Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
1.
Int J Mol Sci ; 24(23)2023 Nov 25.
Article de Anglais | MEDLINE | ID: mdl-38069080

RÉSUMÉ

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.


Sujet(s)
MicroARN circulant , Syndrome métabolique X , microARN , Tumeurs du sein triple-négatives , Humains , Femelle , Tumeurs du sein triple-négatives/anatomopathologie , Syndrome métabolique X/génétique , microARN/métabolisme , MicroARN circulant/usage thérapeutique , Régulation de l'expression des gènes tumoraux
2.
Cell Death Dis ; 10(4): 299, 2019 04 01.
Article de Anglais | MEDLINE | ID: mdl-30931931

RÉSUMÉ

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.


Sujet(s)
Alcohol oxidoreductases/métabolisme , Adhérence cellulaire/génétique , Protéines de liaison à l'ADN/métabolisme , Syndrome métabolique X/génétique , microARN/génétique , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , ARN messager/génétique , Alcohol oxidoreductases/génétique , Animaux , Protéines de liaison à l'ADN/génétique , Alimentation riche en graisse , Modèles animaux de maladie humaine , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Hétérogreffes/cytologie , Hétérogreffes/métabolisme , Humains , Mâle , Syndrome métabolique X/métabolisme , Souris , Souris de lignée NOD , Souris SCID , microARN/métabolisme , Métastase tumorale , Cellules PC-3 , Tumeurs de la prostate/anatomopathologie , Pseudopodes/génétique , Pseudopodes/métabolisme , ARN messager/métabolisme
3.
Int J Cancer ; 144(5): 1115-1127, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30152543

RÉSUMÉ

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.


Sujet(s)
Tissu adipeux/anatomopathologie , Alcohol oxidoreductases/génétique , Aromatase/génétique , Prolifération cellulaire/génétique , Protéines de liaison à l'ADN/génétique , Oestradiol/génétique , Syndrome métabolique X/génétique , Tumeurs de la prostate/génétique , Animaux , Lignée cellulaire tumorale , Techniques de coculture/méthodes , Régulation négative/génétique , Protéine p300-E1A/génétique , Régulation de l'expression des gènes tumoraux/génétique , Humains , Inflammation/génétique , Inflammation/anatomopathologie , Mâle , Syndrome métabolique X/anatomopathologie , Souris , Souris de lignée C57BL , Souris de lignée NOD , Cellules PC-3 , Régions promotrices (génétique)/génétique , Tumeurs de la prostate/anatomopathologie , Transcription génétique/génétique
4.
Oncotarget ; 9(17): 13848-13858, 2018 Mar 02.
Article de Anglais | MEDLINE | ID: mdl-29568399

RÉSUMÉ

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.
Int J Cancer ; 143(4): 897-906, 2018 08 15.
Article de Anglais | MEDLINE | ID: mdl-29536528

RÉSUMÉ

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.


Sujet(s)
Alcohol oxidoreductases/physiologie , Adhérence cellulaire/physiologie , Canaux chlorure/génétique , Protéines de liaison à l'ADN/physiologie , Protéine p300-E1A/physiologie , Épigenèse génétique , Transition épithélio-mésenchymateuse/physiologie , Histone deacetylases/physiologie , Syndrome métabolique X/complications , microARN/physiologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Facteur de transcription Zeb1/physiologie , Animaux , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Hétérogreffes , Humains , Mâle , Souris , Régions promotrices (génétique) , Tumeurs de la prostate/complications , Transcription génétique
6.
Reproduction ; 154(4): R81-R97, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28878093

RÉSUMÉ

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.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Transformation cellulaire néoplasique/génétique , microARN/génétique , Tumeurs de la prostate/génétique , Androgènes/métabolisme , Animaux , Marqueurs biologiques tumoraux/sang , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , MicroARN circulant/sang , MicroARN circulant/génétique , Régulation de l'expression des gènes tumoraux , Humains , Mâle , microARN/sang , microARN/usage thérapeutique , Techniques de diagnostic moléculaire , Valeur prédictive des tests , Tumeurs de la prostate/sang , Tumeurs de la prostate/anatomopathologie , Récepteurs aux androgènes/génétique , Récepteurs aux androgènes/métabolisme , Transduction du signal
7.
Oncotarget ; 7(14): 18798-811, 2016 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-26933806

RÉSUMÉ

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.


Sujet(s)
Alcohol oxidoreductases/métabolisme , Tumeurs du sein/métabolisme , Protéines de liaison à l'ADN/métabolisme , Syndrome métabolique X/métabolisme , microARN/métabolisme , Animaux , Tumeurs du sein/génétique , Alimentation riche en graisse , Femelle , Hétérogreffes , Humains , Cellules MCF-7 , Syndrome métabolique X/génétique , Souris , Souris nude , Cellules NIH 3T3 , Répartition aléatoire , Facteurs de risque
8.
Front Biosci (Elite Ed) ; 8(1): 72-83, 2016 01 01.
Article de Anglais | MEDLINE | ID: mdl-26709647

RÉSUMÉ

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.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux/métabolisme , Gène BRCA1 , Tumeurs/génétique , Antinéoplasiques/pharmacologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Humains , Mitose/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Pronostic
9.
Cell Physiol Biochem ; 30(3): 596-608, 2012.
Article de Anglais | MEDLINE | ID: mdl-22832221

RÉSUMÉ

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.


Sujet(s)
Protéine BRCA1/métabolisme , Protéines de transport/métabolisme , Facteur de transcription E2F1/métabolisme , Protéines nucléaires/métabolisme , Antibiotiques antinéoplasiques/pharmacologie , Protéines mutées dans l'ataxie-télangiectasie , Protéine BRCA1/composition chimique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Altération de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Doxorubicine/pharmacologie , Endodeoxyribonucleases , Humains , Morpholines/pharmacologie , Régions promotrices (génétique) , Liaison aux protéines/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Structure tertiaire des protéines , Pyrones/pharmacologie , Transcription génétique , Protéines suppresseurs de tumeurs/antagonistes et inhibiteurs , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/métabolisme
10.
Mol Cancer Res ; 9(8): 1078-90, 2011 Aug.
Article de Anglais | MEDLINE | ID: mdl-21700680

RÉSUMÉ

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.


Sujet(s)
Antibiotiques antinéoplasiques/pharmacologie , Protéine BRCA1/métabolisme , Carcinomes/métabolisme , Altération de l'ADN/génétique , Doxorubicine/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Tumeurs de la prostate/métabolisme , Facteur de transcription CHOP/métabolisme , Animaux , Protéine BRCA1/génétique , Carcinomes/anatomopathologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/génétique , Lignée cellulaire tumorale , Instabilité du génome/génétique , Humains , Mâle , Souris , Tumeurs de la prostate/anatomopathologie , Facteur de transcription CHOP/génétique
11.
Cell Cycle ; 9(15): 3119-26, 2010 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-20714219

RÉSUMÉ

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.


Sujet(s)
Transformation cellulaire néoplasique/anatomopathologie , Cycline T/métabolisme , Tumeurs/anatomopathologie , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Transformation cellulaire néoplasique/génétique , Clones cellulaires , Cycline T/génétique , Kinase-4 cycline-dépendante/métabolisme , Kinase-9 cycline-dépendante/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou/enzymologie , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Humains , Souris , Cellules NIH 3T3 , Tumeurs/métabolisme , Phosphorylation , Facteur B d'élongation transcriptionnelle positive/métabolisme , Protéine du rétinoblastome/métabolisme , Test clonogénique de cellules souches tumorales , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Mol Cancer Res ; 7(11): 1745-55, 2009 Nov.
Article de Anglais | MEDLINE | ID: mdl-19903769

RÉSUMÉ

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.


Sujet(s)
Heme oxygenase-1/métabolisme , Tumeurs de la prostate/enzymologie , Tumeurs de la prostate/anatomopathologie , Animaux , Processus de croissance cellulaire/effets des médicaments et des substances chimiques , Processus de croissance cellulaire/physiologie , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/physiologie , Régulation négative , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes codant pour des enzymes , Heme oxygenase-1/génétique , Hémine/pharmacologie , Humains , Immunohistochimie , Mâle , Matrix metalloproteinase 9/génétique , Matrix metalloproteinase 9/métabolisme , Souris , Analyse sur microréseau , Invasion tumorale , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Petit ARN interférent/administration et posologie , Petit ARN interférent/génétique , Transfection , Transplantation hétérologue
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE