Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
1.
Nat Commun ; 15(1): 5585, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992040

RÉSUMÉ

MYCN oncogene amplification is frequently observed in aggressive childhood neuroblastoma. Using an unbiased large-scale mutagenesis screen in neuroblastoma-prone transgenic mice, we identify a single germline point mutation in the transcriptional corepressor Runx1t1, which abolishes MYCN-driven tumorigenesis. This loss-of-function mutation disrupts a highly conserved zinc finger domain within Runx1t1. Deletion of one Runx1t1 allele in an independent Runx1t1 knockout mouse model is also sufficient to prevent MYCN-driven neuroblastoma development, and reverse ganglia hyperplasia, a known pre-requisite for tumorigenesis. Silencing RUNX1T1 in human neuroblastoma cells decreases colony formation in vitro, and inhibits tumor growth in vivo. Moreover, RUNX1T1 knockdown inhibits the viability of PAX3-FOXO1 fusion-driven rhabdomyosarcoma and MYC-driven small cell lung cancer cells. Despite the role of Runx1t1 in MYCN-driven tumorigenesis neither gene directly regulates the other. We show RUNX1T1 forms part of a transcriptional LSD1-CoREST3-HDAC repressive complex recruited by HAND2 to enhancer regions to regulate chromatin accessibility and cell-fate pathway genes.


Sujet(s)
Carcinogenèse , Protéine du proto-oncogène N-Myc , Neuroblastome , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Animaux , Protéine du proto-oncogène N-Myc/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Humains , Souris , Carcinogenèse/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Souris transgéniques , Souris knockout , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Protéines corépressives/métabolisme , Protéines corépressives/génétique
2.
Sci Adv ; 10(13): eadk5386, 2024 Mar 29.
Article de Anglais | MEDLINE | ID: mdl-38536927

RÉSUMÉ

While pancreatic ductal adenocarcinomas (PDACs) are addicted to KRAS-activating mutations, inhibitors of downstream KRAS effectors, such as the MEK1/2 kinase inhibitor trametinib, are devoid of therapeutic effects. However, the extensive rewiring of regulatory circuits driven by the attenuation of the KRAS pathway may induce vulnerabilities of therapeutic relevance. An in-depth molecular analysis of the transcriptional and epigenomic alterations occurring in PDAC cells in the initial hours after MEK1/2 inhibition by trametinib unveiled the induction of endogenous retroviruses (ERVs) escaping epigenetic silencing, leading to the production of double-stranded RNAs and the increased expression of interferon (IFN) genes. We tracked ERV activation to the early induction of the transcription factor ELF3, which extensively bound and activated nonsilenced retroelements and synergized with IRF1 (interferon regulatory factor 1) in the activation of IFNs and IFN-stimulated genes. Trametinib-induced viral mimicry in PDAC may be exploited in the rational design of combination therapies in immuno-oncology.


Sujet(s)
Carcinome du canal pancréatique , Rétrovirus endogènes , Tumeurs du pancréas , Humains , Rétrovirus endogènes/génétique , Transduction du signal , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme
3.
Cancers (Basel) ; 13(8)2021 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-33918978

RÉSUMÉ

Ornithine decarboxylase (ODC1), a critical regulatory enzyme in polyamine biosynthesis, is a direct transcriptional target of MYCN, amplification of which is a powerful marker of aggressive neuroblastoma. A single nucleotide polymorphism (SNP), G316A, within the first intron of ODC1, results in genotypes wildtype GG, and variants AG/AA. CRISPR-cas9 technology was used to investigate the effects of AG clones from wildtype MYCN-amplified SK-N-BE(2)-C cells and the effect of the SNP on MYCN binding, and promoter activity was investigated using EMSA and luciferase assays. AG clones exhibited decreased ODC1 expression, growth rates, and histone acetylation and increased sensitivity to ODC1 inhibition. MYCN was a stronger transcriptional regulator of the ODC1 promoter containing the G allele, and preferentially bound the G allele over the A. Two neuroblastoma cohorts were used to investigate the clinical impact of the SNP. In the study cohort, the minor AA genotype was associated with improved survival, while poor prognosis was associated with the GG genotype and AG/GG genotypes in MYCN-amplified and non-amplified patients, respectively. These effects were lost in the GWAS cohort. We have demonstrated that the ODC1 G316A polymorphism has functional significance in neuroblastoma and is subject to allele-specific regulation by the MYCN oncoprotein.

4.
Cancer Res ; 80(19): 4129-4144, 2020 10 01.
Article de Anglais | MEDLINE | ID: mdl-32816860

RÉSUMÉ

Therapeutic checkpoint antibodies blocking programmed death receptor 1/programmed death ligand 1 (PD-L1) signaling have radically improved clinical outcomes in cancer. However, the regulation of PD-L1 expression on tumor cells is still poorly understood. Here we show that intratumoral copper levels influence PD-L1 expression in cancer cells. Deep analysis of the The Cancer Genome Atlas database and tissue microarrays showed strong correlation between the major copper influx transporter copper transporter 1 (CTR-1) and PD-L1 expression across many cancers but not in corresponding normal tissues. Copper supplementation enhanced PD-L1 expression at mRNA and protein levels in cancer cells and RNA sequencing revealed that copper regulates key signaling pathways mediating PD-L1-driven cancer immune evasion. Conversely, copper chelators inhibited phosphorylation of STAT3 and EGFR and promoted ubiquitin-mediated degradation of PD-L1. Copper-chelating drugs also significantly increased the number of tumor-infiltrating CD8+ T and natural killer cells, slowed tumor growth, and improved mouse survival. Overall, this study reveals an important role for copper in regulating PD-L1 and suggests that anticancer immunotherapy might be enhanced by pharmacologically reducing intratumor copper levels. SIGNIFICANCE: These findings characterize the role of copper in modulating PD-L1 expression and contributing to cancer immune evasion, highlighting the potential for repurposing copper chelators as enhancers of antitumor immunity. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4129/F1.large.jpg.


Sujet(s)
Antigène CD274/métabolisme , Tumeurs du cerveau/immunologie , Cuivre/métabolisme , Neuroblastome/immunologie , Échappement de la tumeur à la surveillance immunitaire/physiologie , Animaux , Antigène CD274/génétique , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Lignée cellulaire tumorale , Chélateurs/pharmacologie , Transporteur-1 du cuivre/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/immunologie , Humains , Immunothérapie/méthodes , Cellules tueuses naturelles , Lymphocytes TIL/anatomopathologie , Souris de lignée BALB C , Neuroblastome/traitement médicamenteux , Neuroblastome/métabolisme , 1,1',1''-Phosphoryltriaziridine/pharmacologie , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Theranostics ; 10(14): 6411-6429, 2020.
Article de Anglais | MEDLINE | ID: mdl-32483461

RÉSUMÉ

Targeting glutamine metabolism has emerged as a potential therapeutic strategy for Myc overexpressing cancer cells. Myc proteins contribute to an aggressive neuroblastoma phenotype. Radiotherapy is one of the treatment modalities for high-risk neuroblastoma patients. Herein, we investigated the effect of glutamine deprivation in combination with irradiation in neuroblastoma cells representative of high-risk disease and studied the role of Myc member interplay in regulating neuroblastoma cell radioresistance. Methods: Cell proliferation and viability assays were used to establish the effect of glutamine deprivation in neuroblastoma cells expressing c-Myc or MycN. Gene silencing and overexpression were used to modulate the expression of Myc genes to determine their role in neuroblastoma radioresistance. qPCR and western blot investigated interplay between expression of Myc members. The impact of glutamine deprivation on cell response following irradiation was explored using a radiobiological 3D colony assay. DNA repair gene pathways as well as CSC-related genes were studied by qPCR array. Reactive Oxygen Species (ROS) and glutathione (GSH) levels were detected by fluorescence and luminescence probes respectively. Cancer-stem cell (CSC) properties were investigated by sphere-forming assay and flow cytometry to quantify CSC markers. Expression of DNA repair genes and CSC-related genes was analysed by mining publicly available patient datasets. Results: Our results showed that glutamine deprivation decreased neuroblastoma cell proliferation and viability and modulated Myc member expression. We then demonstrated for the first time that combined glutamine deprivation with irradiation led to a selective radioresistance of MYCN-amplified neuroblastoma cells. By exploring the underlying mechanism of neuroblastoma radioresistance properties, our results highlight interplay between c-Myc and MycN expression suggesting compensatory mechanisms in Myc proteins leading to radioresistance in MYCN-amplified cells. This result was associated with the ability of MYCN-amplified cells to dysregulate the DNA repair gene pathway, maintain GSH and ROS levels and to increase the CSC-like population and properties. Conversely, glutamine deprivation led to radiosensitization in non-MYCN amplified cell lines through a disruption of the cell redox balance and a trend to decrease in the CSC-like populations. Mining publicly available gene expression dataset obtained from pediatric neuroblastoma patients, we identified a correlation pattern between Myc members and CSC-related genes as well as a specific group of DNA repair gene pathways. Conclusions: This study demonstrated that MycN and c-Myc tightly cooperate in regulation of the neuroblastoma CSC phenotypes and radioresistance upon glutamine deprivation. Pharmacologically, strategies targeting glutamine metabolism may prove beneficial in Myc-driven tumors. Consideration of MycN/c-Myc status in selecting neuroblastoma patients for glutamine metabolism treatment will be important to avoid potential radioresistance.


Sujet(s)
Glutamine/métabolisme , Protéine du proto-oncogène N-Myc/métabolisme , Cellules souches tumorales/métabolisme , Lignée cellulaire tumorale , Réparation de l'ADN , Régulation de l'expression des gènes tumoraux , Gènes myc , Humains , Protéine du proto-oncogène N-Myc/génétique , Neuroblastome/thérapie , Radiothérapie/méthodes
6.
Molecules ; 25(9)2020 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-32365886

RÉSUMÉ

Carbon nanostructures (CN) are emerging valuable materials for the assembly of highly engineered multifunctional nanovehicles for cancer therapy, in particular for counteracting the insurgence of multi-drug resistance (MDR). In this regard, carbon nanotubes (CNT), graphene oxide (GO), and fullerenes (F) have been proposed as promising materials due to their superior physical, chemical, and biological features. The possibility to easily modify their surface, conferring tailored properties, allows different CN derivatives to be synthesized. Although many studies have explored this topic, a comprehensive review evaluating the beneficial use of functionalized CNT vs G or F is still missing. Within this paper, the most relevant examples of CN-based nanosystems proposed for MDR reversal are reviewed, taking into consideration the functionalization routes, as well as the biological mechanisms involved and the possible toxicity concerns. The main aim is to understand which functional CN represents the most promising strategy to be further investigated for overcoming MDR in cancer.


Sujet(s)
Antinéoplasiques/composition chimique , Carbone/composition chimique , Résistance aux médicaments antinéoplasiques , Nanostructures/composition chimique , Glycoprotéine P/génétique , Glycoprotéine P/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Vecteurs de médicaments/composition chimique , Systèmes de délivrance de médicaments , Multirésistance aux médicaments , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie
7.
Int J Cancer ; 147(8): 2225-2238, 2020 10 15.
Article de Anglais | MEDLINE | ID: mdl-32277480

RÉSUMÉ

Epithelial ovarian cancer (EOC) is a complex disease comprising discrete histological and molecular subtypes, for which survival rates remain unacceptably low. Tailored approaches for this deadly heterogeneous disease are urgently needed. Efflux pumps belonging to the ATP-binding cassette (ABC) family of transporters are known for roles in both drug resistance and cancer biology and are also highly targetable. Here we have investigated the association of ABCC4/MRP4 expression to clinical outcome and its biological function in endometrioid and serous tumors, common histological subtypes of EOC. We found high expression of ABCC4/MRP4, previously shown to be directly regulated by c-Myc/N-Myc, was associated with poor prognosis in endometrioid EOC (P = .001) as well as in a subset of serous EOC with a "high-MYCN" profile (C5/proliferative; P = .019). Transient siRNA-mediated suppression of MRP4 in EOC cells led to reduced growth, migration and invasion, with the effects being most pronounced in endometrioid and C5-like serous cells compared to non-C5 serous EOC cells. Sustained knockdown of MRP4 also sensitized endometrioid cells to MRP4 substrate drugs. Furthermore, suppression of MRP4 decreased the growth of patient-derived EOC cells in vivo. Together, our findings provide the first evidence that MRP4 plays an important role in the biology of Myc-associated ovarian tumors and highlight this transporter as a potential therapeutic target for EOC.


Sujet(s)
Carcinome épithélial de l'ovaire/génétique , Carcinome épithélial de l'ovaire/anatomopathologie , Gènes myc/génétique , Protéines associées à la multirésistance aux médicaments/génétique , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Carcinome endométrioïde/génétique , Carcinome endométrioïde/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/anatomopathologie , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Humains , Pronostic , Petit ARN interférent/génétique , Taux de survie
8.
ACS Pharmacol Transl Sci ; 3(1): 148-160, 2020 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-32259094

RÉSUMÉ

MYCN-amplified neuroblastoma is one of the deadliest forms of childhood cancer and remains a significant clinical challenge. Direct pharmacological inhibition of MYCN is not currently achievable. One strategy could be to target the AKT/GSK3ß pathway, which directly regulates the stability of the MYCN protein. Numerous potent and isoform-specific small-molecule AKT inhibitors have been developed. However, the selection of the right drug combinations in the relevant indication will have a significant impact on AKT inhibitor clinical success. To maximally exploit the potential of AKT inhibitors, a better understanding of AKT isoform functions in cancer is crucial. Here using RNAi to downregulate specific AKT isoforms, we demonstrated that loss of total AKT activity rather than isoform-specific expression was necessary to decrease MYCN expression and cause a significant decrease in neuroblastoma cell proliferation. Consistent with these observations, isoform-specific pharmacological inhibition of AKT was substantially less effective than pan-AKT inhibition in combination with cytotoxic drugs in MYCN-amplified neuroblastoma. The allosteric pan-AKT inhibitor perifosine had promising in vitro and in vivo activity in combination with conventional cytotoxic drugs in MYCN-amplified neuroblastoma cells. Our results demonstrated that perifosine drug combination was able to induce apoptosis and downregulate ABC transporter expression. Collectively, this study shows that selecting pan-AKT inhibitors rather than isoform-specific drugs to synergize with first-line chemotherapy treatment should be considered for clinical trials for aggressive neuroblastoma and, potentially, other MYCN -driven cancers.

10.
Oncogene ; 39(17): 3555-3570, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32123312

RÉSUMÉ

Amplification of the MYCN oncogene occurs in ~25% of primary neuroblastomas and is the single most powerful biological marker of poor prognosis in this disease. MYCN transcriptionally regulates a range of biological processes important for cancer, including cell metabolism. The MYCN-regulated metabolic gene SLC16A1, encoding the lactate transporter monocarboxylate transporter 1 (MCT1), is a potential therapeutic target. Treatment of neuroblastoma cells with the MCT1 inhibitor SR13800 increased intracellular lactate levels, disrupted the nicotinamide adenine dinucleotide (NADH/NAD+) ratio, and decreased intracellular glutathione levels. Metabolite tracing with 13C-glucose and 13C-glutamine following MCT1 inhibitor treatment revealed increased quantities of tricarboxylic acid (TCA) cycle intermediates and increased oxygen consumption rate. MCT1 inhibition was highly synergistic with vincristine and LDHA inhibition under cell culture conditions, but this combination was ineffective against neuroblastoma xenografts. Posttreatment xenograft tumors had increased synthesis of the MCT1 homolog MCT4/SLC16A, a known resistance factor to MCT1 inhibition. We found that MCT4 was negatively regulated by MYCN in luciferase reporter assays and its synthesis in neuroblastoma cells was increased under hypoxic conditions and following hypoxia-inducible factor (HIF1) induction, suggesting that MCT4 may contribute to resistance to MCT1 inhibitor treatment in hypoxic neuroblastoma tumors. Co-treatment of neuroblastoma cells with inhibitors of MCT1 and LDHA, the enzyme responsible for lactate production, resulted in a large increase in intracellular pyruvate and was highly synergistic in decreasing neuroblastoma cell viability. These results highlight the potential of targeting MCT1 in neuroblastoma in conjunction with strategies that involve disruption of pyruvate homeostasis and indicate possible resistance mechanisms.


Sujet(s)
Antinéoplasiques/pharmacologie , Systèmes de délivrance de médicaments , Transporteurs d'acides monocarboxyliques , Protéines tumorales , Neuroblastome , Symporteurs , Vincristine/pharmacocinétique , Animaux , Lignée cellulaire tumorale , Cycle citrique/effets des médicaments et des substances chimiques , Femelle , Humains , Souris de lignée BALB C , Souris nude , Transporteurs d'acides monocarboxyliques/antagonistes et inhibiteurs , Transporteurs d'acides monocarboxyliques/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Neuroblastome/traitement médicamenteux , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Symporteurs/antagonistes et inhibiteurs , Symporteurs/génétique , Symporteurs/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Materials (Basel) ; 12(18)2019 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-31500165

RÉSUMÉ

A hybrid system composed of multi-walled carbon nanotubes coated with chitosan was proposed as a pH-responsive carrier for the vectorization of methotrexate to lung cancer. The effective coating of the carbon nanostructure by chitosan, quantified (20% by weight) by thermogravimetric analysis, was assessed by combined scanning and transmission electron microscopy, and X-ray photoelectron spectroscopy (N1s signal), respectively. Furthermore, Raman spectroscopy was used to characterize the interaction between polysaccharide and carbon counterparts. Methotrexate was physically loaded onto the nanohybrid and the release profiles showed a pH-responsive behavior with higher and faster release in acidic (pH 5.0) vs. neutral (pH 7.4) environments. Empty nanoparticles were found to be highly biocompatible in either healthy (MRC-5) or cancerous (H1299) cells, with the nanocarrier being effective in reducing the drug toxicity on MRC-5 while enhancing the anticancer activity on H1299.

12.
Pharmaceuticals (Basel) ; 12(2)2019 May 18.
Article de Anglais | MEDLINE | ID: mdl-31109098

RÉSUMÉ

Selective vectorization of Cisplatin (CisPt) to Glioblastoma U87 cells was exploited by the fabrication of a hybrid nanocarrier composed of magnetic γ-Fe2O3 nanoparticles and nanographene oxide (NGO). The magnetic component, obtained by annealing magnetite Fe3O4 and characterized by XRD measurements, was combined with NGO sheets prepared via a modified Hummer's method. The morphological and thermogravimetric analysis proved the effective binding of γ-Fe2O3 nanoparticles onto NGO layers. The magnetization measured under magnetic fields up to 7 Tesla at room temperature revealed superparamagnetic-like behavior with a maximum value of MS = 15 emu/g and coercivity HC ≈ 0 Oe within experimental error. The nanohybrid was found to possess high affinity towards CisPt, and a rather slow fractional release profile of 80% after 250 h. Negligible toxicity was observed for empty nanoparticles, while the retainment of CisPt anticancer activity upon loading into the carrier was observed, together with the possibility to spatially control the drug delivery at a target site.

13.
Sci Transl Med ; 11(477)2019 01 30.
Article de Anglais | MEDLINE | ID: mdl-30700572

RÉSUMÉ

Amplification of the MYCN oncogene is associated with an aggressive phenotype and poor outcome in childhood neuroblastoma. Polyamines are highly regulated essential cations that are frequently elevated in cancer cells, and the rate-limiting enzyme in polyamine synthesis, ornithine decarboxylase 1 (ODC1), is a direct transcriptional target of MYCN. Treatment of neuroblastoma cells with the ODC1 inhibitor difluoromethylornithine (DFMO), although a promising therapeutic strategy, is only partially effective at impeding neuroblastoma cell growth due to activation of compensatory mechanisms resulting in increased polyamine uptake from the surrounding microenvironment. In this study, we identified solute carrier family 3 member 2 (SLC3A2) as the key transporter involved in polyamine uptake in neuroblastoma. Knockdown of SLC3A2 in neuroblastoma cells reduced the uptake of the radiolabeled polyamine spermidine, and DFMO treatment increased SLC3A2 protein. In addition, MYCN directly increased polyamine synthesis and promoted neuroblastoma cell proliferation by regulating SLC3A2 and other regulatory components of the polyamine pathway. Inhibiting polyamine uptake with the small-molecule drug AMXT 1501, in combination with DFMO, prevented or delayed tumor development in neuroblastoma-prone mice and extended survival in rodent models of established tumors. Our findings suggest that combining AMXT 1501 and DFMO with standard chemotherapy might be an effective strategy for treating neuroblastoma.


Sujet(s)
Évolution de la maladie , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Polyamines/métabolisme , Animaux , Voies de biosynthèse/génétique , Lignée cellulaire tumorale , Études de cohortes , Modèles animaux de maladie humaine , Amplification de gène , Régulation de l'expression des gènes , Régulation de l'expression des gènes tumoraux , Protéines de transport membranaire/métabolisme , Souris , Analyse multifactorielle , Protéine du proto-oncogène N-Myc/génétique , Neuroblastome/génétique , Pronostic , Modèles des risques proportionnels , Analyse de survie , Résultat thérapeutique
14.
Eur J Cancer ; 83: 132-141, 2017 09.
Article de Anglais | MEDLINE | ID: mdl-28735070

RÉSUMÉ

The ATP-binding cassette transporter ABCC4 (multidrug resistance protein 4, MRP4) mRNA level is a strong predictor of poor clinical outcome in neuroblastoma which may relate to its export of endogenous signalling molecules and chemotherapeutic agents. We sought to determine whether ABCC4 contributes to development, growth and drug response in neuroblastoma in vivo. In neuroblastoma patients, high ABCC4 protein levels were associated with reduced overall survival. Inducible knockdown of ABCC4 strongly inhibited the growth of human neuroblastoma cells in vitro and impaired the growth of neuroblastoma xenografts. Loss of Abcc4 in the Th-MYCN transgenic neuroblastoma mouse model did not impact tumour formation; however, Abcc4-null neuroblastomas were strongly sensitised to the ABCC4 substrate drug irinotecan. Our findings demonstrate a role for ABCC4 in neuroblastoma cell proliferation and chemoresistance and provide rationale for a strategy where inhibition of ABCC4 should both attenuate the growth of neuroblastoma and sensitise tumours to ABCC4 chemotherapeutic substrates.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Camptothécine/analogues et dérivés , Protéines associées à la multirésistance aux médicaments/déficit , Neuroblastome/traitement médicamenteux , Animaux , Technique de Western , Camptothécine/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Doxycycline/pharmacologie , Hétérogreffes/effets des médicaments et des substances chimiques , Irinotécan , Souris , Souris knockout , Protéines associées à la multirésistance aux médicaments/physiologie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
15.
Oncotarget ; 7(34): 54937-54951, 2016 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-27448979

RÉSUMÉ

Amplification of the MYCN oncogene, a member of the MYC family of transcriptional regulators, is one of the most powerful prognostic markers identified for poor outcome in neuroblastoma, the most common extracranial solid cancer in childhood. While MYCN has been established as a key driver of malignancy in neuroblastoma, the underlying molecular mechanisms are poorly understood. Transcription factor activating enhancer binding protein-4 (TFAP4) has been reported to be a direct transcriptional target of MYC. We show for the first time that high expression of TFAP4 in primary neuroblastoma patients is associated with poor clinical outcome. siRNA-mediated suppression of TFAP4 in MYCN-expressing neuroblastoma cells led to inhibition of cell proliferation and migration. Chromatin immunoprecipitation assay demonstrated that TFAP4 expression is positively regulated by MYCN. Microarray analysis identified genes regulated by both MYCN and TFAP4 in neuroblastoma cells, including Phosphoribosyl-pyrophosphate synthetase-2 (PRPS2) and Syndecan-1 (SDC1), which are involved in cancer cell proliferation and metastasis. Overall this study suggests a regulatory circuit in which MYCN by elevating TFAP4 expression, cooperates with it to control a specific set of genes involved in tumor progression. These findings highlight the existence of a MYCN-TFAP4 axis in MYCN-driven neuroblastoma as well as identifying potential therapeutic targets for aggressive forms of this disease.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes tumoraux , Protéine du proto-oncogène N-Myc/génétique , Neuroblastome/génétique , Facteurs de transcription/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Protéines de liaison à l'ADN/métabolisme , Évolution de la maladie , Analyse de profil d'expression de gènes/méthodes , Humains , Estimation de Kaplan-Meier , Protéine du proto-oncogène N-Myc/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Interférence par ARN , Facteurs de transcription/métabolisme
16.
Hum Mol Genet ; 25(18): 3887-3907, 2016 09 15.
Article de Anglais | MEDLINE | ID: mdl-27466189

RÉSUMÉ

Cyclin-dependent kinase-like 5 (CDKL5) is a Ser/Thr protein kinase predominantly expressed in the brain. Mutations of the CDKL5 gene lead to CDKL5 disorder, a neurodevelopmental pathology that shares several features with Rett Syndrome and is characterized by severe intellectual disability. The phosphorylation targets of CDKL5 are largely unknown, which hampers the discovery of therapeutic strategies for improving the neurological phenotype due to CDKL5 mutations. Here, we show that the histone deacetylase 4 (HDAC4) is a direct phosphorylation target of CDKL5 and that CDKL5-dependent phosphorylation promotes HDAC4 cytoplasmic retention. Nuclear HDAC4 binds to chromatin as well as to MEF2A transcription factor, leading to histone deacetylation and altered neuronal gene expression. By using a Cdkl5 knockout (Cdkl5 -/Y) mouse model, we found that hypophosphorylated HDAC4 translocates to the nucleus of neural precursor cells, thereby reducing histone 3 acetylation. This effect was reverted by re-expression of CDKL5 or by inhibition of HDAC4 activity through the HDAC4 inhibitor LMK235. In Cdkl5 -/Y mice treated with LMK235, defective survival and maturation of neuronal precursor cells and hippocampus-dependent memory were fully normalized. These results demonstrate a critical role of HDAC4 in the neurodevelopmental alterations due to CDKL5 mutations and suggest the possibility of HDAC4-targeted pharmacological interventions.


Sujet(s)
Histone deacetylases/biosynthèse , Déficience intellectuelle/génétique , Protein-Serine-Threonine Kinases/génétique , Syndrome de Rett/génétique , Spasmes infantiles/génétique , Animaux , Modèles animaux de maladie humaine , Antienzymes/administration et posologie , Syndromes épileptiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/croissance et développement , Hippocampe/anatomopathologie , Histone deacetylases/effets des médicaments et des substances chimiques , Histone deacetylases/génétique , Humains , Déficience intellectuelle/traitement médicamenteux , Déficience intellectuelle/physiopathologie , Facteurs de transcription MEF2/génétique , Souris , Souris knockout , Mutation , Cellules souches neurales/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Phosphorylation , Syndrome de Rett/traitement médicamenteux , Syndrome de Rett/anatomopathologie , Spasmes infantiles/traitement médicamenteux , Spasmes infantiles/anatomopathologie
17.
J Biol Chem ; 288(29): 20817-20829, 2013 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-23740250

RÉSUMÉ

Intellectual disability in Down syndrome (DS) appears to be related to severe proliferation impairment during brain development. Recent evidence shows that it is not only cellular proliferation that is heavily compromised in DS, but also cell fate specification and dendritic maturation. The amyloid precursor protein (APP), a gene that is triplicated in DS, plays a key role in normal brain development by influencing neural precursor cell proliferation, cell fate specification, and neuronal maturation. APP influences these processes via two separate domains, the APP intracellular domain (AICD) and the soluble secreted APP. We recently found that the proliferation impairment of neuronal precursors (NPCs) from the Ts65Dn mouse model for DS was caused by derangement of the Shh pathway due to overexpression of patched1(Ptch1), its inhibitory regulator. Ptch1 overexpression was related to increased levels within the APP/AICD system. The overall goal of this study was to determine whether APP contributes to neurogenesis impairment in DS by influencing in addition to proliferation, cell fate specification, and neurite development. We found that normalization of APP expression restored the reduced neuronogenesis, the increased astrogliogenesis, and the reduced neurite length of trisomic NPCs, indicating that APP overexpression underpins all aspects of neurogenesis impairment. Moreover, we found that two different domains of APP impair neuronal differentiation and maturation in trisomic NPCs. The APP/AICD system regulates neuronogenesis and neurite length through the Shh pathway, whereas the APP/secreted AP system promotes astrogliogenesis through an IL-6-associated signaling cascade. These results provide novel insight into the mechanisms underlying brain development alterations in DS.


Sujet(s)
Précurseur de la protéine bêta-amyloïde/composition chimique , Précurseur de la protéine bêta-amyloïde/métabolisme , Différenciation cellulaire , Syndrome de Down/génétique , Cellules souches neurales/anatomopathologie , Neurites/métabolisme , Trisomie/génétique , Animaux , Astrocytes/métabolisme , Lignage cellulaire , Forme de la cellule , Modèles animaux de maladie humaine , Femelle , Extinction de l'expression des gènes , Protéines Hedgehog/métabolisme , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Humains , Interleukine-6/métabolisme , Mâle , Souris , Souris transgéniques , Modèles biologiques , Cellules souches neurales/métabolisme , Neurogenèse , Récepteurs patched , Récepteur Patched-1 , Structure tertiaire des protéines , Récepteurs de surface cellulaire/métabolisme , Transduction du signal , Relation structure-activité
18.
Front Oncol ; 3: 42, 2013.
Article de Anglais | MEDLINE | ID: mdl-23482921

RÉSUMÉ

Neuroblastoma is the most common extra cranial solid tumor in childhood and the most frequently diagnosed neoplasm during the infancy. MYCN amplification and overexpression occur in about 25% of total neuroblastoma cases and this percentage increases at 30% in advanced stage neuroblastoma. So far, MYCN expression profile is still one of the most robust and significant prognostic markers for neuroblastoma outcome. MYCN is a transcription factor that belongs to the family of MYC oncoproteins, comprising c-MYC and MYCL genes. Deregulation of MYC oncoprotein expression is a crucial event involved in the occurrence of different types of malignant tumors. MYCN, as well as c-MYC, can heterodimerize with its partner MAX and activate the transcription of several target genes containing E-Box sites in their promoter regions. However, recent several lines of evidence have revealed that MYCN can repress at least as many genes as it activates, thus proposing a novel function of this protein in neuroblastoma biology. Whereas the mechanism by which MYCN can act as a transcriptional activator is relatively well known, very few studies has been done in the attempt to explain how MYCN can exert its transcription repression function. Here, we will review current knowledge about the mechanism of MYCN-mediated transcriptional repression and will emphasize its role as a repressor in the recruitment of a precise set of proteins to form complexes capable of down-regulating specific subsets of genes whose function is actively involved in apoptosis, cell differentiation, chemosensitivity, and cell motility. The finding that MYCN can also act as a repressor has widen our view on its role in oncogenesis and has posed the bases to search for novel therapeutic drugs that can specifically target its transcriptional repression function.

19.
J Biol Chem ; 288(12): 8332-8341, 2013 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-23362253

RÉSUMÉ

CLU (clusterin) is a tumor suppressor gene that we have previously shown to be negatively modulated by the MYCN proto-oncogene, but the mechanism of repression was unclear. Here, we show that MYCN inhibits the expression of CLU by direct interaction with the non-canonical E box sequence CACGCG in the 5'-flanking region. Binding of MYCN to the CLU gene induces bivalent epigenetic marks and recruitment of repressive proteins such as histone deacetylases and Polycomb members. MYCN physically binds in vitro and in vivo to EZH2, a component of the Polycomb repressive complex 2, required to repress CLU. Notably, EZH2 interacts with the Myc box domain 3, a segment of MYC known to be essential for its transforming effects. The expression of CLU can be restored in MYCN-amplified cells by epigenetic drugs with therapeutic results. Importantly, the anticancer effects of the drugs are ablated if CLU expression is blunted by RNA interference. Our study implies that MYC tumorigenesis can be effectively antagonized by epigenetic drugs that interfere with the recruitment of chromatin modifiers at repressive E boxes of tumor suppressor genes such as CLU.


Sujet(s)
Neuroblastome/traitement médicamenteux , Protéines nucléaires/métabolisme , Protéines oncogènes/métabolisme , Complexe répresseur Polycomb-2/métabolisme , Région 5' flanquante , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Séquence nucléotidique , Lignée cellulaire tumorale/effets des médicaments et des substances chimiques , Mouvement cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Chromatine/métabolisme , Clusterine/génétique , Clusterine/métabolisme , Éléments E-box , Protéine-2 homologue de l'activateur de Zeste , Épigenèse génétique , Régulation de l'expression des gènes tumoraux , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Acides hydroxamiques/pharmacologie , Données de séquences moléculaires , Protéine du proto-oncogène N-Myc , Protéines nucléaires/physiologie , Protéines oncogènes/physiologie , Régions promotrices (génétique) , Liaison aux protéines , Proto-oncogène Mas
20.
PLoS One ; 7(9): e45328, 2012.
Article de Anglais | MEDLINE | ID: mdl-23028937

RÉSUMÉ

The 2.2 Mb long dystrophin (DMD) gene, the largest gene in the human genome, corresponds to roughly 0.1% of the entire human DNA sequence. Mutations in this gene cause Duchenne muscular dystrophy and other milder X-linked, recessive dystrophinopathies. Using a custom-made tiling array, specifically designed for the DMD locus, we identified a variety of novel long non-coding RNAs (lncRNAs), both sense and antisense oriented, whose expression profiles mirror that of DMD gene. Importantly, these transcripts are intronic in origin and specifically localized to the nucleus and are transcribed contextually with dystrophin isoforms or primed by MyoD-induced myogenic differentiation. Furthermore, their forced ectopic expression in both human muscle and neuronal cells causes a specific and negative regulation of endogenous dystrophin full length isoforms and significantly down-regulate the activity of a luciferase reporter construct carrying the minimal promoter regions of the muscle dystrophin isoform. Consistent with this apparently repressive role, we found that, in muscle samples of dystrophinopathic female carriers, lncRNAs expression levels inversely correlate with those of muscle full length DMD isoforms. Overall these findings unveil an unprecedented complexity of the transcriptional pattern of the DMD locus and reveal that DMD lncRNAs may contribute to the orchestration and homeostasis of the muscle dystrophin expression pattern by either selective targeting and down-modulating the dystrophin promoter transcriptional activity.


Sujet(s)
Dystrophine/génétique , Muscles squelettiques/métabolisme , Isoformes d'ARN/génétique , ARN long non codant/génétique , Femelle , Humains , Mâle , Transcription génétique/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...