Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Natl Cancer Inst ; 114(12): 1656-1664, 2022 12 08.
Article in English | MEDLINE | ID: mdl-36053178

ABSTRACT

BACKGROUND: Personalized genomic classifiers have transformed the management of prostate cancer (PCa) by identifying the most aggressive subsets of PCa. Nevertheless, the performance of genomic classifiers to risk classify African American men is thus far lacking in a prospective setting. METHODS: This is a prospective study of the Decipher genomic classifier for National Comprehensive Cancer Network low- and intermediate-risk PCa. Study-eligible non-African American men were matched to African American men. Diagnostic biopsy specimens were processed to estimate Decipher scores. Samples accrued in NCT02723734, a prospective study, were interrogated to determine the genomic risk of reclassification (GrR) between conventional clinical risk classifiers and the Decipher score. RESULTS: The final analysis included a clinically balanced cohort of 226 patients with complete genomic information (113 African American men and 113 non-African American men). A higher proportion of African American men with National Comprehensive Cancer Network-classified low-risk (18.2%) and favorable intermediate-risk (37.8%) PCa had a higher Decipher score than non-African American men. Self-identified African American men were twice more likely than non-African American men to experience GrR (relative risk [RR] = 2.23, 95% confidence interval [CI] = 1.02 to 4.90; P = .04). In an ancestry-determined race model, we consistently validated a higher risk of reclassification in African American men (RR = 5.26, 95% CI = 1.66 to 16.63; P = .004). Race-stratified analysis of GrR vs non-GrR tumors also revealed molecular differences in these tumor subtypes. CONCLUSIONS: Integration of genomic classifiers with clinically based risk classification can help identify the subset of African American men with localized PCa who harbor high genomic risk of early metastatic disease. It is vital to identify and appropriately risk stratify the subset of African American men with aggressive disease who may benefit from more targeted interventions.


Subject(s)
Prostatectomy , Prostatic Neoplasms , Male , Humans , Prospective Studies , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Black or African American/genetics , Genetic Testing
2.
Eur Urol ; 81(4): 325-330, 2022 04.
Article in English | MEDLINE | ID: mdl-33303244

ABSTRACT

Grade group 4 and 5 (GG-45) prostate cancer (PCa) patients are at the highest risk of lethal outcomes, yet lack genomic risk stratification for prognosis and treatment selection. Here, we assess whether transcriptomic interactions between tumor immune content score (ICS) and the Decipher genomic classifier can identify most lethal subsets of GG-45 PCa. We utilized whole transcriptome data from 8071 tumor tissue (6071 prostatectomy and 2000 treatment-naïve biopsy samples) to derive four immunogenomic subtypes using ICS and Decipher. When compared across all grade groups, GG-45 samples had the highest proportion of most aggressive subtype-ICSHigh/DecipherHigh. Subsequent analyses within the GG-45 patient samples (n = 1420) revealed that the ICSHigh/DecipherHigh subtype was associated with increased genomic radiosensitivity. Additionally, in a multivariable model (n = 335), ICSHigh/DecipherHigh subtype had a significantly higher risk of distant metastasis (hazard ratio [HR] = 5.41; 95% confidence interval [CI], 2.76-10.6; p ≤ 0.0001) and PCa-specific mortality (HR = 10.6; 95% CI, 4.18-26.94; p ≤ 0.0001) as compared with ICSLow/DecipherLow. The novel immunogenomic subtypes establish a very strong synergistic interaction between ICS and Decipher in identifying GG-45 patients who experience the most lethal outcomes. PATIENT SUMMARY: In this analysis, we identified a novel interaction between the total immune content of prostate tumors and genomic classifier to identify the most lethal subset of patients with grade groups 4 and 5. Our results will aid in the subtyping of aggressive prostate cancer patients who may benefit from combined immune-radiotherapy modalities.


Subject(s)
Prostatic Neoplasms , Transcriptome , Humans , Male , Neoplasm Grading , Prostate/pathology , Prostatectomy/adverse effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy
3.
Sci Rep ; 11(1): 9264, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33927218

ABSTRACT

Risk classification for prostate cancer (PCa) aggressiveness and underlying mechanisms remain inadequate. Interactions between single nucleotide polymorphisms (SNPs) may provide a solution to fill these gaps. To identify SNP-SNP interactions in the four pathways (the angiogenesis-, mitochondria-, miRNA-, and androgen metabolism-related pathways) associated with PCa aggressiveness, we tested 8587 SNPs for 20,729 cases from the PCa consortium. We identified 3 KLK3 SNPs, and 1083 (P < 3.5 × 10-9) and 3145 (P < 1 × 10-5) SNP-SNP interaction pairs significantly associated with PCa aggressiveness. These SNP pairs associated with PCa aggressiveness were more significant than each of their constituent SNP individual effects. The majority (98.6%) of the 3145 pairs involved KLK3. The 3 most common gene-gene interactions were KLK3-COL4A1:COL4A2, KLK3-CDH13, and KLK3-TGFBR3. Predictions from the SNP interaction-based polygenic risk score based on 24 SNP pairs are promising. The prevalence of PCa aggressiveness was 49.8%, 21.9%, and 7.0% for the PCa cases from our cohort with the top 1%, middle 50%, and bottom 1% risk profiles. Potential biological functions of the identified KLK3 SNP-SNP interactions were supported by gene expression and protein-protein interaction results. Our findings suggest KLK3 SNP interactions may play an important role in PCa aggressiveness.


Subject(s)
Kallikreins/genetics , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Biomarkers, Tumor/genetics , Epistasis, Genetic , Genetic Predisposition to Disease , Genotype , Humans , Male , Polymorphism, Single Nucleotide , Prostatic Neoplasms/pathology
4.
Methods Mol Biol ; 2194: 127-142, 2021.
Article in English | MEDLINE | ID: mdl-32926365

ABSTRACT

Bioinformatic scientists are often asked to do widespread analyses of publicly available datasets in order to identify genetic alterations in cancer for genes of interest; therefore, we sought to create a set of tools to conduct common statistical analyses of The Cancer Genome Atlas (TCGA) data. These tools have been developed in response to requests from our collaborators to ask questions, validate findings, and better understand the function of their gene of interest. We describe here what data we have used, how to obtain it, and what figures we have found useful.


Subject(s)
Databases, Genetic , Neoplasms/genetics , Translational Research, Biomedical/methods , Computational Biology , DNA Methylation , Gene Expression Regulation/genetics , Genetic Heterogeneity , Genomics , Humans , RNA-Seq , Software , Survival Analysis
5.
Clin Cancer Res ; 27(1): 320-329, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33037017

ABSTRACT

PURPOSE: The role of immune-oncologic mechanisms of racial disparities in prostate cancer remains understudied. Limited research exists to evaluate the molecular underpinnings of immune differences in African American men (AAM) and European American men (EAM) prostate tumor microenvironment (TME). EXPERIMENTAL DESIGN: A total of 1,173 radiation-naïve radical prostatectomy samples with whole transcriptome data from the Decipher GRID registry were used. Transcriptomic expressions of 1,260 immune-specific genes were selected to assess immune-oncologic differences between AAM and EAM prostate tumors. Race-specific differential expression of genes was assessed using a rank test, and intergene correlational matrix and gene set enrichment was used for pathway analysis. RESULTS: AAM prostate tumors have significant enrichment of major immune-oncologic pathways, including proinflammatory cytokines, IFNα, IFNγ, TNFα signaling, ILs, and epithelial-mesenchymal transition. AAM TME has higher total immune content score (ICSHIGH) compared with 0 (37.8% vs. 21.9%, P = 0.003). AAM tumors also have lower DNA damage repair and are genomically radiosensitive as compared with EAM. IFITM3 (IFN-inducible transmembrane protein 3) was one of the major proinflammatory genes overexpressed in AAM that predicted increased risk of biochemical recurrence selectively for AAM in both discovery [HRAAM = 2.30; 95% confidence interval (CI), 1.21-4.34; P = 0.01] and validation (HRAAM = 2.42; 95% CI, 1.52-3.86; P = 0.0001) but not in EAM. CONCLUSIONS: Prostate tumors of AAM manifest a unique immune repertoire and have significant enrichment of proinflammatory immune pathways that are associated with poorer outcomes. Observed immune-oncologic differences can aid in a genomically adaptive approach to treating prostate cancer in AAM.


Subject(s)
Black or African American/genetics , Gene Expression Regulation, Neoplastic/immunology , Neoplasm Recurrence, Local/immunology , Prostatic Neoplasms/genetics , Tumor Microenvironment/immunology , Black or African American/statistics & numerical data , Aged , Datasets as Topic , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Follow-Up Studies , Genomics/statistics & numerical data , Health Status Disparities , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/prevention & control , Prostate/immunology , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/mortality , Prostatic Neoplasms/therapy , Risk Assessment/statistics & numerical data , Tumor Microenvironment/genetics , White People/genetics , White People/statistics & numerical data
6.
Prostate ; 81(2): 109-117, 2021 02.
Article in English | MEDLINE | ID: mdl-33141952

ABSTRACT

BACKGROUND: In prostate cancer (PCa), lack of androgen receptor (AR) regulated TMPRSS2-ETS-related gene (ERG) gene fusion (ERGnegative ) status has been associated with African American race; however, the implications of ERG status for the location of dominant tumors within the prostate remains understudied. METHODS: An African American-enriched multiinstitutional cohort of 726 PCa patients consisting of both African American men (AAM; n = 254) and European American men (EAM; n = 472) was used in the analyses. Methods of categorical analysis were used. Messenger RNA (mRNA) expression differences between anterior and posterior tumor lesions were analyzed using Wilcoxon rank-sum tests with multiple comparison corrections. RESULTS: Anti-ERG immunohistochemistry staining showed that the association between ERG status and anterior tumors is independent of race and is consistently robust for both AAM (ERGnegative 81.4% vs. ERGpositive 18.6%; p = .005) and EAM (ERGnegative 60.4% vs. ERGpositive 39.6%; p < .001). In a multivariable model, anterior tumors were more likely to be IHC-ERGnegative (odds ratio [OR]: 3.20; 95% confidence interval [CI]: 2.14-4.78; p < .001). IHC-ERGnegative were also more likely to have high-grade tumors (OR: 1.73; 95% CI: 1.06-2.82; p = .02). In the exploratory genomic analysis, mRNA expression of location-dependent genes is highly influenced by ERG status and African American race. However, tumor location did not impact the expression of AR or the major canonical AR-target genes (KLK3, AMACR, and MYC). CONCLUSIONS: ERGnegative tumor status is the strongest predictor of anterior prostate tumors, regardless of race. Furthermore, AR expression and canonical AR signaling do not impact tumor location.


Subject(s)
Black or African American/genetics , Oncogene Proteins, Fusion/genetics , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Cohort Studies , Humans , Immunohistochemistry , Male , Middle Aged , Prostatic Neoplasms/chemistry , RNA, Messenger , Transcriptional Regulator ERG/analysis , Transcriptional Regulator ERG/genetics
7.
Cancer Epidemiol Biomarkers Prev ; 29(1): 246-253, 2020 01.
Article in English | MEDLINE | ID: mdl-31757784

ABSTRACT

BACKGROUND: Commercial gene expression signatures of prostate cancer prognosis were developed and validated in cohorts of predominantly European American men (EAM). Limited research exists on the value of such signatures in African American men (AAM), who have poor prostate cancer outcomes. We explored differences in gene expression between EAM and AAM for three commercially available panels recommended by the National Comprehensive Cancer Network for prostate cancer prognosis. METHODS: A total of 232 EAM and 95 AAM patients provided radical prostatectomy specimens. Gene expression was quantified using NanoString for 60 genes spanning the Oncotype DX Prostate, Prolaris, and Decipher panels. A continuous expression-based risk score was approximated for each. Differential expression, intrapanel coexpression, and risk by race were assessed. RESULTS: Clinical and pathologic features were similar between AAM and EAM. Differential expression by race was observed for 48% of genes measured, although the magnitudes of expression differences were small. Coexpression patterns were more strongly preserved by race group for Oncotype DX and Decipher than Prolaris. Poorer prognosis was estimated in EAM versus AAM for Oncotype DX (P < 0.001), whereas negligible prognostic differences were predicted between AAM and EAM using Prolaris or Decipher (P > 0.05). CONCLUSIONS: Because of observed racial differences across three commercial gene expression panels for prostate cancer prognosis, caution is warranted when applying these panels in clinical decision-making in AAM. IMPACT: Differences in gene expression by race for three commercial panels for prostate cancer prognosis indicate that further study of their effectiveness in AAM with long-term follow-up is warranted.


Subject(s)
Biomarkers, Tumor/genetics , Black or African American/genetics , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/genetics , Transcriptome , White People/genetics , Humans , Male , Neoplasm Grading , Neoplasm Staging , Predictive Value of Tests , Prostatic Neoplasms/pathology , Race Factors
8.
J Urol ; 202(2): 247-255, 2019 08.
Article in English | MEDLINE | ID: mdl-31107158

ABSTRACT

PURPOSE: Most prostate cancer in African American men lacks the ETS (E26 transforming specific) family fusion event (ETS-). We aimed to establish clinically relevant biomarkers in African American men by studying ETS dependent gene expression patterns to identified race specific genes predictive of outcomes. MATERIALS AND METHODS: Two multicenter cohorts of a total of 1,427 men were used for the discovery and validation (635 and 792 men, respectively) of race specific predictive biomarkers. We used false discovery rate adjusted q values to identify race and ETS dependent genes which were differentially expressed in African American men who experienced biochemical recurrence within 5 years. Principal component modeling along with survival analysis was done to assess the accuracy of the gene panel in predicting recurrence. RESULTS: We identified 3,047 genes which were differentially expressed based on ETS status. Of these genes 362 were differentially expressed in a race specific manner (false discovery rate 0.025 or less). A total of 81 genes were race specific and over expressed in African American men who experienced biochemical recurrence. The final gene panel included APOD, BCL6, EMP1, MYADM, SRGN and TIMP3. These genes were associated with 5-year biochemical recurrence (HR 1.97, 95% CI 1.27-3.06, p = 0.002) and they improved the predictive accuracy of clinicopathological variables only in African American men (60-month time dependent AUC 0.72). CONCLUSIONS: In an effort to elucidate biological features associated with prostate cancer aggressiveness in African American men we identified ETS dependent biomarkers predicting early onset biochemical recurrence only in African American men. Thus, these ETS dependent biomarkers representing ideal candidates for biomarkers of aggressive disease in this patient population.


Subject(s)
Black or African American/genetics , Prostatic Neoplasms/genetics , Aged , Biomarkers, Tumor/genetics , Cohort Studies , Gene Expression Regulation , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Prognosis , Proto-Oncogene Proteins c-ets/genetics
9.
Prostate Cancer Prostatic Dis ; 22(2): 292-302, 2019 05.
Article in English | MEDLINE | ID: mdl-30367117

ABSTRACT

BACKGROUND: Prostate cancer (PCa) tumors harboring translocations of ETS family genes with the androgen responsive TMPRSS2 gene (ETS+ tumors) provide a robust biomarker for detecting PCa in approximately 70% of patients. ETS+ PCa express high levels of the androgen receptor (AR), yet PCa tumors lacking ETS fusions (ETS-) also express AR and demonstrate androgen-regulated growth. In this study, we evaluate the differences in the AR-regulated transcriptomes between ETS+ and ETS- PCa tumors. METHODS: 10,608 patient tumors from three independent PCa datasets classified as ETS+ (samples overexpressing ERG or other ETS family members) or ETS- (all other PCa) were analyzed for differential gene expression using false-discovery-rate adjusted methods and gene-set enrichment analysis (GSEA). RESULTS: Based on the expression of AR-dependent genes and an unsupervised Principal Component Analysis (PCA) model, AR-regulated gene expression alone was able to separate PCa samples into groups based on ETS status in all PCa databases. ETS status distinguished several differentially expressed genes in both TCGA (6.9%) and GRID (6.6%) databases, with 413 genes overlapping in both databases. Importantly, GSEA showed enrichment of distinct androgen-responsive genes in both ETS- and ETS+ tumors, and AR ChIP-seq data identified 131 direct AR-target genes that are regulated in an ETS-specific fashion. Notably, dysregulation of ETS-dependent AR-target genes within the metabolic and non-canonical WNT pathways was associated with clinical outcomes. CONCLUSIONS: ETS status influences the transcriptional repertoire of the AR, and ETS- PCa tumors appear to rely on distinctly different AR-dependent transcriptional programs to drive and sustain tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-ets/genetics , Receptors, Androgen/metabolism , Transcriptome , Cell Line, Tumor , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Humans , Male , Neoplasm Grading , Neoplasm Staging , Prostatic Neoplasms/pathology
10.
Cancer Epidemiol Biomarkers Prev ; 27(11): 1376-1383, 2018 11.
Article in English | MEDLINE | ID: mdl-30108099

ABSTRACT

Background: We studied the utility of the tumor suppressor Tristetraprolin (TTP, ZFP36) as a clinically relevant biomarker of aggressive disease in prostate cancer patients after radical prostatectomy (RP).Methods: TTP RNA expression was measured in an RP cohort of patients treated at Moffitt Cancer Center (MCC) and obtained from six publically available RP datasets with biochemical recurrence (BCR; total n = 1,394) and/or metastatic outcome data (total n = 1,222). TTP protein expression was measured by immunohistochemistry in a tissue microarray of 153 MCC RP samples. The time to BCR or metastasis based on TTP RNA or protein levels was calculated using the Kaplan-Meier analysis. Univariable and multivariable Cox proportional hazard models were performed on multiple cohorts to evaluate if TTP is a clinically relevant biomarker and to assess if TTP improves upon the Cancer of the Prostate Risk Assessment postsurgical (CAPRA-S) score for predicting clinical outcomes.Results: In all of the RP patient cohorts, prostate cancer with low TTP RNA or protein levels had decreased time to BCR or metastasis versus TTP-high tumors. Further, the decreased time to BCR in TTP-low prostate cancer was more pronounced in low-grade tumors. Finally, pooled survival analysis suggests that TTP RNA expression provides independent information beyond CAPRA-S to predict BCR.Conclusions: TTP is a promising prostate cancer biomarker for predicting which RP patients will have poor outcomes, especially for low-grade prostate cancer patients.Impact: This study suggests that TTP RNA expression can be used to enhance the accuracy of CAPRA-S to predict outcomes in patients treated with RP. Cancer Epidemiol Biomarkers Prev; 27(11); 1376-83. ©2018 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , Prostatic Neoplasms/genetics , Tristetraprolin/genetics , Humans , Male , Prostatic Neoplasms/pathology , Risk Factors , Treatment Outcome
11.
BMC Urol ; 17(1): 18, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28320379

ABSTRACT

Prostate cancer is the most common malignancy among men in the US. Though considerable improvement in the diagnosis of prostate cancer has been achieved in the past decade, predicting disease outcome remains a major clinical challenge. Recent expression profiling studies in prostate cancer suggest microRNAs (miRNAs) may serve as potential biomarkers for prostate cancer risk and disease progression. miRNAs comprise a large family of about 22-nucleotide-long non-protein coding RNAs, regulate gene expression post-transcriptionally and participate in the regulation of numerous cellular processes. In this review, we discuss the current status of miRNA in studies evaluating the disease progression of prostate cancer. The discussion highlights key findings from previous studies, which reported the role of miRNAs in risk and progression of prostate cancer, providing an understanding of the influence of miRNA on prostate cancer. Our review indicates that somewhat consistent results exist between these studies and reports on several prostate cancer related miRNAs. Present promising candidates are miR-1, -21, 106b, 141, -145, -205, -221, and -375, which are the most frequently studied and seem to be the most promising for diagnosis and prognosis for prostate cancer. Nevertheless, the findings from previous studies suggest miRNAs may play an important role in the risk and progression of prostate cancer as promising biomarkers.


Subject(s)
Disease Progression , MicroRNAs , Prostatic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/metabolism
12.
Oncotarget ; 7(50): 83462-83475, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27825143

ABSTRACT

Tristetraprolin (TTP) is an RNA-binding protein that post-transcriptionally suppresses gene expression by delivering mRNA cargo to processing bodies (P-bodies) where the mRNA is degraded. TTP functions as a tumor suppressor in a mouse model of B cell lymphoma, and in some human malignancies low TTP expression correlates with reduced survival. Here we report important prognostic and functional roles for TTP in human prostate cancer. First, gene expression analysis of prostate tumors revealed low TTP expression correlates with patients having high-risk Gleason scores and increased biochemical recurrence. Second, in prostate cancer cells with low levels of endogenous TTP, inducible TTP expression inhibits their growth and proliferation, as well as their clonogenic growth. Third, TTP functions as a tumor suppressor in prostate cancer, as forced TTP expression markedly impairs the tumorigenic potential of prostate cancer cells in a mouse xenograft model. Finally, pathway analysis of gene expression data suggested metabolism is altered by TTP expression in prostate tumor cells, and metabolic analyses revealed that such processes are impaired by TTP, including mitochondrial respiration. Collectively, these findings suggest that TTP is an important prognostic indicator for prostate cancer, and augmenting TTP function would effectively disable the metabolism and proliferation of aggressive prostate tumors.


Subject(s)
Cell Proliferation , Energy Metabolism , Prostatic Neoplasms/metabolism , Tristetraprolin/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Energy Metabolism/drug effects , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Kallikreins/blood , Male , Mice, Nude , Neoplasm Grading , Prostate-Specific Antigen/blood , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction , Time Factors , Tristetraprolin/genetics , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
13.
PLoS One ; 9(12): e115517, 2014.
Article in English | MEDLINE | ID: mdl-25541715

ABSTRACT

The RNA-binding protein Tristetraprolin (TTP, ZFP36) functions as a tumor suppressor that impairs the development and disables the maintenance of MYC-driven lymphoma. In addition, other human cancers expressed reduced levels of TTP, suggesting that it may function as a tumor suppressor in several malignancies. To identify genes that may be associated with TTP tumor suppressor functions in human cancer, we analyzed The Cancer Genome Atlas (TCGA) breast cancer, lung adenocarcinoma, lung squamous cell carcinoma, and colon adenocarcinoma datasets. These analyses defined a signature of 50 genes differentially regulated between high and low TTP-expressing tumors. Notably, patients with low TTP-expressing breast cancer and lung adenocarcinoma had decreased survival rates and more aggressive tumors with increased necrosis. In addition, analysis across non-TCGA tumor gene expression databases identified a broad spectrum of human cancers having similarities with the TTP-low tumor gene signature, including pancreatic, bladder, and prostate cancer. TTP has documented roles in regulating mRNAs encoding inflammatory proteins, and pathway analysis identified several inflammatory pathways that are altered in tumors with low TTP expression. Surprisingly, the TTP-low tumor gene signature includes a core component of 20 under-expressed CREB target genes, suggesting that the regulation of CREB activity may be related to the tumor suppressor function of TTP. Thus, reduced levels of TTP are a potential biomarker for human cancers with poor outcome, and targeting the CREB pathway may be a therapeutic route for treating aggressive TTP-low tumors.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Lung Neoplasms/metabolism , Transcriptome , Tristetraprolin/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Carcinoma/diagnosis , Carcinoma/genetics , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Prognosis , Tristetraprolin/genetics
14.
Cancer Res ; 74(3): 908-20, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24285728

ABSTRACT

Myc oncoproteins induce genes driving aerobic glycolysis, including lactate dehydrogenase-A that generates lactate. Here, we report that Myc controls transcription of the lactate transporter SLC16A1/MCT1 and that elevated MCT1 levels are manifest in premalignant and neoplastic Eµ-Myc transgenic B cells and in human malignancies with MYC or MYCN involvement. Notably, disrupting MCT1 function leads to an accumulation of intracellular lactate that rapidly disables tumor cell growth and glycolysis, provoking marked alterations in glycolytic intermediates, reductions in glucose transport, and in levels of ATP, NADPH, and ultimately, glutathione (GSH). Reductions in GSH then lead to increases in hydrogen peroxide, mitochondrial damage, and ultimately, cell death. Finally, forcing glycolysis by metformin treatment augments this response and the efficacy of MCT1 inhibitors, suggesting an attractive combination therapy for MYC/MCT1-expressing malignancies.


Subject(s)
Glutathione/biosynthesis , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/genetics , Proto-Oncogene Proteins c-myc/metabolism , Symporters/genetics , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , Glycolysis/genetics , Homeostasis/drug effects , Humans , Hydrogen Peroxide/pharmacology , Metformin/pharmacology , Mice , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Symporters/antagonists & inhibitors , Symporters/metabolism , Transcription, Genetic
15.
Cell ; 150(3): 563-74, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863009

ABSTRACT

Myc oncoproteins directly regulate transcription by binding to target genes, yet this only explains a fraction of the genes affected by Myc. mRNA turnover is controlled via AU-binding proteins (AUBPs) that recognize AU-rich elements (AREs) found within many transcripts. Analyses of precancerous and malignant Myc-expressing B cells revealed that Myc regulates hundreds of ARE-containing (ARED) genes and select AUBPs. Notably, Myc directly suppresses transcription of Tristetraprolin (TTP/ZFP36), an mRNA-destabilizing AUBP, and this circuit is also operational during B lymphopoiesis and IL7 signaling. Importantly, TTP suppression is a hallmark of cancers with MYC involvement, and restoring TTP impairs Myc-induced lymphomagenesis and abolishes maintenance of the malignant state. Further, there is a selection for TTP loss in malignancy; thus, TTP functions as a tumor suppressor. Finally, Myc/TTP-directed control of select cancer-associated ARED genes is disabled during lymphomagenesis. Thus, Myc targets AUBPs to regulate ARED genes that control tumorigenesis.


Subject(s)
Genes, Tumor Suppressor , Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tristetraprolin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Stability , RNA, Messenger/chemistry
16.
Mol Carcinog ; 49(2): 152-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19798698

ABSTRACT

Deregulation of E2F transcriptional activity as a result of alterations in the p16-cyclin D-Rb pathway is a hallmark of cancer. However, the roles of the different E2F family members in the process of tumorigenesis are still being elucidated. Studies in mice and humans suggest that E2F2 functions as a tumor suppressor. Here we demonstrate that E2f2 inactivation cooperates with transgenic expression of Myc to enhance tumor development in the skin and oral cavity. In fact, hemizygosity at the E2f2 locus was sufficient to increase tumor incidence in this model. Loss of E2F2 enhanced proliferation in Myc transgenic tissue but did not affect Myc-induced apoptosis. E2F2 did not behave as a simple activator of transcription in epidermal keratinocytes but instead appeared to differentially regulate gene expression dependent on the individual target. E2f2 inactivation also altered the changes in gene expression in Myc transgenic cells by enhancing the increase of some genes, such as cyclin E, and reversing the repression of other genes. These findings demonstrate that E2F2 can function as a tumor suppressor in epithelial tissues, perhaps by limiting proliferation in response to Myc.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic , E2F2 Transcription Factor/physiology , Genes, myc , Animals , Blotting, Western , Immunohistochemistry , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
17.
Cancer Res ; 69(2): 547-53, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147568

ABSTRACT

Neuroblastoma is a pediatric malignancy that arises from the neural crest, and patients with high-risk neuroblastoma, which typically harbor amplifications of MYCN, have an extremely poor prognosis. The tyrosine hydroxylase (TH) promoter-driven TH-MYCN transgenic mouse model faithfully recapitulates many hallmarks of human MYCN-amplified neuroblastoma. A key downstream target of Myc oncoproteins in tumorigenesis is ornithine decarboxylase (Odc), the rate-limiting enzyme of polyamine biosynthesis. Indeed, sustained treatment with the Odc suicide inhibitor alpha-difluoromethylornithine (DFMO) or Odc heterozygosity markedly impairs lymphoma development in Emicro-Myc transgenic mice, and these effects are linked to the induction of the cyclin-dependent kinase (Cdk) inhibitor p27(Kip1), which is normally repressed by Myc. Here, we report that DFMO treatment, but not Odc heterozygosity, impairs MYCN-induced neuroblastoma and that, in this malignancy, transient DFMO treatment is sufficient to confer protection. The selective anticancer effects of DFMO on mouse and human MYCN-amplified neuroblastoma also rely on its ability to disable the proliferative response of Myc, yet in this tumor context, DFMO targets the expression of the p21(Cip1) Cdk inhibitor, which is also suppressed by Myc oncoproteins. These findings suggest that agents, such as DFMO, that target the polyamine pathway may show efficacy in high-risk, MYCN-amplified neuroblastoma.


Subject(s)
Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Ornithine Decarboxylase Inhibitors , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Gene Amplification , Humans , Mice , N-Myc Proto-Oncogene Protein , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Nuclear Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
Proc Natl Acad Sci U S A ; 103(5): 1446-51, 2006 Jan 31.
Article in English | MEDLINE | ID: mdl-16432227

ABSTRACT

Overexpression of the c-myc oncogene contributes to the development of a significant number of human cancers. In response to deregulated Myc activity, the p53 tumor suppressor is activated to promote apoptosis and inhibit tumor formation. Here we demonstrate that p53 induction in response to Myc overexpression requires the ataxia-telangiectasia mutated (ATM) kinase, a major regulator of the cellular response to DNA double-strand breaks. In a transgenic mouse model overexpressing Myc in squamous epithelial tissues, inactivation of Atm suppresses apoptosis and accelerates tumorigenesis. Deregulated Myc expression induces DNA damage in primary transgenic keratinocytes and the formation of gammaH2AX and phospho-SMC1 foci in transgenic tissue. These findings suggest that Myc overexpression causes DNA damage in vivo and that the ATM-dependent response to this damage is critical for p53 activation, apoptosis, and the suppression of tumor development.


Subject(s)
Apoptosis , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Neoplasms/pathology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , Comet Assay , DNA Damage , DNA-Binding Proteins/metabolism , Female , Fibroblasts/metabolism , Genotype , Histones/chemistry , Humans , Immunoblotting , Immunohistochemistry , Keratinocytes/cytology , Keratinocytes/metabolism , Lymphoma/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Neoplasms/metabolism , Oncogenes , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Thymus Gland/pathology , Time Factors , Tumor Suppressor Proteins/metabolism
19.
Mol Cell Biol ; 24(17): 7538-47, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314163

ABSTRACT

The proto-oncogene c-myc encodes a transcription factor that is implicated in the regulation of cellular proliferation, differentiation, and apoptosis and that has also been found to be deregulated in several forms of human and experimental tumors. We have shown that forced expression of c-myc in epithelial tissues of transgenic mice (K5-Myc) resulted in keratinocyte hyperproliferation and the development of spontaneous tumors in the skin and oral cavity. Although a number of genes involved in cancer development are regulated by c-myc, the actual mechanisms leading to Myc-induced neoplasia are not known. Among the genes regulated by Myc is the cyclin-dependent kinase 4 (CDK4) gene. Interestingly, previous studies from our laboratory showed that the overexpression of CDK4 led to keratinocyte hyperproliferation, although no spontaneous tumor development was observed. Thus, we tested the hypothesis that CDK4 may be one of the critical downstream genes involved in Myc carcinogenesis. Our results showed that CDK4 inhibition in K5-Myc transgenic mice resulted in the complete inhibition of tumor development, suggesting that CDK4 is a critical mediator of tumor formation induced by deregulated Myc. Furthermore, a lack of CDK4 expression resulted in marked decreases in epidermal thickness and keratinocyte proliferation compared to the results obtained for K5-Myc littermates. Biochemical analysis of the K5-Myc epidermis showed that CDK4 mediates the proliferative activities of Myc by sequestering p21Cip1 and p27Kip1 and thereby indirectly activating CDK2 kinase activity. These results show that CDK4 mediates the proliferative and oncogenic activities of Myc in vivo through a mechanism that involves the sequestration of specific CDK inhibitors.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Epithelium/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins , Skin Neoplasms/metabolism , Animals , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Division/physiology , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclins/metabolism , Epidermal Cells , Epidermis/metabolism , Epidermis/pathology , Epithelium/pathology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/metabolism
20.
Cancer Res ; 62(11): 3276-81, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12036945

ABSTRACT

Previous studies have demonstrated both oncogenic and tumor suppressive properties for the E2F1 transcription factor. In this study, E2f1-null mice were crossed with transgenic mice expressing Myc under the control of an epithelial-specific keratin 5 promoter to determine whether the absence of E2F1 would modulate the oncogenic activity of Myc. Inactivation of E2f1 was found to significantly accelerate tumor development in keratin 5 Myc transgenic mice. Acceleration of tumorigenesis occurred despite the fact that apoptosis levels were increased in transgenic tissue and tumors null for E2f1, whereas Myc-induced proliferation was unaffected by the status of E2f1. These findings provide new insight into the tumor suppressive activity of E2F1 and identify for the first time a specific oncogenic alteration that cooperates with the loss of E2F1 in tumorigenesis.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Head and Neck Neoplasms/genetics , Proto-Oncogene Proteins c-myc/physiology , Skin Neoplasms/genetics , Transcription Factors/physiology , Animals , Apoptosis/genetics , Cell Division/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Gene Silencing , Genes, Tumor Suppressor , Genes, myc/physiology , Genetic Predisposition to Disease , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...