Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancer Cell ; 41(4): 660-677.e7, 2023 04 10.
Article de Anglais | MEDLINE | ID: mdl-37001527

RÉSUMÉ

Pediatric solid and central nervous system tumors are the leading cause of cancer-related death among children. Identifying new targeted therapies necessitates the use of pediatric cancer models that faithfully recapitulate the patient's disease. However, the generation and characterization of pediatric cancer models has significantly lagged behind adult cancers, underscoring the urgent need to develop pediatric-focused cell line resources. Herein, we establish a single-site collection of 261 cell lines, including 224 pediatric cell lines representing 18 distinct extracranial and brain childhood tumor types. We subjected 182 cell lines to multi-omics analyses (DNA sequencing, RNA sequencing, DNA methylation), and in parallel performed pharmacological and genetic CRISPR-Cas9 loss-of-function screens to identify pediatric-specific treatment opportunities and biomarkers. Our work provides insight into specific pathway vulnerabilities in molecularly defined pediatric tumor classes and uncovers biomarker-linked therapeutic opportunities of clinical relevance. Cell line data and resources are provided in an open access portal.


Sujet(s)
Tumeurs du cerveau , Enfant , Humains , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale
2.
Neoplasia ; 35: 100846, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36335802

RÉSUMÉ

Pediatric brain tumors are the leading cause of cancer-related death in children in the United States and contribute a disproportionate number of potential years of life lost compared to adult cancers. Moreover, survivors frequently suffer long-term side effects, including secondary cancers. The Children's Brain Tumor Network (CBTN) is a multi-institutional international clinical research consortium created to advance therapeutic development through the collection and rapid distribution of biospecimens and data via open-science research platforms for real-time access and use by the global research community. The CBTN's 32 member institutions utilize a shared regulatory governance architecture at the Children's Hospital of Philadelphia to accelerate and maximize the use of biospecimens and data. As of August 2022, CBTN has enrolled over 4700 subjects, over 1500 parents, and collected over 65,000 biospecimen aliquots for research. Additionally, over 80 preclinical models have been developed from collected tumors. Multi-omic data for over 1000 tumors and germline material are currently available with data generation for > 5000 samples underway. To our knowledge, CBTN provides the largest open-access pediatric brain tumor multi-omic dataset annotated with longitudinal clinical and outcome data, imaging, associated biospecimens, child-parent genomic pedigrees, and in vivo and in vitro preclinical models. Empowered by NIH-supported platforms such as the Kids First Data Resource and the Childhood Cancer Data Initiative, the CBTN continues to expand the resources needed for scientists to accelerate translational impact for improved outcomes and quality of life for children with brain and spinal cord tumors.


Sujet(s)
Tumeurs du cerveau , Qualité de vie , Adulte , Humains , Enfant , Tumeurs du cerveau/thérapie
3.
Mol Cell Biol ; 41(7): e0052620, 2021 06 23.
Article de Anglais | MEDLINE | ID: mdl-33903225

RÉSUMÉ

How mammalian neuronal identity is progressively acquired and reinforced during development is not understood. We have previously shown that loss of RP58 (ZNF238 or ZBTB18), a BTB/POZ-zinc finger-containing transcription factor, in the mouse brain leads to microcephaly, corpus callosum agenesis, and cerebellum hypoplasia and that it is required for normal neuronal differentiation. The transcriptional programs regulated by RP58 during this process are not known. Here, we report for the first time that in embryonic mouse neocortical neurons a complex set of genes normally expressed in other cell types, such as those from mesoderm derivatives, must be actively repressed in vivo and that RP58 is a critical regulator of these repressed transcriptional programs. Importantly, gene set enrichment analysis (GSEA) analyses of these transcriptional programs indicate that repressed genes include distinct sets of genes significantly associated with glioma progression and/or pluripotency. We also demonstrate that reintroducing RP58 in glioma stem cells leads not only to aspects of neuronal differentiation but also to loss of stem cell characteristics, including loss of stem cell markers and decrease in stem cell self-renewal capacities. Thus, RP58 acts as an in vivo master guardian of the neuronal identity transcriptome, and its function may be required to prevent brain disease development, including glioma progression.


Sujet(s)
Régulation de l'expression des gènes au cours du développement/physiologie , Glioblastome/métabolisme , Neurones/métabolisme , Protéines de répression/métabolisme , Animaux , Différenciation cellulaire/génétique , Mouvement cellulaire/génétique , Souris , Neurogenèse/physiologie , Névroglie/métabolisme , Protéines de répression/génétique
4.
Cell ; 183(7): 1962-1985.e31, 2020 12 23.
Article de Anglais | MEDLINE | ID: mdl-33242424

RÉSUMÉ

We report a comprehensive proteogenomics analysis, including whole-genome sequencing, RNA sequencing, and proteomics and phosphoproteomics profiling, of 218 tumors across 7 histological types of childhood brain cancer: low-grade glioma (n = 93), ependymoma (32), high-grade glioma (25), medulloblastoma (22), ganglioglioma (18), craniopharyngioma (16), and atypical teratoid rhabdoid tumor (12). Proteomics data identify common biological themes that span histological boundaries, suggesting that treatments used for one histological type may be applied effectively to other tumors sharing similar proteomics features. Immune landscape characterization reveals diverse tumor microenvironments across and within diagnoses. Proteomics data further reveal functional effects of somatic mutations and copy number variations (CNVs) not evident in transcriptomics data. Kinase-substrate association and co-expression network analysis identify important biological mechanisms of tumorigenesis. This is the first large-scale proteogenomics analysis across traditional histological boundaries to uncover foundational pediatric brain tumor biology and inform rational treatment selection.


Sujet(s)
Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Protéogénomique , Tumeurs du cerveau/immunologie , Enfant , Variations de nombre de copies de segment d'ADN/génétique , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Génome humain , Gliome/génétique , Gliome/anatomopathologie , Humains , Lymphocytes TIL/immunologie , Mutation/génétique , Grading des tumeurs , Récidive tumorale locale/anatomopathologie , Phosphoprotéines/métabolisme , Phosphorylation , ARN messager/génétique , ARN messager/métabolisme , Transcriptome/génétique
6.
Cell Rep ; 20(13): 3236-3247, 2017 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-28954238

RÉSUMÉ

How transcription factors (TFs) reprogram one cell lineage to another remains unclear. Here, we define chromatin accessibility changes induced by the proneural TF Ascl1 throughout conversion of fibroblasts into induced neuronal (iN) cells. Thousands of genomic loci are affected as early as 12 hr after Ascl1 induction. Surprisingly, over 80% of the accessibility changes occur between days 2 and 5 of the 3-week reprogramming process. This chromatin switch coincides with robust activation of endogenous neuronal TFs and nucleosome phasing of neuronal promoters and enhancers. Subsequent morphological and functional maturation of iN cells is accomplished with relatively little chromatin reconfiguration. By integrating chromatin accessibility and transcriptome changes, we built a network model of dynamic TF regulation during iN cell reprogramming and identified Zfp238, Sox8, and Dlx3 as key TFs downstream of Ascl1. These results reveal a singular, coordinated epigenomic switch during direct reprogramming, in contrast to stepwise cell fate transitions in development.


Sujet(s)
Chromatine/métabolisme , Fibroblastes/métabolisme , Neurones/métabolisme , Reprogrammation cellulaire , Humains
7.
Cancer Res ; 74(17): 4811-21, 2014 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-24994715

RÉSUMÉ

The Hedgehog (HH) signaling pathway represents an important class of emerging developmental signaling pathways that play critical roles in the genesis of a large number of human cancers. The pharmaceutical industry is currently focused on developing small molecules targeting Smoothened (Smo), a key signaling effector of the HH pathway that regulates the levels and activity of the Gli family of transcription factors. Although one of these compounds, vismodegib, is now FDA-approved for patients with advanced basal cell carcinoma, acquired mutations in Smo can result in rapid relapse. Furthermore, many cancers also exhibit a Smo-independent activation of Gli proteins, an observation that may underlie the limited efficacy of Smo inhibitors in clinical trials against other types of cancer. Thus, there remains a critical need for HH inhibitors with different mechanisms of action, particularly those that act downstream of Smo. Recently, we identified the FDA-approved anti-pinworm compound pyrvinium as a novel, potent (IC50, 10 nmol/L) casein kinase-1α (CK1α) agonist. We show here that pyrvinium is a potent inhibitor of HH signaling, which acts by reducing the stability of the Gli family of transcription factors. Consistent with CK1α agonists acting on these most distal components of the HH signaling pathway, pyrvinium is able to inhibit the activity of a clinically relevant, vismodegib -resistant Smo mutant, as well as the Gli activity resulting from loss of the negative regulator suppressor of fused. We go on to demonstrate the utility of this small molecule in vivo, against the HH-dependent cancer medulloblastoma, attenuating its growth and reducing the expression of HH biomarkers.


Sujet(s)
Protéines Hedgehog/métabolisme , Composés du pyrvinium/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Carcinome basocellulaire/traitement médicamenteux , Carcinome basocellulaire/métabolisme , Casein Kinase Ialpha/métabolisme , Lignée cellulaire , Cellules HEK293 , Humains , Médulloblastome/traitement médicamenteux , Médulloblastome/métabolisme , Souris , Souris nude , Cellules NIH 3T3 , Protéines oncogènes , Récepteurs couplés aux protéines G/métabolisme , Transactivateurs , Facteurs de transcription/métabolisme , Protéine à doigt de zinc GLI1
8.
J Cell Sci ; 125(Pt 18): 4383-94, 2012 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-22641694

RÉSUMÉ

Telomeres play crucial roles in the maintenance of genome integrity and control of cellular senescence. Most eukaryotic telomeres can be transcribed to generate a telomeric repeat-containing RNA (TERRA) that persists as a heterogeneous nuclear RNA and can be developmentally regulated. However, the precise function and regulation of TERRA in normal and cancer cell development remains poorly understood. Here, we show that TERRA accumulates in highly proliferating normal and cancer cells, and forms large nuclear foci, which are distinct from previously characterized markers of DNA damage or replication stress. Using a mouse model for medulloblastoma driven by chronic Sonic hedgehog (SHH) signaling, TERRA RNA was detected in tumor, but not adjacent normal cells using both RNA fluorescence in situ hybridization (FISH) and northern blotting. RNA FISH revealed the formation of TERRA foci (TERFs) in the nuclear regions of rapidly proliferating tumor cells. In the normal developing cerebellum, TERRA aggregates could also be detected in highly proliferating zones of progenitor neurons. SHH could enhance TERRA expression in purified granule progenitor cells in vitro, suggesting that proliferation signals contribute to TERRA expression in responsive tissue. TERRA foci did not colocalize with γH2AX foci, promyelocytic leukemia (PML) or Cajal bodies in mouse tumor tissue. We also provide evidence that TERRA is elevated in a variety of human cancers. These findings suggest that elevated TERRA levels reflect a novel early form of telomere regulation during replication stress and cancer cell evolution, and the TERRA RNA aggregates may form a novel nuclear body in highly proliferating mammalian cells.


Sujet(s)
Tumeurs du cervelet/anatomopathologie , Médulloblastome/génétique , Médulloblastome/anatomopathologie , Cellules souches neurales/métabolisme , ARN/génétique , Séquences répétées d'acides nucléiques/génétique , Télomère/génétique , Animaux , Encéphale/embryologie , Encéphale/anatomopathologie , Prolifération cellulaire , Tumeurs du cervelet/génétique , Corps de Cajal/métabolisme , Altération de l'ADN , Modèles animaux de maladie humaine , Régulation de l'expression des gènes tumoraux , Protéines Hedgehog/pharmacologie , Histone/métabolisme , Humains , Hybridation fluorescente in situ , Interphase , Souris , Modèles biologiques , Cellules souches neurales/anatomopathologie
9.
Development ; 139(11): 1903-9, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22513377

RÉSUMÉ

Cerebellum development depends on the correct differentiation of progenitors into neurons, a process controlled by a transcriptional program that remains poorly understood. Here we show that neural-specific deletion of the BTB/POZ zinc-finger transcription factor-encoding gene Rp58 (Znf238, Zfp238) causes severe cerebellar hypoplasia and developmental failure of Purkinje neurons, Bergmann glia and granule neurons. Deletion of Rp58 in mouse embryonic Atoh1(+) progenitors leads to strong defects in growth and foliation owing to its crucial role in the differentiation of granule neurons. Analysis of the Rp58 mutant at E14.5 demonstrates that Rp58 is required for the development of both glutamatergic and GABAergic neurons. Rp58 mutants show decreased proliferation of glutamatergic progenitors at E14.5. In addition, Rp58 ablation results in a reduced number of GABAergic Pax2(+) neurons at E16.5 together with defects in the transcriptional program of ventricular zone progenitors. Our results indicate that Rp58 is essential for the growth and organization of the cerebellum and regulates the development of both GABAergic and glutamatergic neurons.


Sujet(s)
Plan d'organisation du corps/physiologie , Cervelet/embryologie , Régulation de l'expression des gènes au cours du développement/physiologie , Neurogenèse/physiologie , Protéines de répression/physiologie , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice , Cervelet/croissance et développement , Délétion de gène , Régulation de l'expression des gènes au cours du développement/génétique , Immunohistochimie , Hybridation in situ , Souris , Réaction de polymérisation en chaine en temps réel , Protéines de répression/métabolisme , Cellules souches/cytologie , Cellules souches/métabolisme
10.
Tetrahedron ; 67(52): 10261-10266, 2011 Dec 30.
Article de Anglais | MEDLINE | ID: mdl-22199406

RÉSUMÉ

The design, synthesis and biological evaluation of new analogs of the naturally occurring compound cyclopamine, a Hedgehog signaling inhibitor, are described. Stucture-activity relationship studies lead to an evolving model for the pharmacophore of this medically promising compound class of anti-cancer chemotherapeutic agents.

11.
Org Lett ; 13(19): 5140-3, 2011 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-21905689

RÉSUMÉ

Previous work from our laboratory has established that the readily available steroid-based analog 2 of cyclopamine 1 is, like 1, a highly potent inhibitor of Hedgehog signaling. The first structure-activity relationship studies on 2, i.e., the synthesis and biological evaluation of both the C-17 epi analog 4 and the C-3 deoxy analog 11, both of which are more potent than cyclopamine 1, are described. The implications of these results for the emerging pharmacophore of these Sonic Hedgehog signaling inhibitors are discussed.


Sujet(s)
Protéines Hedgehog/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Stéroïdes/synthèse chimique , Lignée cellulaire , Humains , Modèles moléculaires , Structure moléculaire , Stéroïdes/pharmacologie , Relation structure-activité , Facteurs de transcription/génétique , Protéine à doigt de zinc GLI1
12.
Stem Cells ; 29(11): 1752-62, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21948558

RÉSUMÉ

Mouse and human somatic cells can either be reprogrammed to a pluripotent state or converted to another lineage with a combination of transcription factors suggesting that lineage commitment is a reversible process. Here we show that only one factor, the active intracellular form of Notch1, is sufficient to convert mature pigmented epidermal-derived melanocytes into functional multipotent neural crest (NC) stem-like cells. These induced NC stem cells (iNCSCs) proliferate as spheres under stem cell media conditions, re-express NC-related genes, and differentiate into multiple NC-derived mesenchymal and neuronal lineages. Moreover, iNCSCs are highly migratory and functional in vivo. These results demonstrate that mature melanocytes can be reprogrammed toward their primitive NC cell precursors through the activation of a single stem cell-related pathway. Reprogramming of melanocytes to iNCSCs may provide an alternate source of NCSCs for neuroregenerative applications.


Sujet(s)
Reprogrammation cellulaire/physiologie , Mélanocytes/cytologie , Mélanocytes/métabolisme , Crête neurale/cytologie , Cellules souches neurales/cytologie , Récepteur Notch1/métabolisme , Cellules souches/cytologie , Animaux , Technique de Western , Différenciation cellulaire/génétique , Différenciation cellulaire/physiologie , Lignée cellulaire , Mouvement cellulaire/génétique , Mouvement cellulaire/physiologie , Reprogrammation cellulaire/génétique , Embryon de poulet , Humains , Cellules souches neurales/métabolisme , Récepteur Notch1/génétique , Cellules souches/métabolisme
14.
Genome Res ; 21(8): 1260-72, 2011 Aug.
Article de Anglais | MEDLINE | ID: mdl-21712398

RÉSUMÉ

Despite our growing knowledge that many mammalian genes generate multiple transcript variants that may encode functionally distinct protein isoforms, the transcriptomes of various tissues and their developmental stages are poorly defined. Identifying the transcriptome and its regulation in a cell/tissue is the key to deciphering the cell/tissue-specific functions of a gene. We built a genome-wide inventory of noncoding and protein-coding transcripts (transcriptomes), their promoters (promoteromes) and histone modification states (epigenomes) for developing, and adult cerebella using integrative massive-parallel sequencing and bioinformatics approach. The data consists of 61,525 (12,796 novel) distinct mRNAs transcribed by 29,589 (4792 novel) promoters corresponding to 15,669 protein-coding and 7624 noncoding genes. Importantly, our results show that the transcript variants from a gene are predominantly generated using alternative transcriptional rather than splicing mechanisms, highlighting alternative promoters and transcriptional terminations as major sources of transcriptome diversity. Moreover, H3K4me3, and not H3K27me3, defined the use of alternative promoters, and we identified a combinatorial role of H3K4me3 and H3K27me3 in regulating the expression of transcripts, including transcript variants of a gene during development. We observed a strong bias of both H3K4me3 and H3K27me3 for CpG-rich promoters and an exponential relationship between their enrichment and corresponding transcript expression. Furthermore, the majority of genes associated with neurological diseases expressed multiple transcripts through alternative promoters, and we demonstrated aberrant use of alternative promoters in medulloblastoma, cancer arising in the cerebellum. The transcriptomes of developing and adult cerebella presented in this study emphasize the importance of analyzing gene regulation and function at the isoform level.


Sujet(s)
Épissage alternatif , Cervelet/croissance et développement , Transcription génétique , Transcriptome , Animaux , Tumeurs du cervelet/génétique , Tumeurs du cervelet/métabolisme , Cervelet/métabolisme , Biologie informatique , Épigenèse génétique , Régulation de l'expression des gènes au cours du développement , Génome , Médulloblastome/génétique , Médulloblastome/métabolisme , Souris , Lignées consanguines de souris , Régions promotrices (génétique) , ARN messager/métabolisme
15.
J Biol Chem ; 285(4): 2562-8, 2010 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-19920144

RÉSUMÉ

The hedgehog (HH) family of ligands plays an important instructional role in metazoan development. HH proteins are initially produced as approximately 45-kDa full-length proteins, which undergo an intramolecular cleavage to generate an amino-terminal product that subsequently becomes cholesterol-modified (HH-Np). It is well accepted that this cholesterol-modified amino-terminal cleavage product is responsible for all HH-dependent signaling events. Contrary to this model we show here that full-length forms of HH proteins are able to traffic to the plasma membrane and participate directly in cell-cell signaling, both in vitro and in vivo. We were also able to rescue a Drosophila eye-specific hh loss of function phenotype by expressing a full-length form of hh that cannot be processed into HH-Np. These results suggest that in some physiological contexts full-length HH proteins may participate directly in HH signaling and that this novel activity of full-length HH may be evolutionarily conserved.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Protéines Hedgehog , Transduction du signal/physiologie , Animaux , Communication cellulaire/physiologie , Embryon de poulet , Poulets , Drosophila , Protéines de Drosophila/composition chimique , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Évolution moléculaire , Protéines Hedgehog/composition chimique , Protéines Hedgehog/génétique , Protéines Hedgehog/métabolisme , Holoprosencéphalie/génétique , Holoprosencéphalie/physiopathologie , Humains , Mutagenèse dirigée , Tube neural/embryologie , Tube neural/physiologie , Récepteurs patched , Phénotype , Structure tertiaire des protéines , Transport des protéines/physiologie , Lapins , Récepteurs de surface cellulaire/métabolisme , Relation structure-activité
16.
Hum Genet ; 125(1): 95-103, 2009 Feb.
Article de Anglais | MEDLINE | ID: mdl-19057928

RÉSUMÉ

Sonic hedgehog (SHH) plays an important instructional role in vertebrate development, as exemplified by the numerous developmental disorders that occur when the SHH pathway is disrupted. Mutations in the SHH gene are the most common cause of sporadic and inherited holoprosencephaly (HPE), a developmental disorder that is characterized by defective prosencephalon development. SHH HPE mutations provide a unique opportunity to better understand SHH biogenesis and signaling, and to decipher its role in the development of HPE. Here, we analyzed a panel of SHH HPE missense mutations that encode changes in the amino-terminal active domain of SHH. Our results show that SHH HPE mutations affect SHH biogenesis and signaling at multiple steps, which broadly results in low levels of protein expression, defective processing of SHH into its active form and protein with reduced activity. Additionally, we found that some inactive SHH proteins were able to modulate the activity of wt SHH in a dominant negative manner, both in vitro and in vivo. These findings show for the first time the susceptibility of SHH driven developmental processes to perturbations by low-activity forms of SHH. In conclusion, we demonstrate that SHH mutations found in HPE patients affect distinct steps of SHH biogenesis to attenuate SHH activity to different levels, and suggest that these variable levels of SHH activity might contribute to some of the phenotypic variation found in HPE patients.


Sujet(s)
Protéines Hedgehog/génétique , Holoprosencéphalie/génétique , Prosencéphale/embryologie , Séquence d'acides aminés , Animaux , Embryon de poulet , Protéines Hedgehog/biosynthèse , Holoprosencéphalie/anatomopathologie , Humains , Données de séquences moléculaires , Mutation faux-sens , Prosencéphale/anatomopathologie , Alignement de séquences
17.
Nat Cell Biol ; 10(2): 186-93, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18157121

RÉSUMÉ

Rhythmic production of vertebral precursors, the somites, causes bilateral columns of embryonic segments to form. This process involves a molecular oscillator--the segmentation clock--whose signal is translated into a spatial, periodic pattern by a complex signalling gradient system within the presomitic mesoderm (PSM). In mouse embryos, Wnt signalling has been implicated in both the clock and gradient mechanisms, but how the Wnt pathway can perform these two functions simultaneously remains unclear. Here, we use a yellow fluorescent protein (YFP)-based, real-time imaging system in mouse embryos to demonstrate that clock oscillations are independent of beta-catenin protein levels. In contrast, we show that the Wnt-signalling gradient is established through a nuclear beta-catenin protein gradient in the posterior PSM. This gradient of nuclear beta-catenin defines the size of the oscillatory field and controls key aspects of PSM maturation and segment formation, emphasizing the central role of Wnt signalling in this process.


Sujet(s)
Horloges biologiques/physiologie , Plan d'organisation du corps/physiologie , Mésoderme/embryologie , Somites/embryologie , bêta-Caténine/métabolisme , Animaux , Noyau de la cellule/métabolisme , Mésoderme/métabolisme , Souris , Souris transgéniques , Mutation , Transduction du signal/physiologie , Somites/métabolisme , Protéines de type Wingless/métabolisme , bêta-Caténine/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...