Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 10(4): 299, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30931931

ABSTRACT

About 20% of prostate cancer (PCa) patients progress to metastatic disease. Metabolic syndrome (MeS) is a pathophysiological disorder that increases PCa risk and aggressiveness. C-terminal binding protein (CTBP1) is a transcriptional corepressor that is activated by high-fat diet (HFD). Previously, our group established a MeS/PCa mice model that identified CTBP1 as a novel link associating both diseases. Here, we integrated in vitro (prostate tumor cell lines) and in vivo (MeS/PCa NSG mice) models with molecular and cell biology techniques to investigate MeS/CTBP1 impact over PCa progression, particularly over cell adhesion, mRNA/miRNA expression and PCa spontaneous metastasis development. We found that CTBP1/MeS regulated expression of genes relevant to cell adhesion and PCa progression, such as cadherins, integrins, connexins, and miRNAs in PC3 xenografts. CTBP1 diminished PCa cell adhesion, membrane attachment to substrate and increased filopodia number by modulating gene expression to favor a mesenchymal phenotype. NSG mice fed with HFD and inoculated with CTBP1-depleted PC3 cells, showed a decreased number and size of lung metastases compared to control. Finally, CTBP1 and HFD reduce hsa-mir-30b-5p plasma levels in mice. This study uncovers for the first time the role of CTBP1/MeS in PCa progression and its molecular targets.


Subject(s)
Alcohol Oxidoreductases/metabolism , Cell Adhesion/genetics , DNA-Binding Proteins/metabolism , Metabolic Syndrome/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , Alcohol Oxidoreductases/genetics , Animals , DNA-Binding Proteins/genetics , Diet, High-Fat , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Heterografts/cytology , Heterografts/metabolism , Humans , Male , Metabolic Syndrome/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neoplasm Metastasis , PC-3 Cells , Prostatic Neoplasms/pathology , Pseudopodia/genetics , Pseudopodia/metabolism , RNA, Messenger/metabolism
2.
Int J Cancer ; 144(5): 1115-1127, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30152543

ABSTRACT

Metabolic syndrome (MeS) increases prostate cancer (PCa) risk and aggressiveness. C-terminal binding protein 1 (CTBP1) is a transcriptional co-repressor of tumor suppressor genes that is activated by low NAD+ /NADH ratio. Previously, our group established a MeS and PCa mice model that identified CTBP1 as a novel link associating both diseases. We found that CTBP1 controls the transcription of aromatase (CYP19A1), a key enzyme that converts androgens to estrogens. The aim of this work was to investigate the mechanism that explains CTBP1 as a link between MeS and PCa based on CYP19A1 and estrogen synthesis regulation using PCa cell lines, MeS/PCa mice and adipose co-culture systems. We found that CTBP1 and E1A binding protein p300 (EP300) bind to CYP19A1 promoter and downregulate its expression in PC3 cells. Estradiol, through estrogen receptor beta, released CTBP1 from CYP19A1 promoter triggering its transcription and modulating PCa cell proliferation. We generated NSG and C57BL/6J MeS mice by chronically feeding animals with high fat diet. In the NSG model, CTBP1 depleted PCa xenografts showed an increase in CYP19A1 expression with subsequent increment in intratumor estradiol concentrations. Additionally, in C57BL/6J mice, MeS induced hypertrophy, hyperplasia and inflammation of the white adipose tissue, which leads to a proinflammatory phenotype and increased serum estradiol concentration. Thus, MeS increased PCa growth and Ctbp1, Fabp4 and IL-6 expression levels. These results describe, for the first time, a novel CTBP1/CYP19A1/Estradiol axis that explains, in part, the mechanism for prostate tumor growth increase by MeS.


Subject(s)
Adipose Tissue/pathology , Alcohol Oxidoreductases/genetics , Aromatase/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Estradiol/genetics , Metabolic Syndrome/genetics , Prostatic Neoplasms/genetics , Animals , Cell Line, Tumor , Coculture Techniques/methods , Down-Regulation/genetics , E1A-Associated p300 Protein/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Male , Metabolic Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , PC-3 Cells , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/pathology , Transcription, Genetic/genetics
3.
Int J Cancer ; 143(4): 897-906, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29536528

ABSTRACT

Prostate cancer (PCa) is the most common cancer among men. Metabolic syndrome (MeS) is associated with increased PCa aggressiveness and recurrence. Previously, we proposed C-terminal binding protein 1 (CTBP1), a transcriptional co-repressor, as a molecular link between these two conditions. Notably, CTBP1 depletion decreased PCa growth in MeS mice. The aim of this study was to investigate the molecular mechanisms that explain the link between MeS and PCa mediated by CTBP1. We found that CTBP1 repressed chloride channel accessory 2 (CLCA2) expression in prostate xenografts developed in MeS animals. CTBP1 bound to CLCA2 promoter and repressed its transcription and promoter activity in PCa cell lines. Furthermore, we found that CTBP1 formed a repressor complex with ZEB1, EP300 and HDACs that modulates the CLCA2 promoter activity. CLCA2 promoted PCa cell adhesion inhibiting epithelial-mesenchymal transition (EMT) and activating CTNNB1 together with epithelial marker (CDH1) induction, and mesenchymal markers (SNAI2 and TWIST1) repression. Moreover, CLCA2 depletion in PCa cells injected subcutaneously in MeS mice increased the circulating tumor cells foci compared to control. A microRNA (miRNA) expression microarray from PCa xenografts developed in MeS mice, showed 21 miRNAs modulated by CTBP1 involved in angiogenesis, extracellular matrix organization, focal adhesion and adherents junctions, among others. We found that miR-196b-5p directly targets CLCA2 by cloning CLCA2 3'UTR and performing reporter assays. Altogether, we identified a new molecular mechanism to explain PCa and MeS link based on CLCA2 repression by CTBP1 and miR-196b-5p molecules that might act as key factors in the progression onset of this disease.


Subject(s)
Alcohol Oxidoreductases/physiology , Cell Adhesion/physiology , Chloride Channels/genetics , DNA-Binding Proteins/physiology , E1A-Associated p300 Protein/physiology , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/physiology , Histone Deacetylases/physiology , Metabolic Syndrome/complications , MicroRNAs/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Zinc Finger E-box-Binding Homeobox 1/physiology , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Mice , Promoter Regions, Genetic , Prostatic Neoplasms/complications , Transcription, Genetic
4.
Oncotarget ; 9(17): 13848-13858, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29568399

ABSTRACT

Metastatic breast cancer (BrCa) is still one of the main causes of cancer death in women. Metabolic syndrome (MeS), a risk factor for BrCa, is associated to high grade tumors, increased metastasis and recurrence of this disease. C-terminal binding protein 1 (CTBP1) is a co-repressor of tumor suppressor genes that is activated by low NAD+/NADH ratio. Previously, we demonstrated that CTBP1 hyperactivation by MeS increased tumor growth in MDA-MB-231-derived xenografts regulating several genes and miRNAs. In this work, our aim was to elucidate the role of CTBP1 and MeS in BrCa metastasis. We found that CTBP1 protein diminished adhesion while increased migration of triple negative BrCa cells. CTBP1 and MeS modulated the expression of multiple genes (ITGB4, ITGB6, PRSS2, COL17A1 and FABP4) and miRNAs (miR-378a-3p, miR-146a-5p, let-7e-3p, miR-381-5p, miR-194-5p, miR-494-3p) involved in BrCa progression of MDA-MB-231-derived xenografts. Furthermore, we demonstrated that MeS increased lung micrometastasis and liver neoplastic disease in mice. CTBP1 hyperactivation seems to be critical for MeS effect on BrCa metastasis since CTBP1 depletion completely impaired the detection of circulating tumor cells. Our results highlight CTBP1 and MeS impact on BrCa progression positioning them as key properties to be considered for BrCa patient prognosis and management.

5.
Oncotarget ; 7(14): 18798-811, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26933806

ABSTRACT

Metabolic syndrome (MeS) has been identified as a risk factor for breast cancer. C-terminal binding protein 1 (CtBP1) is a co-repressor of tumor suppressor genes that is activated by low NAD+/NADH ratio. High fat diet (HFD) increases intracellular NADH. We investigated the effect of CtBP1 hyperactivation by HFD intake on mouse breast carcinogenesis. We generated a MeS-like disease in female mice by chronically feeding animals with HFD. MeS increased postnatal mammary gland development and generated prominent duct patterns with markedly increased CtBP1 and Cyclin D1 expression. CtBP1 induced breast cancer cells proliferation. Serum from animals with MeS enriched the stem-like/progenitor cell population from breast cancer cells. CtBP1 increased breast tumor growth in MeS mice modulating multiple genes and miRNA expression implicated in cell proliferation, progenitor cells phenotype, epithelial to mesenchymal transition, mammary development and cell communication in the xenografts. These results define a novel function for CtBP1 in breast carcinogenesis.


Subject(s)
Alcohol Oxidoreductases/metabolism , Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Metabolic Syndrome/metabolism , MicroRNAs/metabolism , Animals , Breast Neoplasms/genetics , Diet, High-Fat , Female , Heterografts , Humans , MCF-7 Cells , Metabolic Syndrome/genetics , Mice , Mice, Nude , NIH 3T3 Cells , Random Allocation , Risk Factors
6.
Mol Cancer Res ; 13(11): 1455-64, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26227317

ABSTRACT

UNLABELLED: Prostate cancer is the second leading cause of cancer-related death in men worldwide. Many factors that participate in the development of prostate cancer promote imbalance in the redox state of the cell. Accumulation of reactive oxygen species causes injury to cell structures, ultimately leading to cancer development. The antioxidant enzyme heme oxygenase 1 (HMOX1/HO-1) is responsible for the maintenance of the cellular homeostasis, playing a critical role in the oxidative stress and the regulation of prostate cancer development and progression. In the present study, the transcriptional regulation of HO-1 was investigated in prostate cancer. Interestingly, the tumor suppressor BRCA1 binds to the HO-1 promoter and modulates HO-1, inducing its protein levels through both the increment of its promoter activity and the induction of its transcriptional activation. In addition, in vitro and in vivo analyses show that BRCA1 also controls HO-1-negative targets: MMP9, uPA, and Cyclin D1. HO-1 transcriptional regulation is also modulated by oxidative and genotoxic agents. Induction of DNA damage by mitoxantrone and etoposide repressed HO-1 transcription, whereas hydrogen peroxide and doxorubicin induced its expression. Xenograft studies showed that HO-1 regulation by doxorubicin also occurs in vivo. Immunofluorescence analysis revealed that BRCA1 overexpression and/or doxorubicin exposure induced the cytoplasmic retention of HO-1. Finally, the transcription factor NRF2 cooperates with BRCA1 protein to activate HO-1 promoter activity. In summary, these results show that the activation of BRCA1-NRF2/HO-1 axis defines a new mechanism for the maintenance of the cellular homeostasis in prostate cancer. IMPLICATIONS: Oxidative and genotoxic stress converge on HO-1 transcriptional activity through the combined actions of BRCA1 and NRF2.


Subject(s)
BRCA1 Protein/metabolism , Heme Oxygenase-1/genetics , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , DNA Damage/genetics , Heme Oxygenase-1/metabolism , Heterografts , Humans , Male , Mice , NF-E2-Related Factor 2/metabolism , Oxidative Stress/genetics , Prostatic Neoplasms/pathology , Protein Binding , Transcriptional Activation
7.
Int J Biochem Cell Biol ; 51: 29-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24685678

ABSTRACT

Epithelial to mesenchymal transition (EMT) of cancer cells is an essential process in cancer progression. Cancer cells that undergone EMT loose cell-cell contacts, acquire mesenchymal properties and develop migratory and invasive abilities. In previous studies we have demonstrated that histamine may modify the invasive phenotype of pancreatic and mammary tumor cells. In this work we proposed to investigate whether histamine may also influence the interaction between tumor cells and normal fibroblasts. The potential activation of normal CCD-1059Sk fibroblasts by histamine and EMT phenotypic changes induced in MCF-7 and MDA-MB-231 breast tumor cells by the conditioned media (CM) derived from fibroblasts were evaluated. Initially, we determined the presence of H1, H2 and H4 histamine receptors and matrix metalloproteinase 2 (MMP2) mRNA in CCD-1059Sk fibroblasts. MMP2 gelatinolytic activity, cell migration and alpha-smooth muscle actin expression were increased in fibroblasts by low doses (<1µM) and decreased by high doses (20µM) of histamine. MCF-7 cells cultured with CM from fibroblasts exhibited spindle-shaped morphology, cell spreading and cytoplasmic expression of ß-catenin but there was no change in MMP2 activity and cell migration. MDA-MB-231 cells cultured with CM from fibroblasts showed a more elongated phenotype, cell spreading, cytoplasmic ß-catenin, increased MMP2 activity and endogenous TGF-ß1 expression, and enhanced cell migration and invasion. Notably, all these features were reversed when mammary tumor cells were cultured with CM from fibroblasts treated with 20µM histamine. In conclusion, high doses of histamine may prevent the activation of fibroblasts and also avert the EMT related changes induced in epithelial tumor cells by fibroblasts CM.


Subject(s)
Breast Neoplasms/pathology , Fibroblasts/metabolism , Histamine/pharmacology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Female , Fibroblasts/cytology , Humans , MCF-7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...