Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Mol Sci ; 24(23)2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38069080

ABSTRACT

Metabolic syndrome (MS) is a risk factor for breast cancer (BC) that increases its aggressiveness and metastasis. The prevalence of MS is higher in triple-negative breast cancer (TNBC), which is the molecular subtype with the worst prognosis. The molecular mechanisms underlying this association have not been fully elucidated. MiRNAs are small, non-coding RNAs that regulate gene expression. Aberrant expression of miRNAs in both tissues and fluids are linked to several pathologies. The aim of this work was to identify circulating miRNAs in patients with alterations associated with MS (AAMS) that also impact on BC. Using microarray technology, we detected 23 miRNAs altered in the plasma of women with AAMS that modulate processes linked to cancer. We found that let-7b-5p and miR-28-3p were decreased in plasma from patients with AAMS and also in BC tumors, while miR-877-5p was increased. Interestingly, miR-877-5p expression was associated with lower patient survival, and its expression was higher in PAM50 basal-like BC tumors compared to the other molecular subtypes. Analyses from public databases revealed that miR-877-5p was also increased in plasma from BC patients compared to plasma from healthy donors. We identified IGF2 and TIMP3 as validated target genes of miR-877-5p whose expression was decreased in BC tissue and moreover, was negatively correlated with the levels of this miRNA in the tumors. Finally, a miRNA inhibitor against miR-877-5p diminished viability and tumor growth of the TNBC model 4T1. These results reveal that miR-877-5p inhibition could be a therapeutic option for the treatment of TNBC. Further studies are needed to investigate the role of this miRNA in TNBC progression.


Subject(s)
Circulating MicroRNA , Metabolic Syndrome , MicroRNAs , Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/pathology , Metabolic Syndrome/genetics , MicroRNAs/metabolism , Circulating MicroRNA/therapeutic use , Gene Expression Regulation, Neoplastic
2.
Front Oncol ; 12: 997457, 2022.
Article in English | MEDLINE | ID: mdl-36387263

ABSTRACT

The incidence and mortality of Prostate Cancer (PCa) worldwide correlate with age and bad dietary habits. Previously, we investigated the mRNA/miRNA role on PCa development and progression using high fat diet (HFD) fed mice. Here our main goal was to investigate the effect of HFD on the expression of PCa-related miRNAs and their relevance in PCa patients. We identified 6 up- and 18 down-regulated miRNAs in TRAMP-C1 mice prostate tumors under HFD conditions using miRNA microarrays. Three down-regulated miRNAs: mmu-miR-133a-3p, -1a-3p and -29c-3p were validated in TRAMP-C1 mice prostate tumor by stem-loop RT-qPCR. Hsa-miR-133a-3p/1-3p expression levels were significantly decreased in PCa compared to normal tissues while hsa-miR-133a-3p was found to be further decreased in metastatic prostate cancer tumors compared to non-metastatic PCa. We examined the promoter region of hsa-miR-133a-3p/1-3p genes and compared methylation at these loci with mature miRNA expression. We found that hsa-miR-1-2/miR-133a-1 cluster promoter hypermethylation decreased hsa-miR-133a-3p/1-3p expression in PCa. GOLPH3 and JUP, two hsa-miR-133a-3p and miR-1-3p predicted target genes, were up-regulated in PCa. ROC analysis showed that the combination of hsa-miR-133a-3p, miR-1-3p, GOLPH3 and JUP is a promising panel biomarker to distinguish between PCa and normal adjacent tissue (NAT). These results link PCa aggressiveness to the attenuation of hsa-miR-133a-3p and miR-1-3p expression by promoter hypermethylation. Hsa-miR-133a-3p and miR-1-3p down-regulation may enhance PCa aggressiveness in part by targeting GOLPH3 and JUP.

3.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681793

ABSTRACT

Breast cancer (BCa) is the leading cause of death by cancer in women worldwide. This disease is mainly stratified in four subtypes according to the presence of specific receptors, which is important for BCa aggressiveness, progression and prognosis. MicroRNAs (miRNAs) are small non-coding RNAs that have the capability to modulate several genes. Our aim was to identify a miRNA signature deregulated in preclinical and clinical BCa models for potential biomarker discovery that would be useful for BCa diagnosis and/or prognosis. We identified hsa-miR-21-5p and miR-106b-5p as up-regulated and hsa-miR-205-5p and miR-143-3p as down-regulated in BCa compared to normal breast or normal adjacent (NAT) tissues. We established 51 shared target genes between hsa-miR-21-5p and miR-106b-5p, which negatively correlated with the miRNA expression. Furthermore, we assessed the pathways in which these genes were involved and selected 12 that were associated with cancer and metabolism. Additionally, GAB1, GNG12, HBP1, MEF2A, PAFAH1B1, PPP1R3B, RPS6KA3 and SESN1 were downregulated in BCa compared to NAT. Interestingly, hsa-miR-106b-5p was up-regulated, while GAB1, GNG12, HBP1 and SESN1 were downregulated in aggressive subtypes. Finally, patients with high levels of hsa-miR-106b-5 and low levels of the abovementioned genes had worse relapse free survival and worse overall survival, except for GAB1.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms , MicroRNAs/physiology , Animals , Biomarkers, Tumor/physiology , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Neoplasm Invasiveness , Prognosis , Tumor Cells, Cultured
4.
Am J Cancer Res ; 11(6): 2802-2820, 2021.
Article in English | MEDLINE | ID: mdl-34249429

ABSTRACT

Prostate cancer (PCa) is the most commonly diagnosed male malignancy worldwide. Early diagnosis and metastases detection are crucial features to diminish patient mortality. High fat diet (HFD) and metabolic syndrome increase PCa risk and aggressiveness. Our goal was to identify miRNAs-based biomarkers for PCa diagnosis and prognosis associated with HFD. Mice chronically fed with a HFD or control diet (CD) were subcutaneously inoculated with androgen insensitive PC3 cells. Xenografts from HFD-fed mice showed increased expression of 7 miRNAs that we named "candidates" compared to CD-fed mice. These miRNAs modulate specific metabolic and cancer related pathways. Using bioinformatic tools and human datasets we found that hsa-miR-19b-3p and miR-101-3p showed more than 1,100 validated targets involved in proteoglycans in cancer and fatty acid biosynthesis. These miRNAs were significantly increased in the bloodstream of PCa patients compared to non-PCa volunteers, and in prostate tumors compared to normal adjacent tissues (NAT). Interestingly, both miRNAs were also increased in tumors of metastatic patients compared to tumors of non-metastatic patients. Further receiver-operating characteristic (ROC) analysis determined that hsa-miR-19b-3p and hsa-miR-101-3p in serum showed poor predictive power to discriminate PCa from non-PCa patients. Hsa-miR-19b-3p showed the best score to discriminate between tumor and NAT, while hsa-miR-101-3p was useful to differentiate between metastatic and non-metastatic PCa patients. Hsa-miR-101-3p was increased in exosomes isolated from blood of PCa patients. Although more detailed functional exploration and validation of the molecular mechanisms are required, we identified hsa-miR-19b-3p and hsa-miR-101-3p with high potential for PCa diagnosis and prognosis.

5.
Mol Oncol ; 14(11): 2868-2883, 2020 11.
Article in English | MEDLINE | ID: mdl-32875710

ABSTRACT

Prostate cancer (PCa) remains an important public health concern in Western countries. Metabolic syndrome (MeS) is a cluster of pathophysiological disorders with increasing prevalence in the general population that is a risk factor for PCa. Several studies have determined that a crosstalk between white adipose tissue (WAT) and solid tumors favors cancer aggressiveness. In this work, our main goal was to investigate the interaction between WAT and PCa cells through microRNAs (miRNAs), in MeS mice. We developed a MeS-like disease model using C57BL/6J mice chronically fed with high-fat diet (HFD) that were inoculated with TRAMP-C1 PCa cells. A group of five miRNAs (mmu-miR-221-3p, 27a-3p, 34a-5p, 138-5p, and 146a-5p) were increased in gonadal WAT (gWAT), tumors, and plasma of MeS mice compared to control animals. Three of these five miRNAs were detected in the media from gWAT and TRAMP-C1 cell cocultures, and significantly increased in MeS context. More importantly, hsa-miR-221-3p, 146a-5p, and 27a-3p were increased in bloodstream of PCa patients compared to healthy donors. Using miRNA microarrays, we found that 121 miRNAs were differentially released to the coculture media between HFD-gWAT and tumor cells compared to control diet-gWAT and tumor cells. Target genes for the 66 most deregulated miRNAs were involved in common pathways, mainly related to fatty acid metabolism, ER protein processing, amino acid degradation, PI3K AKT signaling, and PCa. Our findings show for the first time a signature of five miRNAs as important players involved in the interaction between WAT and PCa in MeS mice. Further research will be necessary to track these miRNAs in the interaction between these tissues as well as their role in PCa patients with MeS.


Subject(s)
Gene Expression Regulation, Neoplastic , Metabolic Syndrome/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Transcriptome , Adipose Tissue/metabolism , Animals , Carcinogenesis/genetics , Gene Expression Profiling , Male , Mice, Inbred C57BL
6.
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
7.
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
8.
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
9.
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.

10.
Reproduction ; 154(4): R81-R97, 2017 10.
Article in English | MEDLINE | ID: mdl-28878093

ABSTRACT

MicroRNAs (miRNAs) are non-coding small RNAs that target mRNA to reduce protein expression. They play fundamental roles in several diseases, including prostate cancer (PCa). A single miRNA can target hundreds of mRNAs and coordinately regulate them, which implicates them in nearly every biological pathway. Hence, miRNAs modulate proliferation, cell cycle, apoptosis, adhesion, migration, invasion and metastasis, most of them constituting crucial hallmarks of cancer. Due to these properties, miRNAs emerged as promising tools for diagnostic, prognosis and management of cancer patients. Moreover, they come out as potential targets for cancer treatment, and several efforts are being made to progress in the field of miRNA-based cancer therapy. In this review, we will summarize the recent information about miRNAs in PCa. We will recapitulate all the miRNAs involved in the androgen pathway and the biology of PCa, focusing in PCa initiation and progression. In particular, we will describe the miRNAs associated with cell proliferation, cell cycle and apoptosis in PCa, as well as invasion, adhesion and metastatic miRNAs. We will revise the recent progress made understanding the role of circulating miRNAs identified in PCa that might be useful for PCa patient stratification. Another key aspect to be discussed in this review is miRNAs' role in PCa therapy, including the miRNAs delivery.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Androgens/metabolism , Animals , Biomarkers, Tumor/blood , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Circulating MicroRNA/blood , Circulating MicroRNA/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/blood , MicroRNAs/therapeutic use , Molecular Diagnostic Techniques , Predictive Value of Tests , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction
11.
Oncotarget ; 8(1): 110-117, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27058755

ABSTRACT

The inclusion of genotype at Acute Lymphoblastic Leukemia (ALL) diagnosis as a genetic predictor of disease outcome is under constant study. However, results are inconclusive and seem to be population specific. We analyzed the predictive value of germline polymorphisms for childhood ALL relapse and survival. We retrospectively recruited 140 Argentine patients with de novo ALL. Genotypes were analyzed using PCR-RFLP (GSTP1 c.313A > G, MDR1 c.3435T > C, and MTHFR c.665C > T) and multiplex PCR (GSTT1 null, GSTM1 null). Patients with the GSTP1 c.313GG genotype had an increased risk for relapse in univariate (OR = 2.65, 95% CI = 1.03-6.82, p = 0.04) and multivariate (OR = 3.22, 95% CI = 1.17-8.83, p = 0.02) models. The combined genotype slightly increased risk for relapse in the univariate (OR = 2.82, 95% CI = 1.09-7.32, p = 0.03) and multivariate (OR = 2.98, 95% CI = 1.14-7.79, p = 0.03) models for patients with 2/3-risk-genotypes (GSTT1 null, GSTM1 null, GSTP1 c.313GG). The Recurrence-Free Survival (RFS) was shorter for GSTP1 c.313GG (p = 0.025) and 2/3-risk-genotypes (p = 0.021). GST polymorphisms increased the risk of relapse and RFS of patients with childhood ALL. The inclusion of these genetic markers in ALL treatment protocols might improve risk stratification and reduce the number of relapses and deaths.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Polymorphism, Genetic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Alleles , Female , Genotype , Humans , Male , Odds Ratio , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Proportional Hazards Models , Recurrence , Risk
12.
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
13.
Front Biosci (Elite Ed) ; 8(1): 72-83, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26709647

ABSTRACT

Chemotherapy is still the leader option for cancer treatment. Nevertheless some patients develop chemotherapy resistance. One major research goal is to identify the critical genes involved in chemotherapy response to predict the best therapy option for patients. Germline mutations in the BReast Cancer susceptibility gene (BRCA1) are associated to increased risk of developing breast, ovarian and other types of cancers. However, due to harmful BRCA1 gene mutations are relatively rare in the general population, nowadays most researchers focused on BRCA1 expression downregulation and/or epigenetic inactivation in sporadic tumors as a prognosis tool for chemotherapy response in patients. Chemotherapy response can be dramatically different depending on BRCA1 expression status, tumor type and drug. Hence, the chemotherapy response could be dissimilar in breast, ovarian, uterine, prostate, esophageal, gastric and lung cancers. Additionally, differential BRCA1 expression in sporadic tumors shows different response to DNA-damaging agents, mitotic inhibitors or PARP inhibitors. In this review we will examine the response to different chemotherapy agents in several cancer types depending on BRCA1 expression status.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Genes, BRCA1 , Neoplasms/genetics , Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Humans , Mitosis/drug effects , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prognosis
14.
PLoS One ; 8(11): e80315, 2013.
Article in English | MEDLINE | ID: mdl-24224047

ABSTRACT

Prostate cancer (PCa) is a leading cause of death among males. It is currently estimated that inflammatory responses are linked to 15-20% of all deaths from cancer worldwide. PCa is dominated by complications arising from metastasis to the bone where the tumor cells interact with the bone microenvironment impairing the balance between bone formation and degradation. However, the molecular nature of this interaction is not completely understood. Heme oxygenase-1 (HO-1) counteracts oxidative damage and inflammation. Previous studies from our laboratory showed that HO-1 is implicated in PCa, demonstrating that endogenous HO-1 inhibits bone derived-prostate cancer cells proliferation, invasion and migration and decreases tumor growth and angiogenesis in vivo. The aim of this work was to analyze the impact of HO-1 modulated PCa cells on osteoblasts proliferation in vitro and on bone remodeling in vivo. Using a co-culture system of PC3 cells with primary mice osteoblasts (PMOs), we demonstrated that HO-1 pharmacological induction (hemin treatment) abrogated the diminution of PMOs proliferation induced by PCa cells and decreased the expression of osteoclast-modulating factors in osteoblasts. No changes were detected in the expression of genes involved in osteoblasts differentiation. However, co-culture of hemin pre-treated PC3 cells (PC3 Hem) with PMOs provoked an oxidative status and activated FoxO signaling in osteoblasts. The percentage of active osteoblasts positive for HO-1 increased in calvarias explants co-cultured with PC3 Hem cells. Nuclear HO-1 expression was detected in tumors generated by in vivo bone injection of HO-1 stable transfected PC3 (PC3HO-1) cells in the femur of SCID mice. These results suggest that HO-1 has the potential to modify the bone microenvironment impacting on PCa bone metastasis.


Subject(s)
Heme Oxygenase-1/metabolism , Osteoblasts/metabolism , Prostatic Neoplasms/metabolism , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Heme Oxygenase-1/genetics , Hemin/pharmacology , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Organ Culture Techniques , Osteoblasts/drug effects , Prostatic Neoplasms/genetics
15.
Cell Physiol Biochem ; 30(3): 596-608, 2012.
Article in English | MEDLINE | ID: mdl-22832221

ABSTRACT

Chromosomal instability is a key feature in cancer progression. Recently we have reported that BRCA1 regulates the transcription of several genes in prostate cancer, including ATM (ataxia telangiectasia mutated). Although it is well accepted that ATM is a pivotal mediator in genotoxic stress, it is unknown whether ATM transcription is regulated during the molecular response to DNA damage. Here we investigate ATM transcription regulation in human prostate tumor PC3 cell line. We have found that doxorubicin and mitoxantrone repress ATM transcription in PC3 cells but etoposide and methotrexate do not affect ATM expression. We have demonstrated that BRCA1 binds to ATM promoter and after doxorubicin exposure, it is released. BRCA1 overexpression increases ATM transcription and this enhancement is abolished by BRCA1 depletion. Moreover, BRCA1-BRCT domain loss impairs the ability of BRCA1 to regulate ATM promoter activity, strongly suggesting that BRCT domain is essential for ATM regulation by BRCA1. BRCA1-overexpressing PC3 cells exposed to KU55933 ATM kinase inhibitor showed significant decreased ATM promoter activity compared to untreated cells, suggesting that ATM transcriptional regulation by BRCA1 is partially mediated by the ATM kinase activity. In addition, we have demonstrated E2F1 binding to ATM promoter before and after doxorubicin exposure. E2F1 overexpression diminishes ATM transcription after doxorubicin exposure which is impaired by E2F1 dominant negative mutants. Finally, the co-regulator of transcription CtIP increases ATM transcription. CtIP increases ATM transcription. Altogether, BRCA1/E2F1/CtIP binding to ATM promoter activates ATM transcription. Doxorubicin exposure releases BRCA1 and CtIP from ATM promoter still keeping E2F1 recruited and, in turn, represses ATM expression.


Subject(s)
BRCA1 Protein/metabolism , Carrier Proteins/metabolism , E2F1 Transcription Factor/metabolism , Nuclear Proteins/metabolism , Antibiotics, Antineoplastic/pharmacology , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Damage/drug effects , DNA Repair , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Doxorubicin/pharmacology , Endodeoxyribonucleases , Humans , Morpholines/pharmacology , Promoter Regions, Genetic , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Pyrones/pharmacology , Transcription, Genetic , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
16.
Mol Cancer Res ; 9(8): 1078-90, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21700680

ABSTRACT

BRCA1 plays numerous roles in the regulation of genome integrity and chemoresistance. Although BRCA1 interaction with key proteins involved in DNA repair is well known, its role as a coregulator in the transcriptional response to DNA damage remains poorly understood. In this study, we show that BRCA1 plays a central role in the transcriptional response to genotoxic stress in prostate cancer. BRCA1 expression mediates apoptosis, cell-cycle arrest, and decreased viability in response to doxorubicin treatment. Xenograft studies using human prostate carcinoma PC3 cells show that BRCA1 depletion results in increased tumor growth. A focused survey of BRCA1-regulated genes in prostate carcinoma reveals that multiple regulators of genome stability and cell-cycle control, including BLM, FEN1, DDB2, H3F3B, BRCA2, CCNB2, MAD2L1, and GADD153, are direct transcriptional targets of BRCA1. Furthermore, we show that BRCA1 targets GADD153 promoter to increase its transcription in response to DNA damage. Finally, GADD153 depletion significantly abrogates BRCA1 influence on cell-cycle progression and cell death in response to doxorubicin treatment. These findings define a novel transcriptional pathway through which BRCA1 orchestrates cell fate decisions in response to genotoxic insults, and suggest that BRCA1 status should be considered for new chemotherapeutic treatment strategies in prostate cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , BRCA1 Protein/metabolism , Carcinoma/metabolism , DNA Damage/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Prostatic Neoplasms/metabolism , Transcription Factor CHOP/metabolism , Animals , BRCA1 Protein/genetics , Carcinoma/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Genomic Instability/genetics , Humans , Male , Mice , Prostatic Neoplasms/pathology , Transcription Factor CHOP/genetics
17.
Cell Cycle ; 9(15): 3119-26, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20714219

ABSTRACT

Human PTE Fb is a protein kinase composed by CDK9 and Cyclin T that controls the elongation phase of RNA Pol II. This complex also affects the activation and differentiation program of lymphoid cells. In this study we found that several head and neck tumor cell lines overexpress PTE Fb. We also established that Cyclin T1 is able to induce transformation in vitro, as we determined by foci and colony formation assays. Nu/nu mice s.c. injected with stable transfected Cyclin T1 cells (NIH 3T3 Cyclin T1) developed tumors faster than animals injected with control cells (NIH 3T3 beta-gal). In vitro, NIH 3T3 Cyclin T1 cells show increased proliferation and CDK4-Rb phosphorylation. Even more, silencing E2F1 expression (shRNA E2F1) in NIH 3T3 cells resulted in a dramatic inhibition of Cyclin T1-induced foci. All these data demonstrate for the first time the Cyclin T1 oncogenic function and suggest a role for this protein in controlling cell cycle probably via Rb/E2F1 pathway.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cyclin T/metabolism , Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Clone Cells , Cyclin T/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Mice , NIH 3T3 Cells , Neoplasms/metabolism , Phosphorylation , Positive Transcriptional Elongation Factor B/metabolism , Retinoblastoma Protein/metabolism , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
18.
Mol Cancer Res ; 7(11): 1745-55, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903769

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer-associated death in men. Inflammation has been recognized as a risk factor for this disease. Heme oxygenase 1 (HO-1), the inducible isoform of the rate-limiting enzyme in heme degradation, counteracts oxidative and inflammatory damage. Here, we investigated the regulated expression of HO-1 and its functional consequences in PCa. We studied the effect of genetic and pharmacologic disruption of HO-1 in the growth, invasion, and migration in androgen-sensitive (MDA PCa2b and LNCaP) and androgen-insensitive (PC3) PCa cell lines. Our results show that HO-1 levels are markedly decreased in PC3 compared with MDA PCa2b and LNCaP. Hemin treatment increased HO-1 at both protein and mRNA levels in all cell lines and decreased cell proliferation and invasion. Furthermore, overexpression of HO-1 in PC3 resulted in markedly reduced cell proliferation and migration. Accordingly, small interfering RNA-mediated silencing of HO-1 expression in MDA PCa2b cells resulted in increased proliferation and invasion. Using reverse transcription-quantitative PCR-generated gene array, a set of inflammatory and angiogenic genes were upregulated or downregulated in response to HO-1 overexpression identifying matrix metalloprotease 9 (MMP9) as a novel downstream target of HO-1. MMP9 production and activity was downregulated by HO-1 overexpression. Furthermore, PC3 cells stably transfected with HO-1 (PC3HO-1) and controls were injected into nu/nu mice for analysis of in vivo tumor xenograft phenotype. Tumor growth and MMP9 expression was significantly reduced in PC3HO-1 tumors compared with control xenografts. Taken together, these results implicate HO-1 in PCa cell migration and proliferation suggesting its potential role as a therapeutic target in clinical settings.


Subject(s)
Heme Oxygenase-1/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Down-Regulation , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Hemin/pharmacology , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Microarray Analysis , Neoplasm Invasiveness , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection , Transplantation, Heterologous
19.
Acta bioquím. clín. latinoam ; Acta bioquím. clín. latinoam;36(4): 505-513, dic. 2002. tab
Article in Spanish | LILACS | ID: lil-330744

ABSTRACT

Las porfirias constituyen una familia de enfermedades que son la consecuencia de una deficiencia parcial y específica de una de las enzimas del camino del hemo. Pueden ser adquiridas o genéticas. Las porfirias se pueden clasificar en base a sus manifestaciones clínicas en cutáneas, agudas y mixtas. La porfiria aguda intermitente (PAI) es el tipo más común entre las porfirias agudas, se transmite en forma autosómica dominante y está causada por un defecto en el gen que codifica para la enzima porfobilinógeno deaminasa; su prevalencia en la Argentina es de 1:125.000. Una deficiencia parcial en la protoporfirinógeno oxidasa, otra de las enzimas del camino del hemo, produce la porfiria variegata (PV), la segunda porfiria aguda más frecuente en éste país, heredada también en forma autosómica dominante, con una frecuencia de 1:600.000. En éste trabajo se describen todas las mutaciones detectadas hasta el momento en 43 pacientes con PAI y en 9 pacientes con PV no relacionados


Subject(s)
Humans , Male , Female , Hydroxymethylbilane Synthase , Porphyrias , Acute Disease , Argentina , Mutation , Porphyrias
20.
Acta bioquím. clín. latinoam ; Acta bioquím. clín. latinoam;36(4): 505-513, dic. 2002. tab
Article in Spanish | BINACIS | ID: bin-6866

ABSTRACT

Las porfirias constituyen una familia de enfermedades que son la consecuencia de una deficiencia parcial y específica de una de las enzimas del camino del hemo. Pueden ser adquiridas o genéticas. Las porfirias se pueden clasificar en base a sus manifestaciones clínicas en cutáneas, agudas y mixtas. La porfiria aguda intermitente (PAI) es el tipo más común entre las porfirias agudas, se transmite en forma autosómica dominante y está causada por un defecto en el gen que codifica para la enzima porfobilinógeno deaminasa; su prevalencia en la Argentina es de 1:125.000. Una deficiencia parcial en la protoporfirinógeno oxidasa, otra de las enzimas del camino del hemo, produce la porfiria variegata (PV), la segunda porfiria aguda más frecuente en éste país, heredada también en forma autosómica dominante, con una frecuencia de 1:600.000. En éste trabajo se describen todas las mutaciones detectadas hasta el momento en 43 pacientes con PAI y en 9 pacientes con PV no relacionados (AU)


Subject(s)
Humans , Male , Female , Porphyrias/genetics , Hydroxymethylbilane Synthase/genetics , Argentina , Porphyrias/diagnosis , Porphyrias/epidemiology , Acute Disease , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL