Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Nat Commun ; 15(1): 4914, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38851846

RESUMEN

FOXA family proteins act as pioneer factors by remodeling compact chromatin structures. FOXA1 is crucial for the chromatin binding of the androgen receptor (AR) in both normal prostate epithelial cells and the luminal subtype of prostate cancer (PCa). Recent studies have highlighted the emergence of FOXA2 as an adaptive response to AR signaling inhibition treatments. However, the role of the FOXA1 to FOXA2 transition in regulating cancer lineage plasticity remains unclear. Our study demonstrates that FOXA2 binds to distinct classes of developmental enhancers in multiple AR-independent PCa subtypes, with its binding depending on LSD1. Moreover, we reveal that FOXA2 collaborates with JUN at chromatin and promotes transcriptional reprogramming of AP-1 in lineage-plastic cancer cells, thereby facilitating cell state transitions to multiple lineages. Overall, our findings underscore the pivotal role of FOXA2 as a pan-plasticity driver that rewires AP-1 to induce the differential transcriptional reprogramming necessary for cancer cell lineage plasticity.


Asunto(s)
Linaje de la Célula , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito , Neoplasias de la Próstata , Factor de Transcripción AP-1 , Masculino , Humanos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-1/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Animales , Cromatina/metabolismo , Cromatina/genética , Plasticidad de la Célula/genética , Reprogramación Celular/genética , Ratones , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Elementos de Facilitación Genéticos/genética , Transcripción Genética
2.
Cancer Res ; 84(14): 2313-2332, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38657108

RESUMEN

Solid tumors are highly reliant on lipids for energy, growth, and survival. In prostate cancer, the activity of the androgen receptor (AR) is associated with reprogramming of lipid metabolic processes. Here, we identified acyl-CoA synthetase medium chain family members 1 and 3 (ACSM1 and ACSM3) as AR-regulated mediators of prostate cancer metabolism and growth. ACSM1 and ACSM3 were upregulated in prostate tumors compared with nonmalignant tissues and other cancer types. Both enzymes enhanced proliferation and protected prostate cancer cells from death in vitro, whereas silencing ACSM3 led to reduced tumor growth in an orthotopic xenograft model. ACSM1 and ACSM3 were major regulators of the prostate cancer lipidome and enhanced energy production via fatty acid oxidation. Metabolic dysregulation caused by loss of ACSM1/3 led to mitochondrial oxidative stress, lipid peroxidation, and cell death by ferroptosis. Conversely, elevated ACSM1/3 activity enabled prostate cancer cells to survive toxic levels of medium chain fatty acids and promoted resistance to ferroptosis-inducing drugs and AR antagonists. Collectively, this study reveals a tumor-promoting function of medium chain acyl-CoA synthetases and positions ACSM1 and ACSM3 as key players in prostate cancer progression and therapy resistance. Significance: Androgen receptor-induced ACSM1 and ACSM3 mediate a metabolic pathway in prostate cancer that enables the utilization of medium chain fatty acids for energy production, blocks ferroptosis, and drives resistance to clinically approved antiandrogens.


Asunto(s)
Proliferación Celular , Coenzima A Ligasas , Ácidos Grasos , Ferroptosis , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Ácidos Grasos/metabolismo , Animales , Ratones , Línea Celular Tumoral , Receptores Androgénicos/metabolismo , Metabolismo de los Lípidos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Biol Cell ; 35(2): ar17, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38019605

RESUMEN

The RNA-binding protein Quaking (QKI) has widespread effects on mRNA regulation including alternative splicing, stability, translation, and localization of target mRNAs. Recently, QKI was found to be induced during epithelial-mesenchymal transition (EMT), where it promotes a mesenchymal alternative splicing signature that contributes to the mesenchymal phenotype. QKI is itself alternatively spliced to produce three major isoforms, QKI-5, QKI-6, and QKI-7. While QKI-5 is primarily localized to the nucleus where it controls mesenchymal splicing during EMT, the functions of the two predominantly cytoplasmic isoforms, QKI-6 and QKI-7, in this context remain uncharacterized. Here we used CRISPR-mediated depletion of QKI in a human mammary epithelial cell model of EMT and studied the effects of expressing the QKI isoforms in isolation and in combination. QKI-5 was required to induce mesenchymal morphology, while combined expression of QKI-5 with either QKI-6 or QKI-7 further enhanced mesenchymal morphology and cell migration. In addition, we found that QKI-6 and QKI-7 can partially localize to the nucleus and contribute to alternative splicing of QKI target genes. These findings indicate that the QKI isoforms function in a dynamic and cooperative manner to promote the mesenchymal phenotype.


Asunto(s)
Empalme Alternativo , Empalme del ARN , Humanos , Isoformas de Proteínas/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
RNA Biol ; 21(1): 1-11, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38112323

RESUMEN

Epithelial-mesenchymal transition (EMT) plays important roles in tumour progression and is orchestrated by dynamic changes in gene expression. While it is well established that post-transcriptional regulation plays a significant role in EMT, the extent of alternative polyadenylation (APA) during EMT has not yet been explored. Using 3' end anchored RNA sequencing, we mapped the alternative polyadenylation (APA) landscape following Transforming Growth Factor (TGF)-ß-mediated induction of EMT in human mammary epithelial cells and found APA generally causes 3'UTR lengthening during this cell state transition. Investigation of potential mediators of APA indicated the RNA-binding protein Quaking (QKI), a splicing factor induced during EMT, regulates a subset of events including the length of its own transcript. Analysis of QKI crosslinked immunoprecipitation (CLIP)-sequencing data identified the binding of QKI within 3' untranslated regions (UTRs) was enriched near cleavage and polyadenylation sites. Following QKI knockdown, APA of many transcripts is altered to produce predominantly shorter 3'UTRs associated with reduced gene expression. These findings reveal the changes in APA that occur during EMT and identify a potential role for QKI in this process.


Asunto(s)
Regulación de la Expresión Génica , Poliadenilación , Humanos , Transición Epitelial-Mesenquimal/genética , Secuencia de Bases , Proteínas de Unión al ARN/genética , Regiones no Traducidas 3'
5.
Br J Cancer ; 129(8): 1350-1361, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37673961

RESUMEN

BACKGROUND: Resistance to androgen receptor signalling inhibitors (ARSIs) represents a major clinical challenge in prostate cancer. We previously demonstrated that the ARSI enzalutamide inhibits only a subset of all AR-regulated genes, and hypothesise that the unaffected gene networks represent potential targets for therapeutic intervention. This study identified the hyaluronan-mediated motility receptor (HMMR) as a survival factor in prostate cancer and investigated its potential as a co-target for overcoming resistance to ARSIs. METHODS: RNA-seq, RT-qPCR and Western Blot were used to evaluate the regulation of HMMR by AR and ARSIs. HMMR inhibition was achieved via siRNA knockdown or pharmacological inhibition using 4-methylumbelliferone (4-MU) in prostate cancer cell lines, a mouse xenograft model and patient-derived explants (PDEs). RESULTS: HMMR was an AR-regulated factor that was unaffected by ARSIs. Genetic (siRNA) or pharmacological (4-MU) inhibition of HMMR significantly suppressed growth and induced apoptosis in hormone-sensitive and enzalutamide-resistant models of prostate cancer. Mechanistically, 4-MU inhibited AR nuclear translocation, AR protein expression and subsequent downstream AR signalling. 4-MU enhanced the growth-suppressive effects of 3 different ARSIs in vitro and, in combination with enzalutamide, restricted proliferation of prostate cancer cells in vivo and in PDEs. CONCLUSION: Co-targeting HMMR and AR represents an effective strategy for improving response to ARSIs.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Masculino , Humanos , Ratones , Animales , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Nitrilos/farmacología , ARN Interferente Pequeño/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Proliferación Celular
6.
Cancer Discov ; 13(8): 1771-1788, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37470668

RESUMEN

Lineage plasticity, a process whereby cells change their phenotype to take on a different molecular and/or histologic identity, is a key driver of cancer progression and therapy resistance. Although underlying genetic changes within the tumor can enhance lineage plasticity, it is predominantly a dynamic process controlled by transcriptional and epigenetic dysregulation. This review explores the transcriptional and epigenetic regulators of lineage plasticity and their interplay with other features of malignancy, such as dysregulated metabolism, the tumor microenvironment, and immune evasion. We also discuss strategies for the detection and treatment of highly plastic tumors. SIGNIFICANCE: Lineage plasticity is a hallmark of cancer and a critical facilitator of other oncogenic features such as metastasis, therapy resistance, dysregulated metabolism, and immune evasion. It is essential that the molecular mechanisms of lineage plasticity are elucidated to enable the development of strategies to effectively target this phenomenon. In this review, we describe key transcriptional and epigenetic regulators of cancer cell plasticity, in the process highlighting therapeutic approaches that may be harnessed for patient benefit.


Asunto(s)
Plasticidad de la Célula , Neoplasias , Humanos , Linaje de la Célula/genética , Plasticidad de la Célula/genética , Neoplasias/genética , Epigénesis Genética , Microambiente Tumoral/genética
7.
Cancer Cell ; 41(7): 1309-1326.e10, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37295428

RESUMEN

The first step of oncogenesis is the acquisition of a repertoire of genetic mutations to initiate and sustain the malignancy. An important example of this initiation phase in acute leukemias is the formation of a potent oncogene by chromosomal translocations between the mixed lineage leukemia (MLL) gene and one of 100 translocation partners, known as the MLL recombinome. Here, we show that circular RNAs (circRNAs)-a family of covalently closed, alternatively spliced RNA molecules-are enriched within the MLL recombinome and can bind DNA, forming circRNA:DNA hybrids (circR loops) at their cognate loci. These circR loops promote transcriptional pausing, proteasome inhibition, chromatin re-organization, and DNA breakage. Importantly, overexpressing circRNAs in mouse leukemia xenograft models results in co-localization of genomic loci, de novo generation of clinically relevant chromosomal translocations mimicking the MLL recombinome, and hastening of disease onset. Our findings provide fundamental insight into the acquisition of chromosomal translocations by endogenous RNA carcinogens in leukemia.


Asunto(s)
Leucemia , Translocación Genética , Animales , Ratones , Humanos , ARN Circular/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Leucemia/genética , Leucemia/patología , ADN , Proteínas de Fusión Oncogénica/genética
8.
Cells ; 12(9)2023 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-37174737

RESUMEN

Trinucleotide repeat disorders comprise ~20 severe, inherited, human neuromuscular and neurodegenerative disorders, which result from an abnormal expansion of repetitive sequences in the DNA. The most common of these, Huntington's disease (HD), results from expansion of the CAG repeat region in exon 1 of the HTT gene via an unknown mechanism. Since non-coding RNAs have been implicated in the initiation and progression of many diseases, herein we focused on a circular RNA (circRNA) molecule arising from non-canonical splicing (backsplicing) of HTT pre-mRNA. The most abundant circRNA from HTT, circHTT(2-6), was found to be more highly expressed in the frontal cortex of HD patients, compared with healthy controls, and positively correlated with CAG repeat tract length. Furthermore, the mouse orthologue (mmu_circHTT(2-6)) was found to be enriched within the brain and specifically the striatum, a region enriched for medium spiny neurons that are preferentially lost in HD. Transgenic overexpression of circHTT(2-6) in two human cell lines-SH-SY5Y and HEK293-reduced cell proliferation and nuclear size without affecting cell cycle progression or cellular size, or altering the CAG repeat region length within HTT. CircHTT(2-6) overexpression did not alter total HTT protein levels, but reduced its nuclear localisation. As these phenotypic and genotypic changes resemble those observed in HD patients, our results suggest that circHTT(2-6) may play a functional role in the pathophysiology of this disease.


Asunto(s)
Enfermedad de Huntington , Neuroblastoma , Humanos , Ratones , Animales , Enfermedad de Huntington/metabolismo , ARN Circular/genética , Células HEK293 , Animales Modificados Genéticamente
9.
Prostate Cancer Prostatic Dis ; 26(4): 673-680, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36859711

RESUMEN

OBJECTIVES: The prognostic capacity of positive surgical margins (PSM) for biochemical recurrence (BCR) is unclear, with inconsistent findings across published studies. We aimed to systematically review and perform a meta-analysis exploring the impact of Positive surgical margin length on biochemical recurrence in men after radical prostatectomy. METHODS: A search was conducted using the MEDLINE, Scopus, Embase and Cochrane databases according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. The quality of the studies was assessed using the Newcastle-Ottawa scale, and the protocol was registered in advance (PROSPERO: CRD42020195908). This meta-analysis included 16 studies with BCR as the primary outcome measure. RESULTS: Studies used various dichotomised thresholds for PSM length. A subgroup meta-analysis was performed using the reported multivariable hazard ratio (Continuous, 3, and 1 mm PSM length). PSM length (continuous) was independently associated with an increased risk of BCR (7 studies, HR 1.04 (CI 1.02-1.05), I2 = 8% p < 0.05). PSM length greater than 3 mm conferred a higher risk of BCR compared to less than 3 mm (4 studies, HR 1.99 (1.54-2.58) I2 = 0%, p < 0.05). There was also an increased risk of BCR associated with PSM length of less than 1 mm compared to negative surgical margins (3 studies, HR 1.46 (1.05-2.04), I2 = 0%, P = 0.02). CONCLUSION: PSM length is independently prognostic for BCR after radical prostatectomy. Further long-term studies are needed to estimate the impact on systemic progression.


Asunto(s)
Márgenes de Escisión , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/etiología , Próstata , Prostatectomía/métodos , Pronóstico , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/etiología , Estudios Retrospectivos
10.
Ther Adv Med Oncol ; 15: 17588359231152839, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36743527

RESUMEN

Despite key advances in the treatment of prostate cancer (PCa), a proportion of men have de novo resistance, and all will develop resistance to current therapeutics over time. Aberrant lipid metabolism has long been associated with prostate carcinogenesis and progression, but more recently there has been an explosion of preclinical and clinical data which is informing new clinical trials. This review explores the epidemiological links between obesity and metabolic syndrome and PCa, the evidence for altered circulating lipids in PCa and their potential role as biomarkers, as well as novel therapeutic strategies for targeting lipids in men with PCa, including therapies widely used in cardiovascular disease such as statins, metformin and lifestyle modification, as well as novel targeted agents such as sphingosine kinase inhibitors, DES1 inhibitors and agents targeting FASN and beta oxidation.

11.
Small Methods ; 6(8): e2200471, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35764869

RESUMEN

Measuring tumor cell invasiveness through 3D tissues, particularly at the single-cell level, can provide important mechanistic understanding and assist in identifying therapeutic targets of tumor invasion. However, current experimental approaches, including standard in vitro invasion assays, have limited physiological relevance and offer insufficient insight into the vast heterogeneity in tumor cell migration through tissues. To address these issues, here the concept of optical cellular micromotion is reported on, where digital holographic microscopy is used to map the optical nano- to submicrometer thickness fluctuations within single-cells. These fluctuations are driven by the dynamic movement of subcellular structures including the cytoskeleton and inherently associated with the biological processes involved in cell invasion within tissues. It is experimentally demonstrated that the optical cellular micromotion correlates with tumor cells motility and invasiveness both at the population and single-cell levels. In addition, the optical cellular micromotion significantly reduced upon treatment with migrastatic drugs that inhibit tumor cell invasion. These results demonstrate that micromotion measurements can rapidly and non-invasively determine the invasive behavior of single tumor cells within tissues, yielding a new and powerful tool to assess the efficacy of approaches targeting tumor cell invasiveness.


Asunto(s)
Holografía , Procesos Neoplásicos , Línea Celular Tumoral , Movimiento Celular/fisiología , Geles , Holografía/métodos , Humanos
12.
Cancer Res Commun ; 2(7): 706-724, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-36923279

RESUMEN

Inhibiting the androgen receptor (AR), a ligand-activated transcription factor, with androgen deprivation therapy is a standard-of-care treatment for metastatic prostate cancer. Paradoxically, activation of AR can also inhibit the growth of prostate cancer in some patients and experimental systems, but the mechanisms underlying this phenomenon are poorly understood. This study exploited a potent synthetic androgen, methyltestosterone (MeT), to investigate AR agonist-induced growth inhibition. MeT strongly inhibited growth of prostate cancer cells expressing AR, but not AR-negative models. Genes and pathways regulated by MeT were highly analogous to those regulated by DHT, although MeT induced a quantitatively greater androgenic response in prostate cancer cells. MeT potently downregulated DNA methyltransferases, leading to global DNA hypomethylation. These epigenomic changes were associated with dysregulation of transposable element expression, including upregulation of endogenous retrovirus (ERV) transcripts after sustained MeT treatment. Increased ERV expression led to accumulation of double-stranded RNA and a "viral mimicry" response characterized by activation of IFN signaling, upregulation of MHC class I molecules, and enhanced recognition of murine prostate cancer cells by CD8+ T cells. Positive associations between AR activity and ERVs/antiviral pathways were evident in patient transcriptomic data, supporting the clinical relevance of our findings. Collectively, our study reveals that the potent androgen MeT can increase the immunogenicity of prostate cancer cells via a viral mimicry response, a finding that has potential implications for the development of strategies to sensitize this cancer type to immunotherapies. Significance: Our study demonstrates that potent androgen stimulation of prostate cancer cells can elicit a viral mimicry response, resulting in enhanced IFN signaling. This finding may have implications for the development of strategies to sensitize prostate cancer to immunotherapies.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Masculino , Humanos , Animales , Ratones , Receptores Androgénicos/genética , Andrógenos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Antagonistas de Andrógenos/farmacología , Linfocitos T CD8-positivos/metabolismo , ADN
13.
Nat Commun ; 12(1): 6377, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34737261

RESUMEN

Endocrine therapies for prostate cancer inhibit the androgen receptor (AR) transcription factor. In most cases, AR activity resumes during therapy and drives progression to castration-resistant prostate cancer (CRPC). However, therapy can also promote lineage plasticity and select for AR-independent phenotypes that are uniformly lethal. Here, we demonstrate the stem cell transcription factor Krüppel-like factor 5 (KLF5) is low or absent in prostate cancers prior to endocrine therapy, but induced in a subset of CRPC, including CRPC displaying lineage plasticity. KLF5 and AR physically interact on chromatin and drive opposing transcriptional programs, with KLF5 promoting cellular migration, anchorage-independent growth, and basal epithelial cell phenotypes. We identify ERBB2 as a point of transcriptional convergence displaying activation by KLF5 and repression by AR. ERBB2 inhibitors preferentially block KLF5-driven oncogenic phenotypes. These findings implicate KLF5 as an oncogene that can be upregulated in CRPC to oppose AR activities and promote lineage plasticity.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Células Neuroendocrinas/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptor ErbB-2/metabolismo , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Estadificación de Neoplasias , Células Neuroendocrinas/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Transducción de Señal , Activación Transcripcional
14.
Nat Cell Biol ; 23(9): 1023-1034, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34489572

RESUMEN

Cancers adapt to increasingly potent targeted therapies by reprogramming their phenotype. Here we investigated such a phenomenon in prostate cancer, in which tumours can escape epithelial lineage confinement and transition to a high-plasticity state as an adaptive response to potent androgen receptor (AR) antagonism. We found that AR activity can be maintained as tumours adopt alternative lineage identities, with changes in chromatin architecture guiding AR transcriptional rerouting. The epigenetic regulator enhancer of zeste homologue 2 (EZH2) co-occupies the reprogrammed AR cistrome to transcriptionally modulate stem cell and neuronal gene networks-granting privileges associated with both fates. This function of EZH2 was associated with T350 phosphorylation and establishment of a non-canonical polycomb subcomplex. Our study provides mechanistic insights into the plasticity of the lineage-infidelity state governed by AR reprogramming that enabled us to redirect cell fate by modulating EZH2 and AR, highlighting the clinical potential of reversing resistance phenotypes.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Redes Reguladoras de Genes/fisiología , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/genética , Transducción de Señal/fisiología
15.
Cancer Res ; 81(19): 4981-4993, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34362796

RESUMEN

Dysregulated lipid metabolism is a prominent feature of prostate cancer that is driven by androgen receptor (AR) signaling. Here we used quantitative mass spectrometry to define the "lipidome" in prostate tumors with matched benign tissues (n = 21), independent unmatched tissues (n = 47), and primary prostate explants cultured with the clinical AR antagonist enzalutamide (n = 43). Significant differences in lipid composition were detected and spatially visualized in tumors compared with matched benign samples. Notably, tumors featured higher proportions of monounsaturated lipids overall and elongated fatty acid chains in phosphatidylinositol and phosphatidylserine lipids. Significant associations between lipid profile and malignancy were validated in unmatched samples, and phospholipid composition was characteristically altered in patient tissues that responded to AR inhibition. Importantly, targeting tumor-related lipid features via inhibition of acetyl-CoA carboxylase 1 significantly reduced cellular proliferation and induced apoptosis in tissue explants. This characterization of the prostate cancer lipidome in clinical tissues reveals enhanced fatty acid synthesis, elongation, and desaturation as tumor-defining features, with potential for therapeutic targeting. SIGNIFICANCE: This study identifies malignancy and treatment-associated changes in lipid composition of clinical prostate cancer tissues, suggesting that mediators of these lipidomic changes could be targeted using existing metabolic agents.


Asunto(s)
Metabolismo de los Lípidos , Lipidómica , Lípidos de la Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Biomarcadores , Biología Computacional/métodos , Metabolismo Energético , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipidómica/métodos , Masculino , Metabolómica/métodos , Terapia Molecular Dirigida , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/etiología , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
16.
Elife ; 102021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34382934

RESUMEN

Alterations to the androgen receptor (AR) signalling axis and cellular metabolism are hallmarks of prostate cancer. This study provides insight into both hallmarks by uncovering a novel link between AR and the pentose phosphate pathway (PPP). Specifically, we identify 6-phosphogluoconate dehydrogenase (6PGD) as an androgen-regulated gene that is upregulated in prostate cancer. AR increased the expression of 6PGD indirectly via activation of sterol regulatory element binding protein 1 (SREBP1). Accordingly, loss of 6PGD, AR or SREBP1 resulted in suppression of PPP activity as revealed by 1,2-13C2 glucose metabolic flux analysis. Knockdown of 6PGD also impaired growth and elicited death of prostate cancer cells, at least in part due to increased oxidative stress. We investigated the therapeutic potential of targeting 6PGD using two specific inhibitors, physcion and S3, and observed substantial anti-cancer activity in multiple models of prostate cancer, including aggressive, therapy-resistant models of castration-resistant disease as well as prospectively collected patient-derived tumour explants. Targeting of 6PGD was associated with two important tumour-suppressive mechanisms: first, increased activity of the AMP-activated protein kinase (AMPK), which repressed anabolic growth-promoting pathways regulated by acetyl-CoA carboxylase 1 (ACC1) and mammalian target of rapamycin complex 1 (mTORC1); and second, enhanced AR ubiquitylation, associated with a reduction in AR protein levels and activity. Supporting the biological relevance of positive feedback between AR and 6PGD, pharmacological co-targeting of both factors was more effective in suppressing the growth of prostate cancer cells than single-agent therapies. Collectively, this work provides new insight into the dysregulated metabolism of prostate cancer and provides impetus for further investigation of co-targeting AR and the PPP as a novel therapeutic strategy.


Asunto(s)
Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Línea Celular , Emodina/análogos & derivados , Retroalimentación , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Vía de Pentosa Fosfato , Neoplasias de la Próstata/genética , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
17.
Cancers (Basel) ; 13(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209750

RESUMEN

Breast and prostate cancer are the second and third leading causes of death amongst all cancer types, respectively. Pathogenesis of these malignancies is characterised by dysregulation of sex hormone signalling pathways, mediated by the estrogen receptor-α (ER) in breast cancer and androgen receptor (AR) in prostate cancer. ER and AR are transcription factors whose aberrant function drives oncogenic transcriptional programs to promote cancer growth and progression. While ER/AR are known to stimulate cell growth and survival by modulating gene transcription, emerging findings indicate that their effects in neoplasia are also mediated by dysregulation of protein synthesis (i.e., mRNA translation). This suggests that ER/AR can coordinately perturb both transcriptional and translational programs, resulting in the establishment of proteomes that promote malignancy. In this review, we will discuss relatively understudied aspects of ER and AR activity in regulating protein synthesis as well as the potential of targeting mRNA translation in breast and prostate cancer.

18.
SLAS Discov ; 26(9): 1107-1124, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34111999

RESUMEN

New treatments are required for advanced prostate cancer; however, there are fewer preclinical models of prostate cancer than other common tumor types to test candidate therapeutics. One opportunity to increase the scope of preclinical studies is to grow tissue from patient-derived xenografts (PDXs) as organoid cultures. Here we report a scalable pipeline for automated seeding, treatment and an analysis of the drug responses of prostate cancer organoids. We established organoid cultures from 5 PDXs with diverse phenotypes of prostate cancer, including castrate-sensitive and castrate-resistant disease, as well as adenocarcinoma and neuroendocrine pathology. We robotically embedded organoids in Matrigel in 384-well plates and monitored growth via brightfield microscopy before treatment with poly ADP-ribose polymerase inhibitors or a compound library. Independent readouts including metabolic activity and live-cell imaging-based features provided robust measures of organoid growth and complementary ways of assessing drug efficacy. Single organoid analyses enabled in-depth assessment of morphological differences between patients and within organoid populations and revealed that larger organoids had more striking changes in morphology and composition after drug treatment. By increasing the scale and scope of organoid experiments, this automated assay complements other patient-derived models and will expedite preclinical testing of new treatments for prostate cancer.


Asunto(s)
Descubrimiento de Drogas/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Ensayos Analíticos de Alto Rendimiento , Imagen Molecular/métodos , Organoides , Técnicas de Cultivo de Tejidos , Algoritmos , Animales , Automatización de Laboratorios , Análisis de Datos , Modelos Animales de Enfermedad , Composición de Medicamentos , Xenoinjertos , Humanos , Masculino , Ratones , Neoplasias de la Próstata
19.
BJU Int ; 127 Suppl 1: 13-22, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33973331

RESUMEN

OBJECTIVES: To systematically review and perform a meta-analysis of studies investigating the role of primary Gleason grade (PGG), Gleason score (GS) or Gleason grade group (GGG) at positive surgical margins (PSMs) after radical prostatectomy (RP) in predicting biochemical recurrence (BCR) and oncological outcomes. METHODS: A systematic search was conducted using the MEDLINE, Scopus, Embase and Cochrane databases according to Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. Studies published between 2005 and 2019 were included. The quality of the studies selected was assessed, and a protocol was registered and published in advance (PROSPERO: CRD42019131800). The primary outcome measure was BCR. Secondary outcome measures included cancer-specific survival, metastasis-free survival and overall mortality during the follow-up period. RESULTS: Our systematic search yielded 3116 unique results. Ten studies were selected for meta-analysis. The sample sizes of PSM cohorts varied from 200 to 956, while the median follow-up ranged from 1.5 to 13 years. Most studies used BCR as a surrogate marker for disease progression; only two studies reported other oncological outcomes. Meta-analysis was performed in selected groups (PGG, GS and GGG). PGG 4 or 5 at the PSM was found to be predictive of BCR (hazard ratio [HR] 1.66, 95% confidence interval [CI] 1.37-2.02; P < 0.01). GGG > 1 at margin was also predictive of BCR compared to GGG 1 (GGG 1 vs 2: HR 2.35, 95% CI 1.6 -3.46; P < 0.001; GGG 1 vs 3: HR 3.95, 95% CI 1.82-8.57; P = 0.005; GGG 1 vs 4: HR 7.17, 95% CI 1.76-29.17; P = 0.006; and GGG 1 vs 5: HR 12.37, 95% CI 1.80-84.82; P = 0.01). CONCLUSION: Gleason score, PGG and GGG at the PSM are associated with a significantly increased risk of BCR. Longer-term studies are needed to investigate the utility of PGG, GS or GGG at the PSM in their ability to predict not only BCR but other outcomes such as cancer-specific survival, metastasis-free survival and overall survival.


Asunto(s)
Clasificación del Tumor , Recurrencia Local de Neoplasia/sangre , Recurrencia Local de Neoplasia/patología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Progresión de la Enfermedad , Humanos , Masculino , Márgenes de Escisión , Neoplasia Residual , Antígeno Prostático Específico/sangre , Prostatectomía , Neoplasias de la Próstata/sangre
20.
Cell Mol Life Sci ; 78(8): 4035-4052, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33834258

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) is an important regulator of cellular metabolism that is commonly hyperactivated in cancer. Recent cancer genome screens have identified multiple mutations in Ras-homolog enriched in brain (Rheb), the primary activator of mTORC1 that might act as driver oncogenes by causing hyperactivation of mTORC1. Here, we show that a number of recurrently occurring Rheb mutants drive hyperactive mTORC1 signalling through differing levels of insensitivity to the primary inactivator of Rheb, tuberous sclerosis complex. We show that two activated mutants, Rheb-T23M and E40K, strongly drive increased cell growth, proliferation and anchorage-independent growth resulting in enhanced tumour growth in vivo. Proteomic analysis of cells expressing the mutations revealed, surprisingly, that these two mutants promote distinct oncogenic pathways with Rheb-T23M driving an increased rate of anaerobic glycolysis, while Rheb-E40K regulates the translation factor eEF2 and autophagy, likely through differential interactions with 5' AMP-activated protein kinase (AMPK) which modulate its activity. Our findings suggest that unique, personalized, combination therapies may be utilised to treat cancers according to which Rheb mutant they harbour.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neoplasias/genética , Mutación Puntual , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Células HEK293 , Células HeLa , Humanos , Ratones , Modelos Moleculares , Células 3T3 NIH , Neoplasias/metabolismo , Proteoma/metabolismo , Proteómica , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...