Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Oxid Med Cell Longev ; 2020: 2148562, 2020.
Article in English | MEDLINE | ID: mdl-32411320

ABSTRACT

The incidence of prostate cancer (PCa) is increasing, and it is currently the second most frequent cause of death by cancer in men. Despite advancements in cancer therapies, new therapeutic approaches are still needed for treatment-refractory advanced metastatic PCa. Cross-species analysis presents a robust strategy for the discovery of new potential therapeutic targets. This strategy involves the integration of genomic data from genetically engineered mouse models (GEMMs) and human PCa datasets. Considering the role of antioxidant pathways in tumor initiation and progression, we searched oxidative stress-related genes for a potential therapeutic target for PCa. First, we analyzed RNA-sequencing data from Pb-Cre4; Ptenf/f mice and discovered an increase in sulfiredoxin (Srxn1) mRNA expression in high-grade prostatic intraepithelial neoplasia (PIN), well-differentiated adenocarcinoma (medium-stage tumors), and poor-differentiated adenocarcinoma (advanced-stage prostate tumors). The increase of SRXN1 protein expression was confirmed by immunohistochemistry in mouse prostate tumor paraffin samples. Analyses of human databases and prostate tissue microarrays demonstrated that SRXN1 is overexpressed in a subset of high-grade prostate tumors and correlates with aggressive PCa with worse prognosis and decreased survival. Analyses in vitro showed that SRXN1 expression is also higher in most PCa cell lines compared to normal cell lines. Furthermore, siRNA-mediated downregulation of SRXN1 led to decreased viability of PCa cells LNCaP. In conclusion, we identified the antioxidant enzyme SRXN1 as a potential therapeutic target for PCa. Our results suggest that the use of specific SRXN1 inhibitors may be an effective strategy for the adjuvant treatment of castration-resistant PCa with SRXN1 overexpression.


Subject(s)
Molecular Targeted Therapy , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Survival , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Oxidative Stress/genetics , Oxidoreductases Acting on Sulfur Group Donors/genetics , Prognosis , Prostate/pathology , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis
2.
Mol Cancer Res ; 17(2): 446-456, 2019 02.
Article in English | MEDLINE | ID: mdl-30333152

ABSTRACT

Among prostate cancers containing Gleason pattern 4, cribriform morphology is associated with unfavorable clinicopathologic factors, but its genetic features and association with long-term outcomes are incompletely understood. In this study, genetic, transcriptional, and epigenetic features of invasive cribriform carcinoma (ICC) tumors were compared with non-cribriform Gleason 4 (NC4) in The Cancer Genome Atlas (TCGA) cohort. ICC (n = 164) had distinctive molecular features when compared with NC4 (n = 102). These include: (i) increased somatic copy number variations (SCNV), specifically deletions at 6q, 8p and 10q, which encompassed PTEN and MAP3K7 losses and gains at 3q; (ii) increased SPOP mut and ATMmut ; (iii) enrichment for mTORC1 and MYC pathways by gene expression; and (iv) increased methylation of selected genes. In addition, when compared with the metastatic prostate cancer, ICC clustered more closely to metastatic prostate cancer than NC4. Validation in clinical cohorts and genomically annotated murine models confirmed the association with SPOPmut (n = 38) and PTENloss (n = 818). The association of ICC with lethal disease was evaluated in the Health Professionals Follow-up Study (HPFS) and Physicians' Health Study (PHS) prospective prostate cancer cohorts (median follow-up, 13.4 years; n = 818). Patients with ICC were more likely to develop lethal cancer [HR, 1.62; 95% confidence interval (CI), 1.05-2.49], independent from Gleason score (GS). IMPLICATIONS: ICC has a distinct molecular phenotype that resembles metastatic prostate cancer and is associated with progression to lethal disease.


Subject(s)
Adenocarcinoma/genetics , Epigenomics/methods , Prostatic Neoplasms/genetics , Adenocarcinoma/pathology , Animals , Humans , Male , Mice , Prostatic Neoplasms/pathology
3.
EMBO Mol Med ; 10(3)2018 03.
Article in English | MEDLINE | ID: mdl-29437778

ABSTRACT

Genetically engineered mouse models of cancer can be used to filter genome-wide expression datasets generated from human tumours and to identify gene expression alterations that are functionally important to cancer development and progression. In this study, we have generated RNAseq data from tumours arising in two established mouse models of prostate cancer, PB-Cre/PtenloxP/loxP and p53loxP/loxPRbloxP/loxP, and integrated this with published human prostate cancer expression data to pinpoint cancer-associated gene expression changes that are conserved between the two species. To identify potential therapeutic targets, we then filtered this information for genes that are either known or predicted to be druggable. Using this approach, we revealed a functional role for the kinase MELK as a driver and potential therapeutic target in prostate cancer. We found that MELK expression was required for cell survival, affected the expression of genes associated with prostate cancer progression and was associated with biochemical recurrence.


Subject(s)
Molecular Targeted Therapy , Prostatic Neoplasms/therapy , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genome , Humans , Male , Mice , Naphthyridines/pharmacology , Neoplasm Invasiveness , Phenotype , Phosphorylation/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Species Specificity , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Stathmin/metabolism , Transcriptome/genetics
4.
Cancer Epidemiol Biomarkers Prev ; 27(2): 201-207, 2018 02.
Article in English | MEDLINE | ID: mdl-29141848

ABSTRACT

Background: The proto-oncogene MYC is implicated in prostate cancer progression. Whether MYC tumor expression at the protein or mRNA level is associated with poorer prognosis has not been well studied.Methods: We conducted a cohort study including 634 men from the Physicians' Health Study and Health Professionals Follow-up Study treated with radical prostatectomy for prostate cancer in 1983-2004 and followed up for a median of 13.7 years. MYC protein expression was evaluated using IHC, and we used Cox regression to calculate HRs and 95% confidence intervals (CIs) of its association with lethal prostate cancer (distant metastases/prostate cancer-related death). We assessed the association between MYC mRNA expression and lethal prostate cancer in a case-control study, including 113 lethal cases and 291 indolent controls.Results: MYC nuclear protein expression was present in 97% of tumors. MYC protein expression was positively correlated with tumor proliferation rate (r = 0.37; P < 0.001) and negatively correlated with apoptotic count (r = -0.17; P < 0.001). There were no significant associations between MYC protein expression and stage, grade, or PSA level at diagnosis. The multivariable HR for lethal prostate cancer among men in the top versus bottom quartile of MYC protein expression was 1.09 (95% CI, 0.50-2.35). There was no significant association between MYC mRNA expression and lethal prostate cancer.Conclusions: Neither MYC protein overexpression nor MYC mRNA overexpression are strong prognostic markers in men treated with radical prostatectomy for prostate cancer.Impact: This is the largest study to examine the prognostic role of MYC protein and mRNA expression in prostate cancer. Cancer Epidemiol Biomarkers Prev; 27(2); 201-7. ©2017 AACR.


Subject(s)
Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/metabolism , Adult , Aged , Biomarkers, Tumor/genetics , Follow-Up Studies , Genes, myc , Humans , Male , Middle Aged , Prospective Studies , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/mortality , Prostatic Neoplasms/surgery , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics
5.
Oncotarget ; 7(20): 28891-902, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27049720

ABSTRACT

Prostate cancer (PCa) is the most commonly diagnosed neoplasm and the second leading cause of cancer-related deaths in men. Acquisition of resistance to conventional therapy is a major problem for PCa patient management. Several mechanisms have been described to promote therapy resistance in PCa, such as androgen receptor (AR) activation, epithelial-to-mesenchymal transition (EMT), acquisition of stem cell properties and neuroendocrine transdifferentiation (NEtD). Recently, we identified Brachyury as a new biomarker of PCa aggressiveness and poor prognosis. In the present study we aimed to assess the role of Brachyury in PCa therapy resistance. We showed that Brachyury overexpression in prostate cancer cells lines increased resistance to docetaxel and cabazitaxel drugs, whereas Brachyury abrogation induced decrease in therapy resistance. Through ChiP-qPCR assays we further demonstrated that Brachyury is a direct regulator of AR expression as well as of the biomarker AMACR and the mesenchymal markers Snail and Fibronectin. Furthermore, in vitro Brachyury was also able to increase EMT and stem properties. By in silico analysis, clinically human Brachyury-positive PCa samples were associated with biomarkers of PCa aggressiveness and therapy resistance, including PTEN loss, and expression of NEtD markers, ERG and Bcl-2. Taken together, our results indicate that Brachyury contributes to tumor chemotherapy resistance, constituting an attractive target for advanced PCa patients.


Subject(s)
Drug Resistance, Neoplasm/physiology , Fetal Proteins/metabolism , Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , T-Box Domain Proteins/metabolism , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/metabolism
6.
J Natl Cancer Inst ; 108(5)2016 May.
Article in English | MEDLINE | ID: mdl-26657335

ABSTRACT

BACKGROUND: The androgen receptor (AR) is a major drug target in prostate cancer (PCa). We profiled the AR-regulated kinome to identify clinically relevant and druggable effectors of AR signaling. METHODS: Using genome-wide approaches, we interrogated all AR regulated kinases. Among these, choline kinase alpha (CHKA) expression was evaluated in benign (n = 195), prostatic intraepithelial neoplasia (PIN) (n = 153) and prostate cancer (PCa) lesions (n = 359). We interrogated how CHKA regulates AR signaling using biochemical assays and investigated androgen regulation of CHKA expression in men with PCa, both untreated (n = 20) and treated with an androgen biosynthesis inhibitor degarelix (n = 27). We studied the effect of CHKA inhibition on the PCa transcriptome using RNA sequencing and tested the effect of CHKA inhibition on cell growth, clonogenic survival and invasion. Tumor xenografts (n = 6 per group) were generated in mice using genetically engineered prostate cancer cells with inducible CHKA knockdown. Data were analyzed with χ(2) tests, Cox regression analysis, and Kaplan-Meier methods. All statistical tests were two-sided. RESULTS: CHKA expression was shown to be androgen regulated in cell lines, xenografts, and human tissue (log fold change from 6.75 to 6.59, P = .002) and was positively associated with tumor stage. CHKA binds directly to the ligand-binding domain (LBD) of AR, enhancing its stability. As such, CHKA is the first kinase identified as an AR chaperone. Inhibition of CHKA repressed the AR transcriptional program including pathways enriched for regulation of protein folding, decreased AR protein levels, and inhibited the growth of PCa cell lines, human PCa explants, and tumor xenografts. CONCLUSIONS: CHKA can act as an AR chaperone, providing, to our knowledge, the first evidence for kinases as molecular chaperones, making CHKA both a marker of tumor progression and a potential therapeutic target for PCa.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Choline Kinase/metabolism , Molecular Chaperones , Molecular Targeted Therapy/methods , Prostatectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Receptors, Androgen/metabolism , Signal Transduction , Aged , Animals , Choline Kinase/antagonists & inhibitors , Choline Kinase/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Grading , Neoplasm Staging , Proportional Hazards Models , Prostatectomy/methods , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Sequence Analysis, DNA , Xenograft Model Antitumor Assays
7.
Oncotarget ; 6(25): 21675-84, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26035357

ABSTRACT

Monocarboxylate Transporter 2 (MCT2) is a major pyruvate transporter encoded by the SLC16A7 gene. Recent studies pointed to a consistent overexpression of MCT2 in prostate cancer (PCa) suggesting MCT2 as a putative biomarker and molecular target. Despite the importance of this observation the mechanisms involved in MCT2 regulation are unknown. Through an integrative analysis we have discovered that selective demethylation of an internal SLC16A7/MCT2 promoter is a recurrent event in independent PCa cohorts. This demethylation is associated with expression of isoforms differing only in 5'-UTR translational control motifs, providing one contributing mechanism for MCT2 protein overexpression in PCa. Genes co-expressed with SLC16A7/MCT2 also clustered in oncogenic-related pathways and effectors of these signalling pathways were found to bind at the SLC16A7/MCT2 gene locus. Finally, MCT2 knock-down attenuated the growth of PCa cells. The present study unveils an unexpected epigenetic regulation of SLC16A7/MCT2 isoforms and identifies a link between SLC16A7/MCT2, Androgen Receptor (AR), ETS-related gene (ERG) and other oncogenic pathways in PCa. These results underscore the importance of combining data from epigenetic, transcriptomic and protein level changes to allow more comprehensive insights into the mechanisms underlying protein expression, that in our case provide additional weight to MCT2 as a candidate biomarker and molecular target in PCa.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Prostatic Neoplasms/metabolism , 5' Untranslated Regions , Amino Acid Motifs , Cohort Studies , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Neoplasm Metastasis , Phenotype , Protein Biosynthesis , RNA, Small Interfering/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcriptional Regulator ERG
8.
J Pathol ; 236(4): 517-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25875424

ABSTRACT

Metabolic adaptation is considered an emerging hallmark of cancer, whereby cancer cells exhibit high rates of glucose consumption with consequent lactate production. To ensure rapid efflux of lactate, most cancer cells express high levels of monocarboxylate transporters (MCTs), which therefore may constitute suitable therapeutic targets. The impact of MCT inhibition, along with the clinical impact of altered cellular metabolism during prostate cancer (PCa) initiation and progression, has not been described. Using a large cohort of human prostate tissues of different grades, in silico data, in vitro and ex vivo studies, we demonstrate the metabolic heterogeneity of PCa and its clinical relevance. We show an increased glycolytic phenotype in advanced stages of PCa and its correlation with poor prognosis. Finally, we present evidence supporting MCTs as suitable targets in PCa, affecting not only cancer cell proliferation and survival but also the expression of a number of hypoxia-inducible factor target genes associated with poor prognosis. Herein, we suggest that patients with highly glycolytic tumours have poorer outcome, supporting the notion of targeting glycolytic tumour cells in prostate cancer through the use of MCT inhibitors.


Subject(s)
Glycolysis , Lactic Acid/metabolism , Molecular Targeted Therapy , Monocarboxylic Acid Transporters/metabolism , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation , Disease Progression , Drug Design , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Transgenic , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Neoplasm Staging , Phenotype , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , Time Factors , Transfection , Tumor Burden
9.
J Cell Mol Med ; 19(4): 723-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25639644

ABSTRACT

Previous studies on monocarboxylate transporters expression in prostate cancer (PCa) have shown that monocarboxylate transporter 2 (MCT2) was clearly overexpressed in prostate malignant glands, pointing it out as a putative biomarker for PCa. However, its localization and possible role in PCa cells remained unclear. In this study, we demonstrate that MCT2 localizes mainly at peroxisomes in PCa cells and is able to take advantage of the peroxisomal transport machinery by interacting with Pex19. We have also shown an increase in MCT2 expression from non-malignant to malignant cells that was directly correlated with its peroxisomal localization. Upon analysis of the expression of several peroxisomal ß-oxidation proteins in PIN lesions and PCa cells from a large variety of human prostate samples, we suggest that MCT2 presence at peroxisomes is related to an increase in ß -oxidation levels which may be crucial for malignant transformation. Our results present novel evidence that may not only contribute to the study of PCa development mechanisms but also pinpoint novel targets for cancer therapy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Monocarboxylic Acid Transporters/metabolism , Peroxisomes/metabolism , Prostatic Neoplasms/metabolism , Basigin/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Male , Membrane Proteins/metabolism , Microscopy, Confocal , Oxidation-Reduction , Prostate/cytology , Prostate/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Isoforms/metabolism , Repressor Proteins/metabolism
10.
Int J Mol Sci ; 15(10): 18333-48, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25314297

ABSTRACT

Metabolic changes during malignant transformation have been noted for many years in tumours. Otto Warburg first reported that cancer cells preferentially rely on glycolysis for energy production, even in the presence of oxygen, leading to the production of high levels of lactate. The crucial role of lactate efflux and exchange within the tumour microenvironment drew attention to monocarboxylate transporters (MCTs). MCTs have been recognized as promising targets in cancer therapy, and their expression was described in a large variety of tumours; however, studies showing how these isoforms contribute to the acquisition of the malignant phenotype are scarce and still unclear regarding prostate cancer. In this review, we focus on the role for MCTs in cell metabolism, supporting the development and progression of prostate cancer, and discuss the exploitation of the metabolic nature of prostate cancer for therapeutic and diagnostic purposes.


Subject(s)
Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Tumor Microenvironment , Animals , Energy Metabolism , Glycolysis , Humans , Male , Molecular Targeted Therapy , Monocarboxylic Acid Transporters/analysis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
11.
Clin Cancer Res ; 20(18): 4949-61, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25009296

ABSTRACT

PURPOSE: Successful therapy of patients with prostate cancer is highly dependent on reliable diagnostic and prognostic biomarkers. Brachyury is considered a negative prognostic factor in colon and lung cancer; however, there are no reports on Brachyury's expression in prostate cancer. EXPERIMENTAL DESIGN: In this study, we aimed to assess the impact of Brachyury expression in prostate tumorigenesis using a large series of human prostate samples comprising benign tissue, prostate intraepithelial neoplasia (PIN) lesions, localized tumor, and metastatic tissues. The results obtained were compared with what can be inferred from the Oncomine database. In addition, multiple in vitro models of prostate cancer were used to dissect the biologic role of Brachyury in prostate cancer progression. RESULTS: We found that Brachyury is significantly overexpressed in prostate cancer and metastatic tumors when compared with normal tissues, both at protein and at mRNA levels. Brachyury expression in the cytoplasm correlates with highly aggressive tumors, whereas the presence of Brachyury in the nucleus is correlated with tumor invasion. We found that Brachyury-positive cells present higher viability, proliferation, migration, and invasion rates than Brachyury-negative cells. Microarray analysis further showed that genes co-expressed with Brachyury are clustered in oncogenic-related pathways, namely cell motility, cell-cycle regulation, and cell metabolism. CONCLUSIONS: Collectively, the present study suggests that Brachyury plays an important role in prostate cancer aggressiveness and points, for the first time, to Brachyury as a significant predictor of poor prostate cancer prognosis. Our work paves the way for future studies assessing Brachyury as a possible prostate cancer therapeutic target.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Fetal Proteins/biosynthesis , Prostatic Neoplasms/pathology , T-Box Domain Proteins/biosynthesis , Aged , Blotting, Western , Cell Line, Tumor , Fetal Proteins/analysis , Humans , Immunohistochemistry , Male , Microscopy, Fluorescence , Middle Aged , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/analysis , Transfection
12.
BMC Cancer ; 14: 352, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24886074

ABSTRACT

BACKGROUND: In a malignant tumour, cancer cells are embedded in stromal cells, namely cancer-associated fibroblasts (CAFs). These CAFs are now accepted as important players in cancer dynamics, being involved in tumour growth and progression. Although there are various reports on the interaction between tumour and stromal cells, the clinico-pathological significance of this cross-talk is still largely unknown. In this study, we aimed to characterise the expression of key metabolic proteins involved in glucose transport, pyruvate/lactate shuttle system, glycolytic metabolism and fatty acid oxidation in CAFs and tumour cells in different stages of malignant transformation. We further aimed to contextualise the clinico-pathological significance of these protein expression profiles with reference to known prognostic indicators, including biochemical recurrence in pT stage. METHODS: Prostate tissues were obtained from 480 patients with a median age of 64 years following radical prostatectomy with no previous hormonal therapy. Tissues were analysed for the expression of several key metabolism-related proteins in glands and surrounding fibroblasts by immunohistochemistry. Reliable markers of prognosis such as pT stage and biochemical recurrence were assessed for each case. RESULTS: We observed that prostate cancer cells did not rely mainly on glycolytic metabolism, while there was a high expression of MCT4 and CAIX - in CAFs. This corroborates the hypothesis of the "Reverse Warburg effect" in prostate cancer, in which fibroblasts are under oxidative stress and express CAIX, an established hypoxia marker. We found that alterations in the expression of metabolism-related proteins were already evident in the early stages of malignant transformation, suggesting the continuing alteration of CAFs from an early stage. Additionally, and for the first time, we show that cases showing high MCT4 expression in CAFs with concomitant strong MCT1 expression in prostate cancer (PCa) cells are associated with poor clinical outcome, namely pT3 stage of the tumour. CONCLUSIONS: In summary, this work demonstrates for the first time the clinico-pathological significance of the lactate shuttle in prostate cancer. It also suggests that other alterations in CAFs may be useful prognostic factors, and further supports the use of MCT1/MCT4 as targets for PCa therapy.


Subject(s)
Energy Metabolism , Fibroblasts/chemistry , Lactic Acid/analysis , Prostatic Neoplasms/chemistry , Stromal Cells/chemistry , Biological Transport , Fibroblasts/pathology , Humans , Kallikreins/blood , Male , Middle Aged , Monocarboxylic Acid Transporters/analysis , Muscle Proteins/analysis , Neoplasm Staging , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Stromal Cells/pathology , Symporters/analysis , Treatment Outcome
13.
EMBO J ; 33(12): 1365-82, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24837709

ABSTRACT

Tumour cells sustain their high proliferation rate through metabolic reprogramming, whereby cellular metabolism shifts from oxidative phosphorylation to aerobic glycolysis, even under normal oxygen levels. Hypoxia-inducible factor 1A (HIF1A) is a major regulator of this process, but its activation under normoxic conditions, termed pseudohypoxia, is not well documented. Here, using an integrative approach combining the first genome-wide mapping of chromatin binding for an endocytic adaptor, ARRB1, both in vitro and in vivo with gene expression profiling, we demonstrate that nuclear ARRB1 contributes to this metabolic shift in prostate cancer cells via regulation of HIF1A transcriptional activity under normoxic conditions through regulation of succinate dehydrogenase A (SDHA) and fumarate hydratase (FH) expression. ARRB1-induced pseudohypoxia may facilitate adaptation of cancer cells to growth in the harsh conditions that are frequently encountered within solid tumours. Our study is the first example of an endocytic adaptor protein regulating metabolic pathways. It implicates ARRB1 as a potential tumour promoter in prostate cancer and highlights the importance of metabolic alterations in prostate cancer.


Subject(s)
Arrestins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Metabolic Networks and Pathways/physiology , Models, Biological , Prostatic Neoplasms/physiopathology , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Fumarate Hydratase/metabolism , Gas Chromatography-Mass Spectrometry , Gene Expression Profiling , Humans , Immunoblotting , Immunohistochemistry , Magnetic Resonance Spectroscopy , Male , Metabolomics , Prostatic Neoplasms/metabolism , RNA Interference , Succinate Dehydrogenase/metabolism , Tissue Array Analysis , beta-Arrestin 1 , beta-Arrestins
14.
Prostate ; 73(7): 763-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23192371

ABSTRACT

BACKGROUND: Monocarboxylate transporter 2 (MCT2) is a transmembrane protein involved in the transport of monocarboxylates such as pyruvate and lactate. In a previous study we described overexpression of MCT2 in prostate carcinoma raising the hypothesis of using MCT2 as a possible biomarker in prostate cancer. With the present study we aimed to compare the pattern of expression of MCT2 and alpha-methylacyl-CoA racemase (AMACR), in prostate carcinoma, PIN lesions, non-neoplastic prostate tissue, and normal prostate and compare their sensitivity and specificity. Also, we wanted to evaluate the value of using MCT2 in combination with AMACR and the negative markers 34ßE12 or p63 to detect prostate cancer. METHODS: A total of 349 cases, including prostate carcinoma, non-neoplastic prostate tissue and PIN lesions, from radical prostatectomies were examined by immunohistochemistry for AMACR, MCT2, p63, and 34ßE12, using tissue microarrays (TMAs). Normal prostate from radical cystoprostatectomy was also studied. RESULTS: Our study revealed that MCT2, similarly to AMACR, was consistently expressed in prostate cancer regardless of the Gleason score. In combination with AMACR and p63 or 34ßE12, MCT2 helped to improve the diagnosis of prostate carcinoma. Also, overexpression of MCT2 as well as AMACR in PIN lesions may indicate the involvement of these two proteins in prostate cancer initiation. CONCLUSIONS: We provided evidence for the presence of MCT2 in prostate cancer, selectively labeling malignant glands. Importantly, assessment of MCT2 together with AMACR, along with the negative markers, highly increases the accuracy in prostate cancer diagnosis.


Subject(s)
Monocarboxylic Acid Transporters/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Racemases and Epimerases/metabolism , Biomarkers , Humans , Immunohistochemistry , Male , Prostate/pathology , Protein Array Analysis , Sensitivity and Specificity
15.
BMC Cancer ; 11: 312, 2011 Jul 25.
Article in English | MEDLINE | ID: mdl-21787388

ABSTRACT

BACKGROUND: Monocarboxylate transporters (MCTs) are transmembrane proteins involved in the transport of monocarboxylates across the plasma membrane, which appear to play an important role in solid tumours, however the role of MCTs in prostate cancer is largely unknown. The aim of the present work was to evaluate the clinico-pathological value of monocarboxylate transporters (MCTs) expression, namely MCT1, MCT2 and MCT4, together with CD147 and gp70 as MCT1/4 and MCT2 chaperones, respectively, in prostate carcinoma. METHODS: Prostate tissues were obtained from 171 patients, who performed radical prostatectomy and 14 patients who performed cystoprostatectomy. Samples and clinico-pathological data were retrieved and organized into tissue microarray (TMAs) blocks. Protein expression was evaluated by immunohistochemistry in neoplastic (n = 171), adjacent non-neoplastic tissues (n = 135), PIN lesions (n = 40) and normal prostatic tissue (n = 14). Protein expression was correlated with patients' clinicopathologic characteristics. RESULTS: In the present study, a significant increase of MCT2 and MCT4 expression in the cytoplasm of tumour cells and a significant decrease in both MCT1 and CD147 expression in prostate tumour cells was observed when compared to normal tissue. All MCT isoforms and CD147 were expressed in PIN lesions. Importantly, for MCT2 and MCT4 the expression levels in PIN lesions were between normal and tumour tissue, which might indicate a role for these MCTs in the malignant transformation. Associations were found between MCT1, MCT4 and CD147 expressions and poor prognosis markers; importantly MCT4 and CD147 overexpression correlated with higher PSA levels, Gleason score and pT stage, as well as with perineural invasion and biochemical recurrence. CONCLUSIONS: Our data provides novel evidence for the involvement of MCTs in prostate cancer. According to our results, we consider that MCT2 should be further explored as tumour marker and both MCT4 and CD147 as markers of poor prognosis in prostate cancer.


Subject(s)
Basigin/biosynthesis , Biomarkers, Tumor/biosynthesis , Monocarboxylic Acid Transporters/biosynthesis , Muscle Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Aged , Humans , Immunohistochemistry , Male , Membrane Glycoproteins/biosynthesis , Middle Aged , Molecular Chaperones , Prognosis , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , Symporters/biosynthesis , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL