. H3T3ph is therefore unlikely to be responsible for transcriptional downregulation during cell division.


Asunto(s)
Histonas , Factores de Transcripción , Histonas/metabolismo , Fosforilación , Factores de Transcripción/metabolismo , Lectura , Cromosomas/genética , Cromosomas/metabolismo , Mitosis/genética
15.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37751307

RESUMEN

Aberrant androgen receptor (AR) signaling drives prostate cancer (PC), and it is a key therapeutic target. Although initially effective, the generation of alternatively spliced AR variants (AR-Vs) compromises efficacy of treatments. In contrast to full-length AR (AR-FL), AR-Vs constitutively activate androgenic signaling and are refractory to the current repertoire of AR-targeting therapies, which together drive disease progression. There is an unmet clinical need, therefore, to develop more durable PC therapies that can attenuate AR-V function. Exploiting the requirement of coregulatory proteins for AR-V function has the capacity to furnish tractable routes for attenuating persistent oncogenic AR signaling in advanced PC. DNA-PKcs regulates AR-FL transcriptional activity and is upregulated in both early and advanced PC. We hypothesized that DNA-PKcs is critical for AR-V function. Using a proximity biotinylation approach, we demonstrated that the DNA-PK holoenzyme is part of the AR-V7 interactome and is a key regulator of AR-V-mediated transcription and cell growth in models of advanced PC. Crucially, we provide evidence that DNA-PKcs controls global splicing and, via RBMX, regulates the maturation of AR-V and AR-FL transcripts. Ultimately, our data indicate that targeting DNA-PKcs attenuates AR-V signaling and provide evidence that DNA-PKcs blockade is an effective therapeutic option in advanced AR-V-positive patients with PC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Masculino , Humanos , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Dominio Catalítico , Línea Celular Tumoral , Andrógenos/uso terapéutico , ADN , Regulación Neoplásica de la Expresión Génica
16.
Cancer Res ; 81(13): 3664-3678, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33985973

RESUMEN

Androgen deprivation therapy (ADT) is the standard of care for treatment of nonresectable prostate cancer. Despite high treatment efficiency, most patients ultimately develop lethal castration-resistant prostate cancer (CRPC). In this study, we performed a comparative proteomic analysis of three in vivo, androgen receptor (AR)-responsive orthograft models of matched hormone-naïve prostate cancer and CRPC. Differential proteomic analysis revealed that distinct molecular mechanisms, including amino acid (AA) and fatty acid metabolism, are involved in the response to ADT in the different models. Despite this heterogeneity, Schlafen family member 5 (SLFN5) was identified as an AR-regulated protein in CRPC. SLFN5 expression was high in CRPC tumors and correlated with poor patient outcome. In vivo, SLFN5 depletion strongly impaired tumor growth in castrated conditions. Mechanistically, SLFN5 interacted with ATF4 and regulated the expression of LAT1, an essential AA transporter. Consequently, SLFN5 depletion in CRPC cells decreased intracellular levels of essential AA and impaired mTORC1 signaling in a LAT1-dependent manner. These results confirm that these orthograft models recapitulate the high degree of heterogeneity observed in patients with CRPC and further highlight SLFN5 as a clinically relevant target for CRPC. SIGNIFICANCE: This study identifies SLFN5 as a novel regulator of the LAT1 amino acid transporter and an essential contributor to mTORC1 activity in castration-resistant prostate cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular , Humanos , Transportador de Aminoácidos Neutros Grandes 1/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Metaboloma , Ratones , Ratones Desnudos , Pronóstico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Proteoma , Tasa de Supervivencia , Serina-Treonina Quinasas TOR/genética , Transcriptoma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Cell Biol ; 170(2): 191-200, 2005 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-16027218

RESUMEN

Internalization of activated receptors regulates signaling, and endocytic adaptor proteins are well-characterized in clathrin-mediated uptake. One of these adaptor proteins, huntingtin interacting protein 1 (HIP1), induces cellular transformation and is overexpressed in some prostate cancers. We have discovered that HIP1 associates with the androgen receptor through a central coiled coil domain and is recruited to DNA response elements upon androgen stimulation. HIP1 is a novel androgen receptor regulator, significantly repressing transcription when knocked down using a silencing RNA approach and activating transcription when overexpressed. We have also identified a functional nuclear localization signal at the COOH terminus of HIP1, which contributes to the nuclear translocation of the protein. In conclusion, we have discovered that HIP1 is a nucleocytoplasmic protein capable of associating with membranes and DNA response elements and regulating transcription.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Receptores Androgénicos/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Endocitosis , Metabolismo de los Lípidos , Masculino , Datos de Secuencia Molecular , Mutación , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Neoplasias de la Próstata , Transporte de Proteínas , Interferencia de ARN , Elementos de Respuesta , Transcripción Genética
18.
Stem Cells Transl Med ; 9(7): 734-745, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32170918

RESUMEN

Primary culture of human prostate organoids and patient-derived xenografts is inefficient and has limited access to clinical tissues. This hampers their use for translational study to identify new treatments. To overcome this, we established a complementary approach where rapidly proliferating and easily handled induced pluripotent stem cells enabled the generation of human prostate tissue in vivo and in vitro. By using a coculture technique with inductive urogenital sinus mesenchyme, we comprehensively recapitulated in situ 3D prostate histology, and overcame limitations in the primary culture of human prostate stem, luminal and neuroendocrine cells, as well as the stromal microenvironment. This model now unlocks new opportunities to undertake translational studies of benign and malignant prostate disease.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Próstata/metabolismo , Animales , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Embarazo , Ratas , Ratas Sprague-Dawley
19.
Nat Commun ; 11(1): 2508, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32427840

RESUMEN

Despite the clinical success of Androgen Receptor (AR)-targeted therapies, reactivation of AR signalling remains the main driver of castration-resistant prostate cancer (CRPC) progression. In this study, we perform a comprehensive unbiased characterisation of LNCaP cells chronically exposed to multiple AR inhibitors (ARI). Combined proteomics and metabolomics analyses implicate an acquired metabolic phenotype common in ARI-resistant cells and associated with perturbed glucose and lipid metabolism. To exploit this phenotype, we delineate a subset of proteins consistently associated with ARI resistance and highlight mitochondrial 2,4-dienoyl-CoA reductase (DECR1), an auxiliary enzyme of beta-oxidation, as a clinically relevant biomarker for CRPC. Mechanistically, DECR1 participates in redox homeostasis by controlling the balance between saturated and unsaturated phospholipids. DECR1 knockout induces ER stress and sensitises CRPC cells to ferroptosis. In vivo, DECR1 deletion impairs lipid metabolism and reduces CRPC tumour growth, emphasizing the importance of DECR1 in the development of treatment resistance.


Asunto(s)
Metabolismo de los Lípidos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Antagonistas de Receptores Androgénicos/administración & dosificación , Progresión de la Enfermedad , Homeostasis , Humanos , Masculino , Mitocondrias/enzimología , Mitocondrias/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Fosfolípidos/metabolismo , Próstata/enzimología , Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo
20.
Mol Cell Biol ; 26(17): 6502-10, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16914734

RESUMEN

The androgen receptor (AR) is a hormone-dependent transcription factor critically involved in human prostate carcinogenesis. Optimal transcriptional control of androgen-responsive genes by AR may require complex interaction among multiple coregulatory proteins. We have previously shown that the AR coregulator TIP60 can interact with human PIRH2 (hPIRH2). In this study, we uncover important new functional role(s) for hPIRH2 in AR signaling: (i) hPIRH2 interacts with AR and enhances AR-mediated transcription with a dynamic pattern of recruitment to androgen response elements in the prostate-specific antigen (PSA) gene; (ii) hPIRH2 interacts with the AR corepressor HDAC1, leading to reduced HDAC1 protein levels and inhibition of transcriptional repression; (iii) hPIRH2 is required for optimal PSA expression; and (iv) hPIRH2 is involved in prostate cancer cell proliferation. In addition, overexpression of hPIRH2 protein was detected in 73 of 82 (89%) resected prostate cancers, with a strong correlation between increased hPIRH2 expression and aggressive disease, as signified by high Gleason sum scores and the presence of metastatic disease (P = <0.0001 and 0.0004, respectively). Collectively, our data establish hPIRH2 as a key modulator of AR function, opening a new direction for targeted therapy in aggressive human prostate cancer.


Asunto(s)
Inhibidores de Histona Desacetilasas , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Transducción de Señal , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proliferación Celular , Células Cultivadas , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 1 , Humanos , Masculino , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/patología , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA