Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Am J Hosp Palliat Care ; 36(6): 519-525, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30602311

ABSTRACT

BACKGROUND: Although 50% to 90% of patients who receive epidermal growth factor receptor (EGFR) inhibitors develop a rash, options for rash prevention or palliation remain limited. This issue is particularly important from a palliative care standpoint because these agents are prescribed only to patients with incurable cancer. Here, we report (1) gene expression profiling of skin biopsies from patients with an EGFR inhibitor-induced rash and (2) a randomized, placebo-controlled feasibility trial with the antiandrogen, spironolactone. Both investigations were undertaken to begin to explore the hypothesis that androgens mediate EGFR inhibitor-induced rash and that antiandrogens palliate it. METHODS/RESULTS: First, 4 skin biopsies from patients with EGFR inhibitor-induced rash (3 men and 1 woman) were subject to gene expression microarray profiling. A public data set of normal skin gene expression (Gene Expression Omnibus, GSE22998) served as a reference. Sixty percent of commonly interrogated androgen receptor genes (207 of 308 between the 2 data sets) were differentially expressed ( P < .05) in the rash samples. Second, in a 17-patient double-blinded, placebo-controlled trial with topical spironolactone applied to the face, although the primary feasibility end point was not achieved, patients in the spironolactone arm received more doses of EGFR inhibitor, and anecdotal photographic evidence suggested salutatory effects of spironolactone on rash. CONCLUSIONS: Epidermal growth factor receptor inhibitor-induced rash appears to be androgen-mediated; antiandrogen therapy merits further study for rash prevention/palliation.


Subject(s)
Androgen Antagonists/therapeutic use , ErbB Receptors/antagonists & inhibitors , Exanthema/chemically induced , Exanthema/drug therapy , Spironolactone/therapeutic use , Adult , Aged , Biopsy , Double-Blind Method , Exanthema/pathology , Female , Humans , Male , Middle Aged , Pilot Projects , Transcriptome
2.
Nucleic Acids Res ; 46(4): 1895-1911, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29309643

ABSTRACT

Androgen receptor (AR) splice variants (ARVs) are implicated in development of castration-resistant prostate cancer (CRPC). Upregulation of ARVs often correlates with persistent AR activity after androgen deprivation therapy (ADT). However, the genomic and epigenomic characteristics of ARV-dependent cistrome and the disease relevance of ARV-mediated transcriptome remain elusive. Through integrated chromatin immunoprecipitation coupled sequencing (ChIP-seq) and RNA sequencing (RNA-seq) analysis, we identified ARV-preferential-binding sites (ARV-PBS) and a set of genes preferentially transactivated by ARVs in CRPC cells. ARVs preferentially bind to enhancers located in nucleosome-depleted regions harboring the full AR-response element (AREfull), while full-length AR (ARFL)-PBS are enhancers resided in closed chromatin regions containing the composite FOXA1-nnnn-AREhalf motif. ARV-PBS exclusively overlapped with AR binding sites in castration-resistant (CR) tumors in patients and ARV-preferentially activated genes were up-regulated in abiraterone-resistant patient specimens. Expression of ARV-PBS target genes, such as oncogene RAP2A and cell cycle gene E2F7, were significantly associated with castration resistance, poor survival and tumor progression. We uncover distinct genomic and epigenomic features of ARV-PBS, highlighting that ARVs are useful tools to depict AR-regulated oncogenic genome and epigenome landscapes in prostate cancer. Our data also suggest that the ARV-preferentially activated transcriptional program could be targeted for effective treatment of CRPC.


Subject(s)
Androstenes/therapeutic use , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/metabolism , Animals , Binding Sites , Cell Line, Tumor , Chromatin/metabolism , Disease Progression , Drug Resistance, Neoplasm , Epigenesis, Genetic , Genomics , Humans , Male , Mice, SCID , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Isoforms/metabolism , RNA Splicing , Receptors, Androgen/genetics , rap GTP-Binding Proteins/physiology
3.
Lancet Oncol ; 17(10): 1435-1444, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27575027

ABSTRACT

BACKGROUND: HSD3B1 (1245A>C) has been mechanistically linked to castration-resistant prostate cancer because it encodes an altered enzyme that augments dihydrotestosterone synthesis from non-gonadal precursors. We postulated that men inheriting the HSD3B1 (1245C) allele would exhibit resistance to androgen-deprivation therapy (ADT). METHODS: In this multicohort study, we determined HSD3B1 genotype retrospectively in men treated with ADT for post-prostatectomy biochemical failure and correlated genotype with long-term clinical outcomes. We used data and samples from prospectively maintained prostate cancer registries at the Cleveland Clinic (Cleveland, OH, USA; primary study cohort) and the Mayo Clinic (Rochester, MN, USA; post-prostatectomy and metastatic validation cohorts). In the post-prostatectomy cohorts, patients of any age were eligible if they underwent prostatectomy between Jan 1, 1996, and Dec 31, 2009 (at the Cleveland Clinic; primary cohort), or between Jan 1, 1987, and Dec 31, 2011 (at the Mayo Clinic; post-prostatectomy cohort) and were treated with ADT for biochemical failure or for non-metastatic clinical failure. In the metastatic validation cohort, patients of any age were eligible if they were enrolled at Mayo Clinic between Sept 1, 2009, and July 31, 2013, with metastatic castration-resistant prostate cancer. The primary endpoint was progression-free survival according to HSD3B1 genotype. We did prespecified multivariable analyses to assess the independent predictive value of HSD3B1 genotype on outcomes. FINDINGS: We included and genotyped 443 patients: 118 in the primary cohort (who underwent prostatectomy), 137 in the post-prostatectomy validation cohort, and 188 in the metastatic validation cohort. In the primary study cohort, median progression-free survival diminished as a function of the number of variant alleles inherited: 6·6 years (95% CI 3·8-not reached) in men with homozygous wild-type genotype, 4·1 years (3·0-5·5) in men with heterozygous variant genotype, and 2·5 years (0·7 to not reached) in men with homozygous variant genotype (p=0·011). Relative to the homozygous wild-type genotype, inheritance of two copies of the variant allele was predictive of decreased progression-free survival (hazard ratio [HR] 2·4 [95% CI 1·1-5·3], p=0·029), as was inheritance of one copy of the variant allele (HR 1·7 [1·0-2·9], p=0·041). Findings were similar for distant metastasis-free survival and overall survival. The effect of the HSD3B1 genotype was independently confirmed in the validation cohorts. INTERPRETATION: Inheritance of the HSD3B1 (1245C) allele that enhances dihydrotestosterone synthesis is associated with prostate cancer resistance to ADT. HSD3B1 could therefore potentially be a powerful genetic biomarker capable of distinguishing men who are a priori likely to fare favourably with ADT from those who harbour disease liable to behave more aggressively, and who therefore might warrant early escalated therapy. FUNDING: Prostate Cancer Foundation, National Institutes of Health, US Department of Defense, Howard Hughes Medical Institute, American Cancer Society, Conquer Cancer Foundation of the American Society of Clinical Oncology, Cleveland Clinic Research Programs Committee and Department of Radiation Oncology, Gail and Joseph Gassner Development Funds.


Subject(s)
Androgen Antagonists/therapeutic use , Multienzyme Complexes/genetics , Progesterone Reductase/genetics , Prostatic Neoplasms/drug therapy , Steroid Isomerases/genetics , Aged , Cohort Studies , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Retrospective Studies
4.
Cell Rep ; 15(3): 599-610, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27068475

ABSTRACT

The androgen receptor (AR) is required for castration-resistant prostate cancer (CRPC) progression, but the function and disease relevance of AR-bound enhancers remain unclear. Here, we identify a group of AR-regulated enhancer RNAs (e.g., PSA eRNA) that are upregulated in CRPC cells, patient-derived xenografts (PDXs), and patient tissues. PSA eRNA binds to CYCLIN T1, activates P-TEFb, and promotes cis and trans target gene transcription by increasing serine-2 phosphorylation of RNA polymerase II (Pol II-Ser2p). We define an HIV-1 TAR RNA-like (TAR-L) motif in PSA eRNA that is required for CYCLIN T1 binding. Using TALEN-mediated gene editing we further demonstrate that this motif is essential for increased Pol II-Ser2p occupancy levels and CRPC cell growth. We have uncovered a P-TEFb activation mechanism and reveal altered eRNA expression that is related to abnormal AR function and may potentially be a therapeutic target in CRPC.


Subject(s)
Positive Transcriptional Elongation Factor B/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , RNA/metabolism , Receptors, Androgen/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation , Cyclin T/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic , Genetic Loci , Humans , Male , Models, Biological , Nucleotide Motifs/genetics , Phosphorylation , Prostate-Specific Antigen/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Binding , RNA Polymerase II/metabolism , Serine/metabolism , Up-Regulation/genetics
5.
Cancer Res ; 75(22): 4688-96, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26432404

ABSTRACT

Clinical trials have established the benefit of androgen deprivation therapy (ADT) combined with radiotherapy in prostate cancer. ADT sensitizes prostate cancer to radiotherapy-induced death at least in part through inhibition of DNA repair machinery, but for unknown reasons, adjuvant ADT provides further survival benefits. Here, we show that androgen receptor (AR) expression and activity are durably upregulated following radiotherapy in multiple human prostate cancer models in vitro and in vivo. Moreover, the degree of AR upregulation correlates with survival in vitro and time to tumor progression in animal models. We also provide evidence of AR pathway upregulation, measured by a rise in serum levels of AR-regulated hK2 protein, in nearly 20% of patients after radiotherapy. Furthermore, these men were three-fold more likely to experience subsequent biochemical failure. Collectively, these data demonstrate that radiotherapy can upregulate AR signaling after therapy to an extent that negatively affects disease progression and/or survival.


Subject(s)
Prostatic Neoplasms/radiotherapy , Radiation Tolerance/physiology , Receptors, Androgen/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Comet Assay , Fluorescent Antibody Technique , Humans , Luminescent Measurements , Male , Mice , Mice, SCID , Prostatic Neoplasms/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation , Xenograft Model Antitumor Assays
6.
Onco Targets Ther ; 8: 1915-22, 2015.
Article in English | MEDLINE | ID: mdl-26261420

ABSTRACT

This study tested the potential of circulating RNA-based signals as predictive biomarkers for docetaxel response in patients with metastatic castration-resistant prostate cancer (CRPC). RNA was analyzed in blood from six CRPC patients by whole-transcriptome sequencing (total RNA-sequencing) before and after docetaxel treatment using the Illumina's HiSeq platform. Targeted RNA capture and sequencing was performed in an independent cohort of ten patients with CRPC matching the discovery cohort to confirm differential expression of the genes. Response to docetaxel was defined on the basis of prostate-specific antigen levels and imaging criteria. Two-way analysis of variance was used to compare differential gene expression in patients classified as responders versus nonresponders before and after docetaxel treatment. Thirty-four genes with two-fold differentially expressed transcripts in responders versus nonresponders were selected from total RNA-sequencing for further validation. Targeted RNA capture and sequencing showed that 13/34 genes were differentially expressed in responders. Alpha defensin genes DEFA1, DEFA1B, and DEFA3 exhibited significantly higher expression in responder patients compared with nonresponder patients before administration of chemotherapy (fold change >2.5). In addition, post-docetaxel treatment significantly increased transcript levels of these defensin genes in responders (fold change >2.8). Our results reveal that patients with higher defensin RNA transcripts in blood respond well to docetaxel therapy. We suggest that monitoring DEFA1, DEFA1B, and DEFA3 RNA transcripts in blood prior to treatment will be helpful to determine which patients are better candidates to receive docetaxel chemotherapy.

7.
Nat Rev Urol ; 12(3): 137-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25666893

ABSTRACT

Androgen receptor splice variants (AR-Vs)--which are expressed in castration-resistant prostate cancer (CRPC) cell lines and clinical samples--lack the C-terminal ligand-binding domain and are constitutively active. AR-Vs are, therefore, resistant to traditional androgen deprivation therapy (ADT). AR-Vs are induced by several mechanisms, including ADT, and might contribute to the progression of CRPC and resistance to ADT. AR-Vs could represent a novel therapeutic target for prostate cancer, especially in CRPC.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Alternative Splicing , Animals , Disease Progression , Gene Expression Profiling , Gene Expression Regulation/physiology , Humans , Male , Neoplasms, Hormone-Dependent , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Protein Isoforms , Receptors, Androgen/genetics , Signal Transduction/physiology
8.
J Urol ; 193(2): 690-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25132238

ABSTRACT

PURPOSE: Spliced variant forms of androgen receptor were recently identified in castration resistant prostate cancer cell lines and clinical samples. We identified the cistrome and gene signature of androgen receptor splice variants in castration resistant prostate cancer cell lines and determined the clinical significance of androgen receptor splice variant regulated genes. MATERIALS AND METHODS: The castration resistant prostate cancer cell line 22Rv1, which expresses full-length androgen receptor and androgen receptor splice variants endogenously, was used as the research model. We established 22Rv1-ARFL(-)/ARV(+) and 22Rv1-ARFL(-)/ARV(-) through RNA interference. Chromatin immunoprecipitation coupled with next generation sequencing and microarray techniques were used to identify the cistrome and gene expression profiles of androgen receptor splice variants in the absence of androgen. RESULTS: Androgen receptor splice variant binding sites were identified in 22Rv1-ARFL(-)/ARV(+). A gene set was regulated uniquely by androgen receptor splice variants but not by full-length androgen receptor in the absence of androgen. Integrated analysis revealed that some genes were directly modulated by androgen receptor splice variants. Unsupervised clustering analysis showed that the androgen receptor splice variant gene signature differentiated benign from malignant prostate tissue as well as localized prostate cancer from metastatic castration resistant prostate cancer specimens. Some genes that were modulated uniquely by androgen receptor splice variants also correlated with histological grade and biochemical failure. CONCLUSIONS: Androgen receptor splice variants can bind to DNA independent of full-length androgen receptor in the absence of androgen and modulate a unique set of genes that is not regulated by full-length androgen receptor. The androgen receptor splice variant gene signature correlates with disease progression. It distinguishes primary cancer from castration resistant prostate cancer specimens and benign from malignant prostate specimens.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Transcriptome , High-Throughput Nucleotide Sequencing , Humans , Male , Protein Isoforms , Tumor Cells, Cultured
9.
Urology ; 84(2): 393-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24916669

ABSTRACT

OBJECTIVE: To explore explanations for the numerical imbalance of biopsy-detected Gleason 8-10 prostate cancers (PCa) diagnosed in years 3-4 in the dutasteride and placebo groups of the Reduction by Dutasteride of Prostate Cancer Events (REDUCE) study. METHODS: REDUCE was a 4-year, randomized, double-blind, placebo-controlled trial of dutasteride (0.5 mg/d) vs placebo for PCa risk reduction. We modeled the incidence of Gleason 8-10 cancer and used logistic regression analysis to evaluate the effects of baseline predictors of PCa, as well as post-baseline prostate volume at the time of biopsy, on PCa diagnosis. We compared needle biopsy Gleason scores with corresponding surgery Gleason scores. All statistical tests conducted were 2-sided. RESULTS: Had there been a scheduled biopsy occurring only at year 4, we estimated a similar incidence of Gleason 8-10 PCa in the dutasteride (n = 45) and placebo (n = 46) groups. Two biopsy Gleason 7 cancers in the placebo group (n = 150) were upgraded to Gleason 8-10 cancer on prostatectomy, and no patients in the dutasteride group (n = 111) were upgraded. Logistic regression analysis demonstrated the effect of prostate volume on Gleason 8-10 cancer diagnosis. CONCLUSION: Although modeling of REDUCE data showed a similar incidence of Gleason 8-10 cancer in the dutasteride and placebo groups at year 4, an association between dutasteride and Gleason 8-10 cancer cannot be definitely excluded. It is likely that several biases, notably study design and prostate size at the time of biopsy, contributed to the numerical imbalance in Gleason 8-10 cancers observed between the treatment groups in years 3-4.


Subject(s)
5-alpha Reductase Inhibitors/therapeutic use , Azasteroids/therapeutic use , Models, Statistical , Prostatic Neoplasms/classification , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Biopsy, Needle , Dutasteride , Humans , Male , Neoplasm Grading , Randomized Controlled Trials as Topic , Time Factors
10.
Int J Biol Sci ; 10(6): 614-9, 2014.
Article in English | MEDLINE | ID: mdl-24948874

ABSTRACT

Androgens and the androgen receptor (AR) are essential for growth and differentiation of the normal prostate gland as well as proliferation and survival of prostate cancer (PCa). Increasing evidence suggests that reactivation of the AR plays a pivotal role in disease progression to castration-resistant PCa (CRPC). Forkhead box (FOX) factors exert two distinct effects on AR function in PCa. The A-class of FOX proteins, especially FOXA1, functions as a pioneer factor to facilitate AR transactivation and PCa growth. In contrast, the O-class of FOX proteins such as FOXO1 and FOXO3, which are downstream effectors of the PTEN tumor suppressor, inhibit the transcriptional activity of either full-length AR or constitutively active splice variants of AR in a direct or indirect manner in PCa. FOXO1 also contributes to taxane-mediated inhibition of the AR and CRPC growth. Therefore, FOX family members not only have a tight relationship with AR, but also represent a pivotal group of proteins to be targeted for PCa therapy. The present review focuses primarily on recent advances in the epigenetic, mechanistic and clinical relevant aspects of regulation of the AR by FOXA1 and FOXO1 factors in PCa.


Subject(s)
Forkhead Transcription Factors/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgens/metabolism , Animals , Forkhead Box Protein O1 , Humans , Male
11.
Mol Cancer Ther ; 13(5): 1067-77, 2014 May.
Article in English | MEDLINE | ID: mdl-24674886

ABSTRACT

Azadirachta indica, commonly known as neem, has gained worldwide prominence because of its medical properties, namely antitumor, antiviral, anti-inflammatory, antihyperglycemic, antifungal, and antibacterial activities. Despite these promising results, gaps remain in our understanding of the molecular mechanism of action of neem compounds and their potential for use in clinical trials. We investigated supercritical extract of neem leaves (SENL) for the following: molecular targets in vitro, in vivo efficacy to inhibit tumor growth, and bioactive compounds that exert antitumor activity. Treatment of LNCaP-luc2 prostate cancer cells with SENL suppressed dihydrotestosterone-induced androgen receptor and prostate-specific antigen levels. SENL inhibited integrin ß1, calreticulin, and focal adhesion kinase activation in LNCaP-luc2 and PC3 prostate cancer cells. Oral administration of SENL significantly reduced LNCaP-luc2 xenograft tumor growth in mice with the formation of hyalinized fibrous tumor tissue, reduction in the prostate-specific antigen, and increase in AKR1C2 levels. To identify the active anticancer compounds, we fractionated SENL by high-pressure liquid chromatography and evaluated 16 peaks for cytotoxic activity. Four of the 16 peaks exhibited significant cytotoxic activity against prostate cancer cells. Mass spectrometry of the isolated peaks suggested the compounds with cytotoxic activity were nimbandiol, nimbolide, 2',3'-dihydronimbolide, and 28-deoxonimbolide. Analysis of tumor tissue and plasma samples from mice treated with SENL indicated 28-deoxonimbolide and nimbolide as the bioactive compounds. Overall, our data revealed the bioactive compounds in SENL and suggested that the anticancer activity could be mediated through alteration in androgen receptor and calreticulin levels in prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Azadirachta/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Prostatic Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Focal Adhesions/drug effects , Humans , Inhibitory Concentration 50 , Male , Mice , Plant Extracts/pharmacokinetics , Plant Extracts/toxicity , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Res ; 74(6): 1870-1880, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24480624

ABSTRACT

Overexpression of the histone acetyltransferase p300 is implicated in the proliferation and progression of prostate cancer, but evidence of a causal role is lacking. In this study, we provide genetic evidence that this generic transcriptional coactivator functions as a positive modifier of prostate tumorigenesis. In a mouse model of PTEN deletion-induced prostate cancer, genetic ablation of p300 attenuated expression of the androgen receptor (AR). This finding was confirmed in human prostate cancer cells in which PTEN expression was abolished by RNA interference-mediated attenuation. These results were consistent with clinical evidence that the expression of p300 and AR correlates positively in human prostate cancer specimens. Mechanistically, PTEN inactivation increased AR phosphorylation at serine 81 (Ser81) to promote p300 binding and acetylation of AR, thereby precluding its polyubiquitination and degradation. In support of these findings, in PTEN-deficient prostate cancer in the mouse, we found that p300 was crucial for AR target gene expression. Taken together, our work identifies p300 as a molecular determinant of AR degradation and highlights p300 as a candidate target to manage prostate cancer, especially in cases marked by PTEN loss.


Subject(s)
Carcinogenesis/metabolism , PTEN Phosphohydrolase/genetics , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Neoplasms/enzymology , Receptors, Androgen/metabolism , p300-CBP Transcription Factors/physiology , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Female , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , PTEN Phosphohydrolase/deficiency , Phosphorylation , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Proteolysis , Transcription, Genetic , Ubiquitination
13.
Cancer Res ; 74(7): 2050-61, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24491799

ABSTRACT

Despite the high incidence and mortality of prostate cancer, the etiology of this disease is not fully understood. In this study, we develop functional evidence for CBP and PTEN interaction in prostate cancer based on findings of their correlate expression in the human disease. Cbp(pc-/-);Pten(pc+/-) mice exhibited higher cell proliferation in the prostate and an early onset of high-grade prostatic intraepithelial neoplasia. Levels of EZH2 methyltransferase were increased along with its Thr350 phosphorylation in both mouse Cbp(-/-); Pten(+/-) and human prostate cancer cells. CBP loss and PTEN deficiency cooperated to trigger a switch from K27-acetylated histone H3 to K27-trimethylated bulk histones in a manner associated with decreased expression of the growth inhibitory EZH2 target genes DAB2IP, p27(KIP1), and p21(CIP1). Conversely, treatment with the histone deacetylase inhibitor panobinostat reversed this switch, in a manner associated with tumor suppression in Cbp(pc-/-);Pten(pc+/-) mice. Our findings show how CBP and PTEN interact to mediate tumor suppression in the prostate, establishing a central role for histone modification in the etiology of prostate cancer and providing a rationale for clinical evaluation of epigenetic-targeted therapy in patients with prostate cancer.


Subject(s)
Epigenesis, Genetic , Haploinsufficiency , PTEN Phosphohydrolase/genetics , Peptide Fragments/physiology , Prostatic Neoplasms/genetics , Sialoglycoproteins/physiology , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Enhancer of Zeste Homolog 2 Protein , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Male , Membrane Proteins/physiology , Mice , PTEN Phosphohydrolase/physiology , Panobinostat , Peptide Fragments/genetics , Phosphoproteins/physiology , Polycomb Repressive Complex 2/physiology , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins c-akt/physiology , Sialoglycoproteins/genetics , ras GTPase-Activating Proteins/genetics
14.
Asian J Urol ; 1(1): 2, 2014 Oct.
Article in English | MEDLINE | ID: mdl-29511630
15.
Int J Mol Sci ; 14(7): 14833-59, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23863692

ABSTRACT

The androgen receptor (AR) is important in the development of the prostate by regulating transcription, cellular proliferation, and apoptosis. AR undergoes posttranslational modifications that alter its transcription activity, translocation to the nucleus and stability. The posttranslational modifications that regulate these events are of utmost importance to understand the functional role of AR and its activity. The majority of these modifications occur in the activation function-1 (AF1) region of the AR, which contains the transcriptional activation unit 1 (TAU1) and 5 (TAU5). Identification of the modifications that occur to these regions may increase our understanding of AR activation in prostate cancer and the role of AR in the progression from androgen-dependent to castration-resistant prostate cancer (CRPC). Most of the posttranslational modifications identified to date have been determined using the full-length AR in androgen dependent cells. Further investigations into the role of posttranslational modifications in androgen-independent activation of full-length AR and constitutively active splicing variants are warranted, findings from which may provide new therapeutic options for CRPC.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Acetylation , Humans , Male , Methylation , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational , Protein Structure, Tertiary , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Sumoylation , Ubiquitination
16.
Curr Drug Targets ; 14(4): 401-7, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23565753

ABSTRACT

Androgens and the androgen receptor have been the focus of prostate cancer research since the early 1940s, when Huggins and Hodges demonstrated that removal of androgens caused advanced prostate cancer to regress. Since that time, a large number of androgen deprivation therapies have been developed in an effort to cure this disease, but prostate cancer remains one of the leading causes of cancer death in males worldwide. This is due in part to the emergence of castration- recurrent prostate cancer in patients with advanced disease who have failed androgen deprivation therapy. The androgen receptor is still a major player in castration-recurrent disease, and though much has been discovered since the early work of Huggins and Hodges regarding how prostate cancer cells manage to avoid the effects of androgen deprivation, survival times for men with advanced prostate cancer have changed only modestly. Research is now directed toward delineating the mechanisms of action of the androgen receptor under castrate conditions, whether through amplification of the AR, mutation, expression of splice variants, use of alternate signaling pathways, aberrant expression and activation of coregulators, or intratumoral androgen biosynthesis. Genome-wide association studies are also adding to the wealth of knowledge surrounding the androgen receptor, and with this knowledge comes the ability to design new drug therapies directed toward eradication of this disease.


Subject(s)
Androgen Antagonists/therapeutic use , Prostatic Neoplasms/drug therapy , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Androgens/genetics , Androgens/metabolism , Animals , Genome-Wide Association Study/methods , Humans , Male , Mutation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Signal Transduction
17.
Cancer Lett ; 335(1): 136-44, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23402817

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising therapeutic agent for prostate cancer because it selectively induces apoptosis in cancer cells but not in normal cells. Previous reports have suggested that androgens regulate TRAIL-induced apoptosis in prostate cancer cells. However, there are discrepancies between these reports of how androgens affect TRAIL-induced cell death. To clarify the role of androgens on TRAIL-induced apoptosis in prostate cancer cells, we investigated the effects of androgen on TRAIL-induced cell death in a dose-response manner. Our results showed that although androgens sensitize LNCaP cells to TRAIL-induced apoptosis, this effect is dose-dependent and biphasic. We found that low levels of androgen are superior to high levels of androgen in term of sensitizing LNCaP cells to TRAIL. We also found that upregulation of DR5 (TRAIL-R2) expression by androgens is critical for sensitizing LNCaP cells to TRAIL. However, low levels of androgen are sufficient to induce DR5 expression and sensitize LNCaP cells to TRAIL-induced cell death. High levels of androgen alter the TRADD/RIP1 ratio, which may contribute to NF-κB activation and sequentially inhibit TRAIL-induced apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Metribolone/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Testosterone Congeners/pharmacology , Androstadienes/pharmacology , Apoptosis , Cell Cycle/drug effects , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Male , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , Prostatic Neoplasms , RNA-Binding Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Death Domain Protein/metabolism , Testosterone Congeners/physiology , Wortmannin
18.
Cancer Cell Int ; 13(1): 4, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23368843

ABSTRACT

BACKGROUND: TWIST1 plays a key role in EMT-mediated tumor invasion and metastasis. Since bone metastasis is a hallmark of advanced prostate cancer and is detected in at least 85% of patients who die of this disease, it is of great importance to understand the regulation of the cellular signaling pathways involved in the metastatic process. METHODS: Prostatic cell lines were analyzed using real time RT-PCR, chromatin immunoprecipitations (ChIP) and transfection of siRNA's and reporter constructs. RESULTS: We report in this paper that TWIST1 is an androgen-regulated gene under tight regulation of NKX3-1. Androgens repress the expression of TWIST1 via NKX3-1, which is a prostate-specific tumor suppressor that is down-regulated in the majority of metastatic prostate tumors. We show that NKX3-1 binds to the TWIST1 promoter and that NKX3-1 over-expression reduces the activity of a TWIST1 promoter reporter construct, whereas NKX3-1 siRNA up-regulates endogenous TWIST1 mRNA in prostate cancer cells. CONCLUSION: Our finding that NKX3-1 represses TWIST1 expression emphasizes the functional importance of NKX3-1 in regulating TWIST1 expression during prostate cancer progression to metastatic disease.

19.
Cancer Res ; 73(1): 160-71, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23149913

ABSTRACT

A major contributor to cancer mortality is recurrence and subsequent metastatic transformation following therapeutic intervention. Therefore, in order to develop new treatment modalities and improve the efficacy of current ones, it is important to understand the molecular mechanisms that promote resistance to therapy in cancer cells. One pathway contributing to therapy resistance is autophagy, a self-digestive process that can eliminate unnecessary or damaged organelles to protect cancer cells from death. We have found that the VEGF-C/NRP-2 axis is involved in the activation of autophagy, which helps cancer cell survival following treatment. Inhibition of mTOR complex 1 activity by this axis is the underlying mechanism for the activation of autophagy. Furthermore, we identified two VEGF-C/NRP-2-regulated genes, LAMP-2 and WDFY-1, that have previously been suggested to participate in autophagy and vesicular trafficking. Upregulation of WDFY-1 following VEGF-C or NRP-2 depletion contributes to cytotoxic drug-mediated cell death. Together, these data suggest a link between the VEGF-C/NRP-2 axis and cancer cell survival despite the presence of chemotherapy-induced stress. Effective targeting of this pathway may lead to the development of new cancer therapies.


Subject(s)
Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Neoplasms/metabolism , Neuropilin-2/metabolism , Vascular Endothelial Growth Factor C/metabolism , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic/genetics , Humans , Microscopy, Confocal , Neoplasms/genetics , Neuropilin-2/genetics , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Transfection , Vascular Endothelial Growth Factor C/genetics
20.
Clin Cancer Res ; 18(10): 2882-95, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22589488

ABSTRACT

PURPOSE: The aim of this study was to investigate the promoter hypermethylation as diagnostic markers to detect malignant prostate cells and as prognostic markers to predict the clinical recurrence of prostate cancer. EXPERIMENTAL DESIGN: DNA was isolated from prostate cancer and normal adjacent tissues. After bisulfite conversion, methylation of 14,495 genes was evaluated using the Methylation27 microarrays in 238 prostate tissues. We analyzed methylation profiles in four different groups: (i) tumor (n = 198) versus matched normal tissues (n = 40), (ii) recurrence (n = 123) versus nonrecurrence (n = 75), (iii) clinical recurrence (n = 80) versus biochemical recurrence (n = 43), and (iv) systemic recurrence (n = 36) versus local recurrence (n = 44). Group 1, 2, 3, and 4 genes signifying biomarkers for diagnosis, prediction of recurrence, clinical recurrence, and systemic progression were determined. Univariate and multivariate analyses were conducted to predict risk of recurrence. We validated the methylation of genes in 20 independent tissues representing each group by pyrosequencing. RESULTS: Microarray analysis revealed significant methylation of genes in four different groups of prostate cancer tissues. The sensitivity and specificity of methylation for 25 genes from 1, 2, and 4 groups and 7 from group 3 were shown. Validation of genes by pyrosequencing from group 1 (GSTP1, HIF3A, HAAO, and RARß), group 2 (CRIP1, FLNC, RASGRF2, RUNX3, and HS3ST2), group 3 (PHLDA3, RASGRF2, and TNFRSF10D), and group 4 (BCL11B, POU3F3, and RASGRF2) confirmed the microarray results. CONCLUSIONS: Our study provides a global assessment of DNA methylation in prostate cancer and identifies the significance of genes as diagnostic and progression biomarkers of prostate cancer.


Subject(s)
DNA Methylation , DNA, Neoplasm/chemistry , Neoplasm Recurrence, Local/diagnosis , Prostatic Neoplasms/genetics , Biomarkers, Tumor/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Prognosis , Promoter Regions, Genetic , Prostatic Neoplasms/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...