Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Chem Biol ; 20(1): 19-29, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37308732

RESUMO

O-linked ß-N-acetyl glucosamine (O-GlcNAc) is at the crossroads of cellular metabolism, including glucose and glutamine; its dysregulation leads to molecular and pathological alterations that cause diseases. Here we report that O-GlcNAc directly regulates de novo nucleotide synthesis and nicotinamide adenine dinucleotide (NAD) production upon abnormal metabolic states. Phosphoribosyl pyrophosphate synthetase 1 (PRPS1), the key enzyme of the de novo nucleotide synthesis pathway, is O-GlcNAcylated by O-GlcNAc transferase (OGT), which triggers PRPS1 hexamer formation and relieves nucleotide product-mediated feedback inhibition, thereby boosting PRPS1 activity. PRPS1 O-GlcNAcylation blocked AMPK binding and inhibited AMPK-mediated PRPS1 phosphorylation. OGT still regulates PRPS1 activity in AMPK-deficient cells. Elevated PRPS1 O-GlcNAcylation promotes tumorigenesis and confers resistance to chemoradiotherapy in lung cancer. Furthermore, Arts-syndrome-associated PRPS1 R196W mutant exhibits decreased PRPS1 O-GlcNAcylation and activity. Together, our findings establish a direct connection among O-GlcNAc signals, de novo nucleotide synthesis and human diseases, including cancer and Arts syndrome.


Assuntos
Proteínas Quinases Ativadas por AMP , Processamento de Proteína Pós-Traducional , Humanos , Proteínas Quinases Ativadas por AMP/metabolismo , Fosforilação , Glucose , Nucleotídeos/metabolismo , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo
2.
Cancers (Basel) ; 14(22)2022 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-36428626

RESUMO

Neuroblastoma (NB) is a pediatric tumor of the peripheral nervous system. Approximately 80% of relapsed NB show RAS-MAPK pathway mutations that activate ERK, resulting in the promotion of cell proliferation and drug resistance. Ulixertinib, a first-in-class ERK-specific inhibitor, has shown promising antitumor activity in phase 1 clinical trials for advanced solid tumors. Here, we show that ulixertinib significantly and dose-dependently inhibits cell proliferation and colony formation in different NB cell lines, including PDX cells. Transcriptomic analysis revealed that ulixertinib extensively inhibits different oncogenic and neuronal developmental pathways, including EGFR, VEGF, WNT, MAPK, NGF, and NTRK1. The proteomic analysis further revealed that ulixertinib inhibits the cell cycle and promotes apoptosis in NB cells. Additionally, ulixertinib treatment significantly sensitized NB cells to the conventional chemotherapeutic agent doxorubicin. Furthermore, ulixertinib potently inhibited NB tumor growth and prolonged the overall survival of the treated mice in two different NB mice models. Our preclinical study demonstrates that ulixertinib, either as a single agent or in combination with current therapies, is a novel and practical therapeutic approach for NB.

3.
Clin Cancer Res ; 28(19): 4278-4291, 2022 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-35736214

RESUMO

PURPOSE: Patients with MYC-amplified medulloblastoma (MB) have poor prognosis and frequently develop recurrence, thus new therapeutic approaches to prevent recurrence are needed. EXPERIMENTAL DESIGN: We evaluated OLIG2 expression in a panel of mouse Myc-driven MB tumors, patient MB samples, and patient-derived xenograft (PDX) tumors and analyzed radiation sensitivity in OLIG2-high and OLIG2-low tumors in PDX lines. We assessed the effect of inhibition of OLIG2 by OLIG2-CRISPR or the small molecule inhibitor CT-179 combined with radiotherapy on tumor progression in PDX models. RESULTS: We found that MYC-associated MB can be stratified into OLIG2-high and OLIG2-low tumors based on OLIG2 protein expression. In MYC-amplified MB PDX models, OLIG2-low tumors were sensitive to radiation and rarely relapsed, whereas OLIG2-high tumors were resistant to radiation and consistently developed recurrence. In OLIG2-high tumors, irradiation eliminated the bulk of tumor cells; however, a small number of tumor cells comprising OLIG2- tumor cells and rare OLIG2+ tumor cells remained in the cerebellar tumor bed when examined immediately post-irradiation. All animals harboring residual-resistant tumor cells developed relapse. The relapsed tumors mirrored the cellular composition of the primary tumors with enriched OLIG2 expression. Further studies demonstrated that OLIG2 was essential for recurrence, as OLIG2 disruption with CRISPR-mediated deletion or with the small molecule inhibitor CT-179 prevented recurrence from the residual radioresistant tumor cells. CONCLUSIONS: Our studies reveal that OLIG2 is a biomarker and an effective therapeutic target in a high-risk subset of MYC-amplified MB, and OLIG2 inhibitor combined with radiotherapy represents a novel effective approach for treating this devastating disease.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Animais , Biomarcadores , Linhagem Celular Tumoral , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/genética , Modelos Animais de Doenças , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Meduloblastoma/radioterapia , Camundongos , Recidiva Local de Neoplasia/genética , Fator de Transcrição 2 de Oligodendrócitos/genética , Fator de Transcrição 2 de Oligodendrócitos/uso terapêutico , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
4.
J Transl Med ; 17(1): 321, 2019 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-31547819

RESUMO

BACKGROUND: Medulloblastoma (MB), the most common pediatric brain cancer, presents with a poor prognosis in a subset of patients with high risk disease, or at recurrence, where current therapies are ineffective. Cord blood (CB) natural killer (NK) cells may be promising off-the-shelf effector cells for immunotherapy due to their recognition of malignant cells without the need for a known target, ready availability from multiple banks, and their potential to expand exponentially. However, they are currently limited by immune suppressive cytokines secreted in the MB tumor microenvironment including Transforming Growth Factor ß (TGF-ß). Here, we address this challenge in in vitro models of MB. METHODS: CB-derived NK cells were modified to express a dominant negative TGF-ß receptor II (DNRII) using retroviral transduction. The ability of transduced CB cells to maintain function in the presence of medulloblastoma-conditioned media was then assessed. RESULTS: We observed that the cytotoxic ability of nontransduced CB-NK cells was reduced in the presence of TGF-ß-rich, medulloblastoma-conditioned media (21.21 ± 1.19% killing at E:T 5:1 in the absence vs. 14.98 ± 2.11% in the presence of medulloblastoma-conditioned media, n = 8, p = 0.02), but was unaffected in CB-derived DNRII-transduced NK cells (21.11 ± 1.84% killing at E:T 5:1 in the absence vs. 21.81 ± 3.37 in the presence of medulloblastoma-conditioned media, n = 8, p = 0.85. We also observed decreased expression of CCR2 in untransduced NK cells (mean CCR2 MFI 826 ± 117 in untransduced NK + MB supernatant from mean CCR2 MFI 1639.29 ± 215 in no MB supernatant, n = 7, p = 0.0156), but not in the transduced cells. Finally, we observed that CB-derived DNRII-transduced NK cells may protect surrounding immune cells by providing a cytokine sink for TGF-ß (decreased TGF-ß levels of 610 ± 265 pg/mL in CB-derived DNRII-transduced NK cells vs. 1817 ± 342 pg/mL in untransduced cells; p = 0.008). CONCLUSIONS: CB NK cells expressing a TGF-ß DNRII may have a functional advantage over unmodified NK cells in the presence of TGF-ß-rich MB, warranting further investigation on its potential applications for patients with medulloblastoma.


Assuntos
Neoplasias Cerebelares/imunologia , Células Matadoras Naturais/imunologia , Meduloblastoma/imunologia , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Sangue Fetal/citologia , Humanos , Células Matadoras Naturais/transplante , Testes de Neutralização , Receptores CCR2/metabolismo , Transplante Homólogo
5.
Cell Rep ; 26(11): 3132-3144.e7, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30865899

RESUMO

Identification of human disease signature genes typically requires samples from many donors to achieve statistical significance. Here, we show that single-cell heterogeneity analysis may overcome this hurdle by significantly improving the test sensitivity. We analyzed the transcriptome of 39,905 single islets cells from 9 donors and observed distinct ß cell heterogeneity trajectories associated with obesity or type 2 diabetes (T2D). We therefore developed RePACT, a sensitive single-cell analysis algorithm to identify both common and specific signature genes for obesity and T2D. We mapped both ß-cell-specific genes and disease signature genes to the insulin regulatory network identified from a genome-wide CRISPR screen. Our integrative analysis discovered the previously unrecognized roles of the cohesin loading complex and the NuA4/Tip60 histone acetyltransferase complex in regulating insulin transcription and release. Our study demonstrated the power of combining single-cell heterogeneity analysis and functional genomics to dissect the etiology of complex diseases.


Assuntos
Diabetes Mellitus Tipo 2/genética , Heterogeneidade Genética , Células Secretoras de Insulina/metabolismo , Transcriptoma , Animais , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Camundongos , Análise de Célula Única , Coesinas
6.
Cancer Res ; 79(8): 1967-1980, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30862721

RESUMO

A subset of group 3 medulloblastoma frequently harbors amplification or overexpression of MYC lacking additional focal aberrations, yet it remains unclear whether MYC overexpression alone can induce tumorigenesis and which cells give rise to these tumors. Here, we showed that astrocyte progenitors in the early postnatal cerebellum were susceptible to transformation by MYC. The resulting tumors specifically resembled human group 3 medulloblastoma based on histology and gene-expression profiling. Gene-expression analysis of MYC-driven medulloblastoma cells revealed altered glucose metabolic pathways with marked overexpression of lactate dehydrogenase A (LDHA). LDHA abundance correlated positively with MYC expression and was associated with poor prognosis in human group 3 medulloblastoma. Inhibition of LDHA significantly reduced growth of both mouse and human MYC-driven tumors but had little effect on normal cerebellar cells or SHH-associated medulloblastoma. By generating a new mouse model, we demonstrated for the first time that astrocyte progenitors can be transformed by MYC and serve as the cells of origin for group 3 medulloblastoma. Moreover, we identified LDHA as a novel, specific therapeutic target for this devastating disease. SIGNIFICANCE: Insights from a new model identified LDHA as a novel target for group 3 medulloblastoma, paving the way for the development of effective therapies against this disease.


Assuntos
Biomarcadores Tumorais/metabolismo , Transformação Celular Neoplásica/patologia , Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Células-Tronco/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Células-Tronco/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
J Neurooncol ; 142(3): 411-422, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30725256

RESUMO

PURPOSE: Pediatric brain cancer medulloblastoma (MB) standard-of-care results in numerous comorbidities. MB is comprised of distinct molecular subgroups. Group 3 molecular subgroup patients have the highest relapse rates and after standard-of-care have a 20% survival. Group 3 tumors have high expression of GABRA5, which codes for the α5 subunit of the γ-aminobutyric acid type A receptor (GABAAR). We are advancing a therapeutic approach for group 3 based on GABAAR modulation using benzodiazepine-derivatives. METHODS: We performed analysis of GABR and MYC expression in MB tumors and used molecular, cell biological, and whole-cell electrophysiology approaches to establish presence of a functional 'druggable' GABAAR in group 3 cells. RESULTS: Analysis of expression of 763 MB tumors reveals that group 3 tumors share high subgroup-specific and correlative expression of GABR genes, which code for GABAAR subunits α5, ß3 and γ2 and 3. There are ~ 1000 functional α5-GABAARs per group 3 patient-derived cell that mediate a basal chloride-anion efflux of 2 × 109 ions/s. Benzodiazepines, designed to prefer α5-GABAAR, impair group 3 cell viability by enhancing chloride-anion efflux with subtle changes in their structure having significant impact on potency. A potent, non-toxic benzodiazepine ('KRM-II-08') binds to the α5-GABAAR (0.8 µM EC50) enhancing a chloride-anion efflux that induces mitochondrial membrane depolarization and in response, TP53 upregulation and p53, constitutively phosphorylated at S392, cytoplasmic localization. This correlates with pro-apoptotic Bcl-2-associated death promoter protein localization. CONCLUSION: GABRA5 expression can serve as a diagnostic biomarker for group 3 tumors, while α5-GABAAR is a therapeutic target for benzodiazepine binding, enhancing an ion imbalance that induces apoptosis.


Assuntos
Benzodiazepinas/farmacologia , Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Receptores de GABA-A/química , Regulação Alostérica , Morte Celular/efeitos dos fármacos , Neoplasias Cerebelares/tratamento farmacológico , Neoplasias Cerebelares/metabolismo , Perfilação da Expressão Gênica , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/metabolismo , Receptores de GABA-A/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
Int J Mol Sci ; 17(10)2016 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-27775567

RESUMO

Brain tumors are the leading cause of cancer-related deaths in children, and medulloblastoma is the most prevalent malignant childhood/pediatric brain tumor. Providing effective treatment for these cancers, with minimal damage to the still-developing brain, remains one of the greatest challenges faced by clinicians. Understanding the diverse events driving tumor formation, maintenance, progression, and recurrence is necessary for identifying novel targeted therapeutics and improving survival of patients with this disease. Genomic copy number alteration data, together with clinical studies, identifies c-MYC amplification as an important risk factor associated with the most aggressive forms of medulloblastoma with marked metastatic potential. Yet despite this, very little is known regarding the impact of such genomic abnormalities upon the functional biology of the tumor cell. We discuss here how recent advances in quantitative proteomic techniques are now providing new insights into the functional biology of these aggressive tumors, as illustrated by the use of proteomics to bridge the gap between the genotype and phenotype in the case of c-MYC-amplified/associated medulloblastoma. These integrated proteogenomic approaches now provide a new platform for understanding cancer biology by providing a functional context to frame genomic abnormalities.


Assuntos
Neoplasias Cerebelares/genética , Regulação Neoplásica da Expressão Gênica , Meduloblastoma/genética , Recidiva Local de Neoplasia/genética , Proteogenômica/métodos , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Criança , Dosagem de Genes , Genótipo , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Camundongos , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Risco , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Cancer Cell ; 29(3): 311-323, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26977882

RESUMO

Medulloblastoma (MB) is a highly malignant pediatric brain tumor. Despite aggressive therapy, many patients succumb to the disease, and survivors experience severe side effects from treatment. MYC-driven MB has a particularly poor prognosis and would greatly benefit from more effective therapies. We used an animal model of MYC-driven MB to screen for drugs that decrease viability of tumor cells. Among the most effective compounds were histone deacetylase inhibitors (HDACIs). HDACIs potently inhibit survival of MYC-driven MB cells in vitro, in part by inducing expression of the FOXO1 tumor suppressor gene. HDACIs also synergize with phosphatidylinositol 3-kinase inhibitors to inhibit tumor growth in vivo. These studies identify an effective combination therapy for the most aggressive form of MB.


Assuntos
Proliferação de Células/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Meduloblastoma/tratamento farmacológico , Meduloblastoma/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Genes Supressores de Tumor/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL
10.
Clin Cancer Res ; 22(3): 582-95, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26405194

RESUMO

PURPOSE: Despite significant strides in the identification and characterization of potential therapeutic targets for medulloblastoma, the role of the immune system and its interplay with the tumor microenvironment within these tumors are poorly understood. To address this, we adapted two syngeneic animal models of human Sonic Hedgehog (SHH)-driven and group 3 medulloblastoma for preclinical evaluation in immunocompetent C57BL/6 mice. EXPERIMENTAL DESIGN AND RESULTS: Multicolor flow cytometric analyses were used to phenotype and characterize immune infiltrating cells within established cerebellar tumors. We observed significantly higher percentages of dendritic cells, infiltrating lymphocytes, myeloid-derived suppressor cells, and tumor-associated macrophages in murine SHH model tumors compared with group 3 tumors. However, murine group 3 tumors had higher percentages of CD8(+) PD-1(+) T cells within the CD3 population. PD-1 blockade conferred superior antitumor efficacy in animals bearing intracranial group 3 tumors compared with SHH group tumors, indicating that immunologic differences within the tumor microenvironment can be leveraged as potential targets to mediate antitumor efficacy. Further analysis of anti-PD-1 monoclonal antibody localization revealed binding to PD-1(+) peripheral T cells, but not tumor infiltrating lymphocytes within the brain tumor microenvironment. Peripheral PD-1 blockade additionally resulted in a marked increase in CD3(+) T cells within the tumor microenvironment. CONCLUSIONS: This is the first immunologic characterization of preclinical models of molecular subtypes of medulloblastoma and demonstration that response to immune checkpoint blockade differs across subtype classification. Our findings also suggest that effective anti-PD-1 blockade does not require that systemically administered antibodies penetrate the brain tumor microenvironment.


Assuntos
Antineoplásicos/farmacologia , Meduloblastoma/imunologia , Meduloblastoma/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Animais , Biomarcadores , Modelos Animais de Doenças , Imunofenotipagem , Contagem de Linfócitos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Meduloblastoma/mortalidade , Camundongos , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Receptores Patched , Fenótipo , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Microambiente Tumoral/genética
11.
Development ; 139(10): 1724-33, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22461560

RESUMO

The WNT pathway plays multiple roles in neural development and is crucial for establishment of the embryonic cerebellum. In addition, WNT pathway mutations are associated with medulloblastoma, the most common malignant brain tumor in children. However, the cell types within the cerebellum that are responsive to WNT signaling remain unknown. Here we investigate the effects of canonical WNT signaling on two important classes of progenitors in the developing cerebellum: multipotent neural stem cells (NSCs) and granule neuron precursors (GNPs). We show that WNT pathway activation in vitro promotes proliferation of NSCs but not GNPs. Moreover, mice that express activated ß-catenin in the cerebellar ventricular zone exhibit increased proliferation of NSCs in that region, whereas expression of the same protein in GNPs impairs proliferation. Although ß-catenin-expressing NSCs proliferate they do not undergo prolonged expansion or neoplastic growth; rather, WNT signaling markedly interferes with their capacity for self-renewal and differentiation. At a molecular level, mutant NSCs exhibit increased expression of c-Myc, which might account for their transient proliferation, but also express high levels of bone morphogenetic proteins and the cyclin-dependent kinase inhibitor p21, which might contribute to their altered self-renewal and differentiation. These studies suggest that the WNT pathway is a potent regulator of cerebellar stem cell growth and differentiation.


Assuntos
Cerebelo/citologia , Cerebelo/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Cerebelo/embriologia , Citometria de Fluxo , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
12.
Cancer Cell ; 21(2): 155-67, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22340590

RESUMO

Medulloblastoma (MB) is the most common malignant brain tumor in children. Patients whose tumors exhibit overexpression or amplification of the MYC oncogene (c-MYC) usually have an extremely poor prognosis, but there are no animal models of this subtype of the disease. Here, we show that cerebellar stem cells expressing Myc and mutant Trp53 (p53) generate aggressive tumors following orthotopic transplantation. These tumors consist of large, pleiomorphic cells and resemble human MYC-driven MB at a molecular level. Notably, antagonists of PI3K/mTOR signaling, but not Hedgehog signaling, inhibit growth of tumor cells. These findings suggest that cerebellar stem cells can give rise to MYC-driven MB and identify a novel model that can be used to test therapies for this devastating disease.


Assuntos
Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Aminopiridinas/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Neoplasias Cerebelares/tratamento farmacológico , Cerebelo/patologia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Genes p53/fisiologia , Imidazóis/farmacologia , Meduloblastoma/tratamento farmacológico , Camundongos , Morfolinas/farmacologia , Células-Tronco Neurais/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Quinolinas/farmacologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/fisiologia
13.
Nature ; 468(7327): 1095-9, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-21150899

RESUMO

Medulloblastoma encompasses a collection of clinically and molecularly diverse tumour subtypes that together comprise the most common malignant childhood brain tumour. These tumours are thought to arise within the cerebellum, with approximately 25% originating from granule neuron precursor cells (GNPCs) after aberrant activation of the Sonic Hedgehog pathway (hereafter, SHH subtype). The pathological processes that drive heterogeneity among the other medulloblastoma subtypes are not known, hindering the development of much needed new therapies. Here we provide evidence that a discrete subtype of medulloblastoma that contains activating mutations in the WNT pathway effector CTNNB1 (hereafter, WNT subtype) arises outside the cerebellum from cells of the dorsal brainstem. We found that genes marking human WNT-subtype medulloblastomas are more frequently expressed in the lower rhombic lip (LRL) and embryonic dorsal brainstem than in the upper rhombic lip (URL) and developing cerebellum. Magnetic resonance imaging (MRI) and intra-operative reports showed that human WNT-subtype tumours infiltrate the dorsal brainstem, whereas SHH-subtype tumours are located within the cerebellar hemispheres. Activating mutations in Ctnnb1 had little impact on progenitor cell populations in the cerebellum, but caused the abnormal accumulation of cells on the embryonic dorsal brainstem which included aberrantly proliferating Zic1(+) precursor cells. These lesions persisted in all mutant adult mice; moreover, in 15% of cases in which Tp53 was concurrently deleted, they progressed to form medulloblastomas that recapitulated the anatomy and gene expression profiles of human WNT-subtype medulloblastoma. We provide the first evidence, to our knowledge, that subtypes of medulloblastoma have distinct cellular origins. Our data provide an explanation for the marked molecular and clinical differences between SHH- and WNT-subtype medulloblastomas and have profound implications for future research and treatment of this important childhood cancer.


Assuntos
Tronco Encefálico/patologia , Neoplasias Cerebelares/patologia , Meduloblastoma/patologia , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Mutação , beta Catenina/genética
14.
Cancer Cell ; 18(6): 546-7, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21156279

RESUMO

Recent studies have suggested that brain tumors arise from neural stem cells and are maintained by stem-like tumor-initiating cells (TICs). In this issue of Cancer Cell, Persson et al. report that oligodendrogliomas, unlike malignant astrocytomas, originate from-and are propagated by-cells that resemble oligodendrocyte progenitors.

15.
Neuroreport ; 15(12): 1847-50, 2004 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-15305122

RESUMO

The transplantation of dopaminergic (DA) neurons is used for treating Parkinson's disease. However, their actual application is restricted by a limited source of DA cells. Here we report that DA cells can be increased 5- to 10-fold in vitro by the soluble factors from cortex in early developmental stages, which is much more than any previously identified growth factors such as BDNF, GDNF and NT3. We also show that the effect of the soluble factors from cortex is stronger than those of midbrain at embryonic early developmental ages. In contrast, at middle ages the soluble factors from midbrain present a much stronger effect. These findings suggest that the development of DA cells may be regulated by growth factors in a complex spatial and temporal network.


Assuntos
Diferenciação Celular/fisiologia , Córtex Cerebral/química , Dopamina/metabolismo , Mesencéfalo/citologia , Neurônios/fisiologia , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Tamanho Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , AMP Cíclico/farmacologia , Embrião de Mamíferos , Imuno-Histoquímica/métodos , Mesencéfalo/embriologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/efeitos dos fármacos , Ratos , Tirosina 3-Mono-Oxigenase/metabolismo
16.
Neuroreport ; 15(10): 1643-7, 2004 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-15232299

RESUMO

Heparin plays an important role in the survival and differentiation of mesencephalic progenitors mediated by FGF-2 in vitro. If the heparin concentration is gradually increased, cell survival mediated by FGF-2 can be greatly enhanced, to a maximum concentration of 20 ng/ml FGF-2 from 5 microg/ml heparin. However, differentiation of FGF-2 responsive mesencephalic progenitors is inhibited by heparin. When cortical, mesencephalic and hippocampal astrocytes were primed with FGF-2 and heparin, the latter two astrocytes promoted the differentiation of TH-positive neurons from mesencephalic progenitors. RT-PCR analysis showed that FGFR1, FGFR2 and FGFR3 were expressed in the cortical astrocytes, but only FGFR1 and FGFR3 were expressed in the mesencephalic and hippocampal astrocytes.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/fisiologia , Heparina/farmacologia , Mesencéfalo/citologia , Células-Tronco/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Contagem de Células/métodos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura/métodos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Regulação da Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica/métodos , RNA Mensageiro/biossíntese , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Células-Tronco/citologia , Sais de Tetrazólio , Tiazóis , Tubulina (Proteína)/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
17.
Neuroreport ; 14(12): 1567-70, 2003 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-14502077

RESUMO

Extrinsic signals play an important role in the differentiation of neural progenitor cells, but little is known about the underlying mechanism. In order to investigate how extrinsic signals influence the fate switch of progenitors during development, we cultured ventral mesencephalic progenitors from E13 and E16 rats in the presence of soluble factors. Here we report that soluble factors in early developmental stages can induce the production of dopaminergic neurons. E16 may be an important developmental stage in which the responsiveness of the progenitors to the soluble factors is much more sensitive. Our results indicate that a combination of cell-intrinsic changes and extrinsic cues controls the competence of mesencephalic progenitors to produce dopaminergic neurons. Cell fate restriction may result from a series of extrinsic factors acting on a multipotent progenitor progressively.


Assuntos
Diferenciação Celular/fisiologia , Dopamina/fisiologia , Mesencéfalo/citologia , Mesencéfalo/embriologia , Neurônios/citologia , Animais , Células Cultivadas , Mesencéfalo/fisiologia , Neurônios/fisiologia , Ratos , Células-Tronco/citologia , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA