Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 5578, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552068

ABSTRACT

Retinoblastoma is the most frequent intraocular malignancy in children, originating from a maturing cone precursor in the developing retina. Little is known on the molecular basis underlying the biological and clinical behavior of this cancer. Here, using multi-omics data, we demonstrate the existence of two retinoblastoma subtypes. Subtype 1, of earlier onset, includes most of the heritable forms. It harbors few genetic alterations other than the initiating RB1 inactivation and corresponds to differentiated tumors expressing mature cone markers. By contrast, subtype 2 tumors harbor frequent recurrent genetic alterations including MYCN-amplification. They express markers of less differentiated cone together with neuronal/ganglion cell markers with marked inter- and intra-tumor heterogeneity. The cone dedifferentiation in subtype 2 is associated with stemness features including low immune and interferon response, E2F and MYC/MYCN activation and a higher propensity for metastasis. The recognition of these two subtypes, one maintaining a cone-differentiated state, and the other, more aggressive, associated with cone dedifferentiation and expression of neuronal markers, opens up important biological and clinical perspectives for retinoblastomas.


Subject(s)
Retinal Cone Photoreceptor Cells/pathology , Retinal Ganglion Cells/metabolism , Retinal Neoplasms/classification , Retinoblastoma/classification , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Dedifferentiation/genetics , Child, Preschool , DNA Methylation , Female , Gene Expression , Genetic Heterogeneity , Humans , Infant , Male , Mutation , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Metastasis , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/pathology , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma/pathology
2.
Clin Cancer Res ; 26(21): 5735-5746, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32900798

ABSTRACT

PURPOSE: Medulloblastoma is an important cause of mortality and morbidity in pediatric oncology. Here, we investigated whether the DNA repair inhibitor, AsiDNA, could help address a significant unmet clinical need in medulloblastoma care, by improving radiotherapy efficacy without increasing radiation-associated toxicity. EXPERIMENTAL DESIGN: To evaluate the brain permeability of AsiDNA upon systemic delivery, we intraperitoneally injected a fluorescence form of AsiDNA in models harboring brain tumors and in models still in development. Studies evaluated toxicity associated with combination of AsiDNA with radiation in the treatment of young developing animals at subacute levels, related to growth and development, and at chronic levels, related to brain organization and cognitive skills. Efficacy of the combination of AsiDNA with radiation was tested in two different preclinical xenografted models of high-risk medulloblastoma and in a panel of medulloblastoma cell lines from different molecular subgroups and TP53 status. Role of TP53 on the AsiDNA-mediated radiosensitization was analyzed by RNA-sequencing, DNA repair recruitment, and cell death assays. RESULTS: Capable of penetrating young brain tissues, AsiDNA showed no added toxicity to radiation. Combination of AsiDNA with radiotherapy improved the survival of animal models more efficiently than increasing radiation doses. Medulloblastoma radiosensitization by AsiDNA was not restricted to a specific molecular group or status of TP53. Molecular mechanisms of AsiDNA, previously observed in adult malignancies, were conserved in pediatric models and resembled dose increase when combined with irradiation. CONCLUSIONS: Our results suggest that AsiDNA is an attractive candidate to improve radiotherapy in medulloblastoma, with no indication of additional toxicity in developing brain tissues.


Subject(s)
DNA/pharmacology , Medulloblastoma/drug therapy , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53/genetics , Adult , Animals , Cell Line, Tumor , Child , DNA/adverse effects , DNA Repair/genetics , DNA Repair/radiation effects , Heterografts , Humans , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Medulloblastoma/radiotherapy , Pediatrics , RNA-Seq , Radiation-Sensitizing Agents/adverse effects
3.
EMBO Mol Med ; 11(8): e9830, 2019 08.
Article in English | MEDLINE | ID: mdl-31328883

ABSTRACT

Medulloblastoma (MB) is a pediatric tumor of the cerebellum divided into four groups. Group 3 is of bad prognosis and remains poorly characterized. While the current treatment involving surgery, radiotherapy, and chemotherapy often fails, no alternative therapy is yet available. Few recurrent genomic alterations that can be therapeutically targeted have been identified. Amplifications of receptors of the TGFß/Activin pathway occur at very low frequency in Group 3 MB. However, neither their functional relevance nor activation of the downstream signaling pathway has been studied. We showed that this pathway is activated in Group 3 MB with some samples showing a very strong activation. Beside genetic alterations, we demonstrated that an ActivinB autocrine stimulation is responsible for pathway activation in a subset of Group 3 MB characterized by high PMEPA1 levels. Importantly, Galunisertib, a kinase inhibitor of the cognate receptors currently tested in clinical trials for Glioblastoma patients, showed efficacy on orthotopically grafted MB-PDX. Our data demonstrate that the TGFß/Activin pathway is active in a subset of Group 3 MB and can be therapeutically targeted.


Subject(s)
Autocrine Communication , Cerebellar Neoplasms/metabolism , Inhibin-beta Subunits/metabolism , Medulloblastoma/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibin-beta Subunits/genetics , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Nude , Phosphorylation , Pyrazoles/pharmacology , Quinolines/pharmacology , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta3/genetics , Tumor Burden , Xenograft Model Antitumor Assays
4.
Cancer Cell ; 33(3): 435-449.e6, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29533784

ABSTRACT

Cancer cells often express differentiation programs unrelated to their tissue of origin, although the contribution of these aberrant phenotypes to malignancy is poorly understood. An aggressive subgroup of medulloblastoma, a malignant pediatric brain tumor of the cerebellum, expresses a photoreceptor differentiation program normally expressed in the retina. We establish that two photoreceptor-specific transcription factors, NRL and CRX, are master regulators of this program and are required for tumor maintenance in this subgroup. Beyond photoreceptor lineage genes, we identify BCL-XL as a key transcriptional target of NRL and provide evidence substantiating anti-BCL therapy as a rational treatment opportunity for select MB patients. Our results highlight the utility of studying aberrant differentiation programs in cancer and their potential as selective therapeutic vulnerabilities.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Medulloblastoma/genetics , Trans-Activators/genetics , Animals , Cell Differentiation/genetics , Cerebellar Neoplasms/genetics , Humans , Mice, Nude , Retina/pathology , Transcription, Genetic/genetics
5.
Nat Commun ; 8: 15262, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28497782

ABSTRACT

NRAS and its effector BRAF are frequently mutated in melanoma. Paradoxically, CRAF but not BRAF was shown to be critical for various RAS-driven cancers, raising the question of the role of RAF proteins in NRAS-induced melanoma. Here, using conditional ablation of Raf genes in NRAS-induced mouse melanoma models, we investigate their contribution in tumour progression, from the onset of benign tumours to malignant tumour maintenance. We show that BRAF expression is required for ERK activation and nevi development, demonstrating a critical role in the early stages of NRAS-driven melanoma. After melanoma formation, single Braf or Craf ablation is not sufficient to block tumour growth, showing redundant functions for RAF kinases. Finally, proliferation of resistant cells emerging in the absence of BRAF and CRAF remains dependent on ARAF-mediated ERK activation. These results reveal specific and compensatory functions for BRAF and CRAF and highlight an addiction to RAF signalling in NRAS-driven melanoma.


Subject(s)
Melanoma/metabolism , Monomeric GTP-Binding Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Disease Progression , Humans , MAP Kinase Signaling System/genetics , Melanoma/genetics , Melanoma/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monomeric GTP-Binding Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/genetics , ras Proteins/genetics
6.
Cell Rep ; 2(4): 774-80, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23022482

ABSTRACT

B-Raf and C-Raf kinases have emerged as critical players in melanoma. However, little is known about their role during development and homeostasis of the melanocyte lineage. Here, we report that knockout of B-raf and C-raf genes in this lineage results in normal pigmentation at birth with no defect in migration, proliferation, or differentiation of melanoblasts in mouse hair follicles. In contrast, the double raf knockout mice displayed hair graying resulting from a defect in cell-cycle entry of melanocyte stem cells (MSCs) and their subsequent depletion in the hair follicle bulge. Therefore, Raf signaling is dispensable for early melanocyte lineage development, but necessary for MSC maintenance.


Subject(s)
Melanocytes/cytology , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Extracellular Signal-Regulated MAP Kinases/metabolism , Hair Follicle/physiology , Mice , Mice, Knockout , Proto-Oncogene Proteins B-raf/deficiency , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-raf/deficiency , Proto-Oncogene Proteins c-raf/genetics , Signal Transduction , Stem Cell Factor/metabolism , Xenopus/growth & development
7.
Clin Cancer Res ; 18(1): 263-72, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22096025

ABSTRACT

PURPOSE: The emergence of skin tumors in patients treated with sorafenib or with more recent BRAF inhibitors is an intriguing and potentially serious event. We carried out a clinical, pathologic, and molecular study of skin lesions occurring in patients receiving sorafenib. EXPERIMENTAL DESIGN: Thirty-one skin lesions from patients receiving sorafenib were characterized clinically and pathologically. DNA extracted from the lesions was screened for mutation hot spots of HRAS, NRAS, KiRAS, TP53, EGFR, BRAF, AKT1, PI3KCA, TGFBR1, and PTEN. Biological effect of sorafenib was studied in vivo in normal skin specimen and in vitro on cultured keratinocytes. RESULTS: We observed a continuous spectrum of lesions: from benign to more inflammatory and proliferative lesions, all seemingly initiated in the hair follicles. Eight oncogenic HRAS, TGFBR1, and TP53 mutations were found in 2 benign lesions, 3 keratoacanthomas (KA) and 3 KA-like squamous cell carcinoma (SCC). Six of them correspond to the typical UV signature. Treatment with sorafenib led to an increased keratinocyte proliferation and a tendency toward increased mitogen-activated protein kinase (MAPK) pathway activation in normal skin. Sorafenib induced BRAF-CRAF dimerization in cultured keratinocytes and activated CRAF with a dose-dependent effect on MAP-kinase pathway activation and on keratinocyte proliferation. CONCLUSION: Sorafenib induces keratinocyte proliferation in vivo and a time- and dose-dependent activation of the MAP kinase pathway in vitro. It is associated with a spectrum of lesions ranging from benign follicular cystic lesions to KA-like SCC. Additional and potentially preexisting somatic genetic events, like UV-induced mutations, might influence the evolution of benign lesions to more proliferative and malignant tumors.


Subject(s)
Benzenesulfonates/adverse effects , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/adverse effects , Receptors, Transforming Growth Factor beta/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Cells, Cultured , Female , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/radiation effects , Male , Middle Aged , Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction , Skin/drug effects , Skin/radiation effects , Skin Neoplasms/diagnosis , Sorafenib , Ultraviolet Rays/adverse effects , raf Kinases/genetics , ras Proteins/genetics
8.
Dev Neurobiol ; 70(7): 485-97, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20213756

ABSTRACT

Dorsal root ganglia proceed from the coalescence of cell bodies of sensory neurons, which have migrated dorsoventrally from the delaminating neural crest. They are composed of different neuronal subtypes with specific sensory functions, including nociception, thermal sensation, proprioception, and mechanosensation. In contrast to proprioceptors and thermonociceptors, little is known about the molecular mechanisms governing the early commitment and later differentiation into mechanosensitive neurons. This is mainly due to the absence of specific molecular markers for this particular cell type. Using knockout mice, we identified the bZIP transcription factor MafA as the first specific marker of a subpopulation of "early c-ret" positive neurons characterized by medium-to-large diameters. This marker will allow further functional characterization of these neurons.


Subject(s)
Ganglia, Spinal/embryology , Maf Transcription Factors, Large/genetics , Mechanoreceptors/metabolism , Neural Crest/embryology , Proto-Oncogene Proteins c-ret/biosynthesis , Sensory Receptor Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Size , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/genetics , Genetic Markers/genetics , Maf Transcription Factors, Large/biosynthesis , Mechanoreceptors/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutagenesis, Insertional , Neural Crest/cytology , Proto-Oncogene Proteins c-ret/genetics , Sensory Receptor Cells/cytology
9.
PLoS One ; 5(12): e15272, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21203559

ABSTRACT

The B-raf proto-oncogene exerts essential functions during development and adulthood. It is required for various processes, such as placental development, postnatal nervous system myelination and adult learning and memory. The mouse B-raf gene encodes several isoforms resulting from alternative splicing of exons 8b and 9b located in the hinge region upstream of the kinase domain. These alternative sequences modulate the biochemical and biological properties of B-Raf proteins. To gain insight into the physiological importance of B-raf alternative splicing, we generated two conditional knockout mice of exons 8b and 9b. Homozygous animals with a constitutive deletion of either exon are healthy and fertile, and survive up to 18 months without any visible abnormalities, demonstrating that alternative splicing is not essential for embryonic development and brain myelination. However, behavioural analyses revealed that expression of exon 9b-containing isoforms is required for B-Raf function in hippocampal-dependent learning and memory. In contrast, mice mutated on exon 8b are not impaired in this function. Interestingly, our results suggest that exon 8b is present only in eutherians and its splicing is differentially regulated among species.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Hippocampus/metabolism , Learning , Memory , Proto-Oncogene Proteins B-raf/metabolism , Animals , Exons , Fear , Hippocampus/pathology , Homozygote , Mice , Mice, Knockout , Myelin Sheath/chemistry , NIH 3T3 Cells , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...