Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 3(8): 1447-1459, 2023 08.
Article in English | MEDLINE | ID: mdl-37546702

ABSTRACT

Although recent efforts have led to the development of highly effective androgen receptor (AR)-directed therapies for the treatment of advanced prostate cancer, a significant subset of patients will progress with resistant disease including AR-negative tumors that display neuroendocrine features [neuroendocrine prostate cancer (NEPC)]. On the basis of RNA sequencing (RNA-seq) data from a clinical cohort of tissue from benign prostate, locally advanced prostate cancer, metastatic castration-resistant prostate cancer and NEPC, we developed a multi-step bioinformatics pipeline to identify NEPC-specific, overexpressed gene transcripts that encode cell surface proteins. This included the identification of known NEPC surface protein CEACAM5 as well as other potentially targetable proteins (e.g., HMMR and CESLR3). We further showed that cadherin EGF LAG seven-pass G-type receptor 3 (CELSR3) knockdown results in reduced NEPC tumor cell proliferation and migration in vitro. We provide in vivo data including laser capture microdissection followed by RNA-seq data supporting a causal role of CELSR3 in the development and/or maintenance of the phenotype associated with NEPC. Finally, we provide initial data that suggests CELSR3 is a target for T-cell redirection therapeutics. Further work is now needed to fully evaluate the utility of targeting CELSR3 with T-cell redirection or other similar therapeutics as a potential new strategy for patients with NEPC. Significance: The development of effective treatment for patients with NEPC remains an unmet clinical need. We have identified specific surface proteins, including CELSR3, that may serve as novel biomarkers or therapeutic targets for NEPC.


Subject(s)
Neuroendocrine Tumors , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Neuroendocrine Tumors/genetics , Prostate/metabolism , Cell Membrane/metabolism , Cadherins/genetics
2.
Bioorg Med Chem ; 87: 117275, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37156065

ABSTRACT

Achondroplasia is a rare disease affecting bone growth and is caused by a missense mutation in the fibroblast growth factor receptor 3 (FGFR3) gene. In the past few years, there were multiple experimental drugs entering into clinical trials for treating achondroplasia including vosoritide, the first precision medicine approved for this indication. This perspective presents the mechanism of action, benefit, and potential mechanistic limitation of the drugs currently being evaluated in clinical trials for achondroplasia. This article also discusses the potential impact of those drugs not only in increasing the growth of individuals living with achondroplasia but also in improving their quality of life.


Subject(s)
Achondroplasia , Precision Medicine , Humans , Quality of Life , Achondroplasia/drug therapy , Achondroplasia/genetics , Mutation
3.
J Clin Invest ; 129(9): 3924-3940, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31260412

ABSTRACT

Despite recent therapeutic advances, prostate cancer remains a leading cause of cancer-related death. A subset of castration resistant prostate cancers become androgen receptor (AR) signaling-independent and develop neuroendocrine prostate cancer (NEPC) features through lineage plasticity. These NEPC tumors, associated with aggressive disease and poor prognosis, are driven, in part, by aberrant expression of N-Myc, through mechanisms that remain unclear. Integrative analysis of the N-Myc transcriptome, cistrome and interactome using in vivo, in vitro and ex vivo models (including patient-derived organoids) identified a lineage switch towards a neural identity associated with epigenetic reprogramming. N-Myc and known AR-co-factors (e.g., FOXA1 and HOXB13) overlapped, independently of AR, at genomic loci implicated in neural lineage specification. Moreover, histone marks specifically associated with lineage-defining genes were reprogrammed by N-Myc. We also demonstrated that the N-Myc-induced molecular program accurately classifies our cohort of patients with advanced prostate cancer. Finally, we revealed the potential for EZH2 inhibition to reverse the N-Myc-induced suppression of epithelial lineage genes. Altogether, our data provide insights on how N-Myc regulates lineage plasticity and epigenetic reprogramming associated with lineage-specification. The N-Myc signature we defined could also help predict the evolution of prostate cancer and thus better guide the choice of future therapeutic strategies.


Subject(s)
Cell Lineage , Epigenesis, Genetic , N-Myc Proto-Oncogene Protein/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Plasticity , DNA/chemistry , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Transplantation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Signal Transduction , Transcriptome
4.
Sci Transl Med ; 11(484)2019 03 20.
Article in English | MEDLINE | ID: mdl-30894499

ABSTRACT

Histologic transformation to small cell neuroendocrine prostate cancer occurs in a subset of patients with advanced prostate cancer as a mechanism of treatment resistance. Rovalpituzumab tesirine (SC16LD6.5) is an antibody-drug conjugate that targets delta-like protein 3 (DLL3) and was initially developed for small cell lung cancer. We found that DLL3 is expressed in most of the castration-resistant neuroendocrine prostate cancer (CRPC-NE) (36 of 47, 76.6%) and in a subset of castration-resistant prostate adenocarcinomas (7 of 56, 12.5%). It shows minimal to no expression in localized prostate cancer (1 of 194) and benign prostate (0 of 103). DLL3 expression correlates with neuroendocrine marker expression, RB1 loss, and aggressive clinical features. DLL3 in circulating tumor cells was concordant with matched metastatic biopsy (87%). Treatment of DLL3-expressing prostate cancer xenografts with a single dose of SC16LD6.5 resulted in complete and durable responses, whereas DLL3-negative models were insensitive. We highlight a patient with neuroendocrine prostate cancer with a meaningful clinical and radiologic response to SC16LD6.5 when treated on a phase 1 trial. Overall, our findings indicate that DLL3 is preferentially expressed in CRPC-NE and provide rationale for targeting DLL3 in patients with DLL3-positive metastatic prostate cancer.


Subject(s)
Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Molecular Targeted Therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Aged , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Benzodiazepinones/pharmacology , Benzodiazepinones/therapeutic use , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Time Factors , Treatment Outcome
5.
Clin Cancer Res ; 25(1): 43-51, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30232224

ABSTRACT

PURPOSE: Neuroendocrine prostate cancer (NEPC) is an aggressive variant of prostate cancer that may develop de novo or as a mechanism of treatment resistance. N-myc is capable of driving NEPC progression. Alisertib inhibits the interaction between N-myc and its stabilizing factor Aurora-A, inhibiting N-myc signaling, and suppressing tumor growth. PATIENTS AND METHODS: Sixty men were treated with alisertib 50 mg twice daily for 7 days every 21 days. Eligibility included metastatic prostate cancer and at least one: small-cell neuroendocrine morphology; ≥50% neuroendocrine marker expression; new liver metastases without PSA progression; or elevated serum neuroendocrine markers. The primary endpoint was 6-month radiographic progression-free survival (rPFS). Pretreatment biopsies were evaluated by whole exome and RNA-seq and patient-derived organoids were developed. RESULTS: Median PSA was 1.13 ng/mL (0.01-514.2), number of prior therapies was 3, and 68% had visceral metastases. Genomic alterations involved RB1 (55%), TP53 (46%), PTEN (29%), BRCA2 (29%), and AR (27%), and there was a range of androgen receptor signaling and NEPC marker expression. Six-month rPFS was 13.4% and median overall survival was 9.5 months (7.3-13). Exceptional responders were identified, including complete resolution of liver metastases and prolonged stable disease, with tumors suggestive of N-myc and Aurora-A overactivity. Patient organoids exhibited concordant responses to alisertib and allowed for the dynamic testing of Aurora-N-myc complex disruption. CONCLUSIONS: Although the study did not meet its primary endpoint, a subset of patients with advanced prostate cancer and molecular features supporting Aurora-A and N-myc activation achieved significant clinical benefit from single-agent alisertib.


Subject(s)
Aurora Kinase A/genetics , Azepines/administration & dosage , Carcinoma, Neuroendocrine/drug therapy , N-Myc Proto-Oncogene Protein/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Pyrimidines/administration & dosage , Aged , Aged, 80 and over , Aurora Kinase A/antagonists & inhibitors , Azepines/adverse effects , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Disease Progression , Humans , Male , Middle Aged , Orchiectomy , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Pyrimidines/adverse effects , Receptors, Androgen/genetics , Signal Transduction/drug effects
6.
Cancer Cell ; 30(4): 563-577, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27728805

ABSTRACT

The transition from castration-resistant prostate adenocarcinoma (CRPC) to neuroendocrine prostate cancer (NEPC) has emerged as an important mechanism of treatment resistance. NEPC is associated with overexpression and gene amplification of MYCN (encoding N-Myc). N-Myc is an established oncogene in several rare pediatric tumors, but its role in prostate cancer progression is not well established. Integrating a genetically engineered mouse model and human prostate cancer transcriptome data, we show that N-Myc overexpression leads to the development of poorly differentiated, invasive prostate cancer that is molecularly similar to human NEPC. This includes an abrogation of androgen receptor signaling and induction of Polycomb Repressive Complex 2 signaling. Altogether, our data establishes N-Myc as an oncogenic driver of NEPC.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Animals , Azepines/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Genes, myc , Heterografts , Humans , Male , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/biosynthesis , N-Myc Proto-Oncogene Protein/metabolism , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Transcription, Genetic
7.
Cell Rep ; 15(11): 2348-56, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264179

ABSTRACT

Mutations in transcription factor (TF) genes are frequently observed in tumors, often leading to aberrant transcriptional activity. Unfortunately, TFs are often considered undruggable due to the absence of targetable enzymatic activity. To address this problem, we developed CRAFTT, a computational drug-repositioning approach for targeting TF activity. CRAFTT combines ChIP-seq with drug-induced expression profiling to identify small molecules that can specifically perturb TF activity. Application to ENCODE ChIP-seq datasets revealed known drug-TF interactions, and a global drug-protein network analysis supported these predictions. Application of CRAFTT to ERG, a pro-invasive, frequently overexpressed oncogenic TF, predicted that dexamethasone would inhibit ERG activity. Dexamethasone significantly decreased cell invasion and migration in an ERG-dependent manner. Furthermore, analysis of electronic medical record data indicates a protective role for dexamethasone against prostate cancer. Altogether, our method provides a broadly applicable strategy for identifying drugs that specifically modulate TF activity.


Subject(s)
Computer Simulation , Drug Repositioning/methods , Oncogenes , Transcription Factors/metabolism , Azepines/pharmacology , Cell Line, Tumor , Dexamethasone/pharmacology , Electronic Health Records , Humans , Kaplan-Meier Estimate , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/metabolism , Small Molecule Libraries/pharmacology , Triazoles/pharmacology
8.
Cell Rep ; 7(6): 1900-13, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24910439

ABSTRACT

The RNA helicases DDX5 and DDX17 are members of a large family of highly conserved proteins that are involved in gene-expression regulation; however, their in vivo targets and activities in biological processes such as cell differentiation, which requires reprogramming of gene-expression programs at multiple levels, are not well characterized. Here, we uncovered a mechanism by which DDX5 and DDX17 cooperate with heterogeneous nuclear ribonucleoprotein (hnRNP) H/F splicing factors to define epithelial- and myoblast-specific splicing subprograms. We then observed that downregulation of DDX5 and DDX17 protein expression during myogenesis and epithelial-to-mesenchymal transdifferentiation contributes to the switching of splicing programs during these processes. Remarkably, this downregulation is mediated by the production of miRNAs induced upon differentiation in a DDX5/DDX17-dependent manner. Since DDX5 and DDX17 also function as coregulators of master transcriptional regulators of differentiation, we propose to name these proteins "master orchestrators" of differentiation that dynamically orchestrate several layers of gene expression.


Subject(s)
DEAD-box RNA Helicases/genetics , MicroRNAs/genetics , Alternative Splicing , Animals , Cell Differentiation/genetics , DEAD-box RNA Helicases/metabolism , Down-Regulation , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/genetics , Exons , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , MCF-7 Cells , Mice , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Myoblasts/enzymology , Myoblasts/metabolism , Myoblasts/physiology , Transcription, Genetic
9.
Nucleic Acids Res ; 42(4): 2197-207, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275493

ABSTRACT

Estrogen and androgen receptors (ER and AR) play key roles in breast and prostate cancers, respectively, where they regulate the transcription of large arrays of genes. The activities of ER and AR are controlled by large networks of protein kinases and transcriptional coregulators, including Ddx5 and its highly related paralog Ddx17. The Ddx5 and Ddx17 RNA helicases are also splicing regulators. Here, we report that Ddx5 and Ddx17 are master regulators of the estrogen- and androgen-signaling pathways by controlling transcription and splicing both upstream and downstream of the receptors. First, Ddx5 and Ddx17 are required downstream of ER and AR for the transcriptional and splicing regulation of a large number of steroid hormone target genes. Second, Ddx5 and Ddx17 act upstream of ER and AR by controlling the expression, at the splicing level, of several key regulators of ER and AR activities. Of particular interest, we demonstrate that Ddx5 and Ddx17 control alternative splicing of the GSK3ß kinase, which impacts on both ER and AR protein stability. We also provide a freely available online resource which gives information regarding splicing variants of genes involved in the estrogen- and androgen-signaling pathways.


Subject(s)
Alternative Splicing , Androgens/pharmacology , DEAD-box RNA Helicases/metabolism , Estrogens/pharmacology , Signal Transduction , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , MCF-7 Cells , Protein Stability , Receptors, Androgen/metabolism
10.
Nat Struct Mol Biol ; 19(11): 1139-46, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23022728

ABSTRACT

Both epigenetic and splicing regulation contribute to tumor progression, but the potential links between these two levels of gene-expression regulation in pathogenesis are not well understood. Here, we report that the mouse and human RNA helicases Ddx17 and Ddx5 contribute to tumor-cell invasiveness by regulating alternative splicing of several DNA- and chromatin-binding factors, including the macroH2A1 histone. We show that macroH2A1 splicing isoforms differentially regulate the transcription of a set of genes involved in redox metabolism. In particular, the SOD3 gene that encodes the extracellular superoxide dismutase and plays a part in cell migration is regulated in an opposite manner by macroH2A1 splicing isoforms. These findings reveal a new regulatory pathway in which splicing factors control the expression of histone variant isoforms that in turn drive a transcription program to switch tumor cells to an invasive phenotype.


Subject(s)
Alternative Splicing/genetics , DEAD-box RNA Helicases/metabolism , Epigenesis, Genetic/physiology , Gene Expression Regulation, Neoplastic/physiology , Histones/genetics , Neoplasm Invasiveness/genetics , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers/genetics , Humans , Mice , Neoplasm Invasiveness/physiopathology , ROC Curve , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
11.
Nat Struct Mol Biol ; 17(11): 1358-66, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20972445

ABSTRACT

Pre-mRNA splicing is functionally coupled to transcription, and genotoxic stresses can enhance alternative exon inclusion by affecting elongating RNA polymerase II. We report here that various genotoxic stress inducers, including camptothecin (CPT), inhibit the interaction between Ewing's sarcoma proto-oncoprotein (EWS), an RNA polymerase II-associated factor, and YB-1, a spliceosome-associated factor. This results in the cotranscriptional skipping of several exons of the MDM2 gene, which encodes the main p53 ubiquitin ligase. This reversible exon skipping participates in the regulation of MDM2 expression that may contribute to the accumulation of p53 during stress exposure and its rapid shut-off when stress is removed. Finally, a splicing-sensitive microarray identified numerous exons that are skipped in response to CPT and EWS-YB-1 depletion. These data demonstrate genotoxic stress-induced alteration of the communication between the transcriptional and splicing machineries, which results in widespread exon skipping and plays a central role in the genotoxic stress response.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exons/genetics , Models, Genetic , Nuclear Proteins/metabolism , RNA-Binding Protein EWS/metabolism , Camptothecin/pharmacology , Cell Line , DNA Polymerase II/metabolism , Gene Expression Regulation , Humans , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Splicing/physiology , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Y-Box-Binding Protein 1
12.
Cancer Res ; 70(9): 3760-70, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20406972

ABSTRACT

Alternative promoters (AP) occur in >30% protein-coding genes and contribute to proteome diversity. However, large-scale analyses of AP regulation are lacking, and little is known about their potential physiopathologic significance. To better understand the transcriptomic effect of estrogens, which play a major role in breast cancer, we analyzed gene and AP regulation by estradiol in MCF7 cells using pan-genomic exon arrays. We thereby identified novel estrogen-regulated genes (ERG) and determined the regulation of AP-encoded transcripts in 150 regulated genes. In <30% cases, APs were regulated in a similar manner by estradiol, whereas in >70% cases, they were regulated differentially. The patterns of AP regulation correlated with the patterns of estrogen receptor alpha (ERalpha) and CCCTC-binding factor (CTCF) binding sites at regulated gene loci. Interestingly, among genes with differentially regulated (DR) APs, we identified cases where estradiol regulated APs in an opposite manner, sometimes without affecting global gene expression levels. This promoter switch was mediated by the DDX5/DDX17 family of ERalpha coregulators. Finally, genes with DR promoters were preferentially involved in specific processes (e.g., cell structure and motility, and cell cycle). We show, in particular, that isoforms encoded by the NET1 gene APs, which are inversely regulated by estradiol, play distinct roles in cell adhesion and cell cycle regulation and that their expression is differentially associated with prognosis in ER(+) breast cancer. Altogether, this study identifies the patterns of AP regulation in ERGs and shows the contribution of AP-encoded isoforms to the estradiol-regulated transcriptome as well as their physiopathologic significance in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , CCCTC-Binding Factor , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Exons , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis/methods , Protein Isoforms , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...