Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Oncogene ; 40(2): 396-407, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33159168

RESUMEN

The immune microenvironment of tumors can play a critical role in promoting or inhibiting tumor progression depending on the context. We present evidence that tumor-associated macrophages/microglia (TAMs) can promote tumor progression in the sonic hedgehog subgroup of medulloblastoma (SHH-MB). By combining longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) and immune profiling of a sporadic mouse model of SHH-MB, we found the density of TAMs is higher in the ~50% of tumors that progress to lethal disease. Furthermore, reducing regulatory T cells or eliminating B and T cells in Rag1 mutants does not alter SHH-MB tumor progression. As TAMs are a dominant immune component in tumors and are normally dependent on colony-stimulating factor 1 receptor (CSF1R), we treated mice with a CSF1R inhibitor, PLX5622. Significantly, PLX5622 reduces a subset of TAMs, prolongs mouse survival, and reduces the volume of most tumors within 4 weeks of treatment. Moreover, concomitant with a reduction in TAMs the percentage of infiltrating cytotoxic T cells is increased, indicating a change in the tumor environment. Our studies in an immunocompetent preclinical mouse model demonstrate TAMs can have a functional role in promoting SHH-MB progression. Thus, CSF1R inhibition could have therapeutic potential for a subset of SHH-MB patients.


Asunto(s)
Neoplasias Cerebelosas/prevención & control , Modelos Animales de Enfermedad , Proteínas Hedgehog/fisiología , Meduloblastoma/prevención & control , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Macrófagos Asociados a Tumores/inmunología , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proliferación Celular , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Femenino , Humanos , Masculino , Meduloblastoma/etiología , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pronóstico , Células Tumorales Cultivadas , Microambiente Tumoral
2.
J Neurooncol ; 142(3): 395-407, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30788681

RESUMEN

PURPOSE: Medulloblastoma (MB) is the most common malignant brain tumor in children. Recent studies have shown the ability of natural killer (NK) cells to lyse MB cell lines in vitro, but in vivo successes remain elusive and the efficacy and fate of NK cells in vivo remain unknown. METHODS: To address these questions, we injected MB cells into the cerebellum of immunodeficient mice and examined tumor growth at various days after tumor establishment via bioluminescence imaging. NK cells were labeled with a fluorine-19 (19F) MRI probe and subsequently injected either intratumorally or contralaterally to the tumor in the cerebellum and effect on tumor growth was monitored. RESULTS: The 19F probe efficiently labeled the NK cells and exhibited little cytotoxicity. Fluorine-19 MRI confirmed the successful and accurate delivery of the labeled NK cells to the cerebellum of the mice. Administration of 19F-labeled NK cells suppressed MB growth, with the same efficacy as unlabeled cells. Immunohistochemistry confirmed the presence of NK cells within the tumor, which was associated with induction of apoptosis in tumor cells. NK cell migration to the tumor from a distal location as well as activation of apoptosis was also demonstrated by immunohstochemistry. CONCLUSIONS: Our results show that NK cells present a novel opportunity for new strategies in MB treatment. Further, 19F-labeled NK cells can suppress MB growth while enabling 19F MRI to provide imaging feedback that can facilitate study and optimization of therapeutic paradigms.


Asunto(s)
Neoplasias Cerebelosas/prevención & control , Monitoreo de Drogas/métodos , Radioisótopos de Flúor/uso terapéutico , Células Asesinas Naturales/trasplante , Imagen por Resonancia Magnética/métodos , Meduloblastoma/prevención & control , Animales , Apoptosis , Proliferación Celular , Neoplasias Cerebelosas/inmunología , Neoplasias Cerebelosas/patología , Humanos , Meduloblastoma/inmunología , Meduloblastoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Res ; 77(6): 1416-1426, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28031228

RESUMEN

The CXCR4 chemokine and Sonic Hedgehog (SHH) morphogen pathways are well-validated therapeutic targets in cancer, including medulloblastoma. However, single-agent treatments with SHH or CXCR4 antagonists have not proven efficacious in clinical trials to date. Here, we discovered that dual inhibition of the SHH and CXCR4 pathways in a murine model of SHH-subtype medulloblastoma exerts potent antitumor effects. This therapeutic synergy resulted in the suppression of tumor-propagating cell function and correlated with increased histone H3 lysine 27 trimethylation within the promoters of stem cell genes, resulting in their decreased expression. These results demonstrate that CXCR4 contributes to the epigenetic regulation of a tumor-propagating cell phenotype. Moreover, they provide a mechanistic rationale to evaluate the combination of SHH and CXCR4 inhibitors in clinical trials for the treatment of medulloblastoma, as well as other cancers driven by SHH that coexpress high levels of CXCR4. Cancer Res; 77(6); 1416-26. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Reprogramación Celular , Neoplasias Cerebelosas/prevención & control , Proteínas Hedgehog/antagonistas & inhibidores , Meduloblastoma/prevención & control , Receptores CXCR4/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Receptores CXCR4/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Biol Chem ; 289(51): 35494-502, 2014 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-25355313

RESUMEN

Epigenetic enzymes modulate signal transduction pathways in different biological contexts. We reasoned that epigenetic regulators might modulate the Hedgehog (HH) signaling pathway, a main driver of cell proliferation in various cancers including medulloblastoma. To test this hypothesis, we performed an unbiased small-molecule screen utilizing an HH-dependent reporter cell line (Light2 cells). We incubated Light2 cells with small molecules targeting different epigenetic modulators and identified four histone deacetylase inhibitors and a bromodomain and extra terminal domain (BET) protein inhibitor (I-BET151) that attenuate HH activity. I-BET151 was also able to inhibit the expression of HH target genes in Sufu(-/-) mouse embryonic fibroblasts, in which constitutive Gli activity is activated in a Smoothened (Smo)-independent fashion, consistent with it acting downstream of Smo. Knockdown of Brd4 (which encodes one of the BET proteins) phenocopies I-BET151 treatment, suggesting that Brd4 is a regulator of the HH signaling pathway. Consistent with this suggestion, Brd4 associates with the proximal promoter region of the Gli1 locus, and does so in a manner that can be reversed by I-BET151. Importantly, I-BET151 also suppressed the HH activity-dependent growth of medulloblastoma cells, in vitro and in vivo. These studies suggest that BET protein modulation may be an attractive therapeutic strategy for attenuating the growth of HH-dependent cancers, such as medulloblastoma.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas Hedgehog/genética , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Meduloblastoma/prevención & control , Receptores Acoplados a Proteínas G/genética , Animales , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones Noqueados , Ratones Desnudos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Smoothened , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1
5.
Oncotarget ; 5(9): 2355-71, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24796395

RESUMEN

Medulloblastoma is a pediatric brain tumor with a variable prognosis due to clinical and genomic heterogeneity. Among the 4 major genomic sub-groups, patients with MYC amplified tumors have a particularly poor prognosis despite therapy with surgery, radiation and chemotherapy. Targeting the MYC oncogene has traditionally been problematic. Here we report that MYC driven medulloblastoma can be targeted by inhibition of the bromodomain protein BRD4. We show that bromodomain inhibition with JQ1 restricts c-MYC driven transcriptional programs in medulloblastoma, suppresses medulloblastoma cell growth and induces a cell cycle arrest. Importantly JQ1 suppresses stem cell associated signaling in medulloblastoma cells and inhibits medulloblastoma tumor cell self-renewal. Additionally JQ1 also promotes senescence in medulloblastoma cells by activating cell cycle kinase inhibitors and inhibiting activity of E2F1. Furthermore BRD4 inhibition displayed an anti-proliferative, pro-senescence effect in a medulloblastoma model in vivo. In clinical samples we found that transcriptional programs suppressed by JQ1 are associated with adverse risk in medulloblastoma patients. Our work indicates that BRD4 inhibition attenuates stem cell signaling in MYC driven medulloblastoma and demonstrates the feasibility BET domain inhibition as a therapeutic approach in vivo.


Asunto(s)
Proliferación Celular , Neoplasias Cerebelosas/prevención & control , Meduloblastoma/prevención & control , Células Madre Neoplásicas/patología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Animales , Apoptosis , Azepinas/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Ciclo Celular , Proteínas de Ciclo Celular , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/patología , Niño , Perfilación de la Expresión Génica , Humanos , Meduloblastoma/genética , Meduloblastoma/mortalidad , Meduloblastoma/patología , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Triazoles/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Acta Oncol ; 53(4): 462-70, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24274390

RESUMEN

BACKGROUND: A mathematical framework is presented for simultaneously quantifying and evaluating the trade-off between tumor control and late complications for risk-based radiation therapy (RT) decision-support. To demonstrate this, we estimate life years lost (LYL) attributable to tumor recurrence, late cardiac toxicity and secondary cancers for standard-risk pediatric medulloblastoma (MB) patients and compare the effect of dose re-distribution on a common scale. METHODS: Total LYL were derived, based on the LYL attributable to radiation-induced late complications and the LYL from not controlling the primary disease. We compared the estimated LYL for three different treatments in 10 patients: 1) standard 3D conformal RT; 2) proton therapy; 3) risk-adaptive photon treatment lowering the dose to part of the craniospinal (CS) target volume situated close to critical risk organs. RESULTS: Late toxicity is important, with 0.75 LYL (95% CI 0.60-7.2 years) for standard uniform 24 Gy CS irradiation. However, recurrence risk dominates the total LYL with 14.2 years (95% CI 13.4-16.6 years). Compared to standard treatment, a risk-adapted strategy prescribing 12 Gy to the spinal volume encompassing the 1st-10th thoracic vertebrae (Th1-Th10), and 36 Gy to the remaining CS volume, estimated a LYL reduction of 0.90 years (95% CI -0.18-2.41 years). Proton therapy with 36 Gy to the whole CS volume was associated with significantly fewer LYL compared to the risk-adapted photon strategies, with a mean LYL difference of 0.50 years (95% CI 0.25-2.60 years). CONCLUSIONS: Optimization of RT prescription strategies considering both late complications and the risk of recurrence, an all-cause mortality dose painting approach, was demonstrated. The risk-adapted techniques compared favorably to the standard, and although in this context, the gain is small compared to estimated uncertainty, this study demonstrates a framework for all-cause mortality risk estimation, rather than evaluates direct clinical applicability of risk-adapted strategies.


Asunto(s)
Neoplasias Cerebelosas/radioterapia , Irradiación Craneoespinal/efectos adversos , Meduloblastoma/radioterapia , Modelos Teóricos , Recurrencia Local de Neoplasia/diagnóstico , Traumatismos por Radiación/etiología , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/prevención & control , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Masculino , Meduloblastoma/mortalidad , Meduloblastoma/prevención & control , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Fotones/efectos adversos , Pronóstico , Terapia de Protones/efectos adversos , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador/efectos adversos , Radioterapia Conformacional/efectos adversos , Radioterapia de Intensidad Modulada/efectos adversos , Factores de Riesgo , Tasa de Supervivencia , Adulto Joven
7.
Am J Clin Oncol ; 37(1): 1-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23111362

RESUMEN

OBJECTIVE: To assess the survival and role of adjuvant chemotherapy in adult medulloblastoma. METHODS: We reviewed outcomes of 66 patients (aged 18 y or more; median age, 33 y) with medulloblastoma. Forty-four (67%) patients had M0 disease, 9 had M1-M4, and 13 had MX. Thirty-one patients each for whom risk stratification was available were classified as high risk or standard risk. Fifty-six patients had histologic results: classic histology was the most common (n=46 [84%]), followed by desmoplastic (n=9), and large cell/anaplastic (n=1). Overall survival (OS) and progression-free survival (PFS) were estimated with Kaplan-Meier curves and log-rank tests. Cox regression analysis was used to compare recurrences. RESULTS: Median follow-up was 6.7 years. The estimated 5-year OS and PFS were 74% and 59%, respectively. High-risk versus standard-risk classification was associated with worse OS (61% vs. 86%; P=0.03) and recurrence (hazard ratio, 2.56; P=0.05) and a trend for worse PFS (49% vs. 69%; P=0.13). Gross total resection was associated with improved OS (P=0.03) and a trend toward improved PFS (P=0.09). No chemotherapy benefit could be demonstrated for the group as a whole. For high-risk patients with classic histology (n=25), chemotherapy was associated with a trend for improvement in 5-year PFS from 36% to 71% (P=0.10) and in 5-year OS from 49% to 100% (P=0.08). CONCLUSIONS: In adult patients with medulloblastoma, the extent of resection and risk classification predicts the outcome. These results suggest a chemotherapy benefit for high-risk patients with classic histology.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/prevención & control , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/prevención & control , Recurrencia Local de Neoplasia/prevención & control , Adulto , Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/cirugía , Quimioterapia Adyuvante , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Infratentoriales/diagnóstico , Estimación de Kaplan-Meier , Masculino , Meduloblastoma/mortalidad , Meduloblastoma/patología , Meduloblastoma/cirugía , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Radioterapia Adyuvante , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Resultado del Tratamiento
8.
Neuro Oncol ; 15(1): 83-90, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23172372

RESUMEN

Medulloblastoma is the most common malignant brain tumor in children, and a substantial number of patients die as a result of tumor progression. Overexpression of CDK6 is present in approximately one-third of medulloblastomas and is an independent poor prognostic marker for this disease. MicroRNA (miR)-124 inhibits expression of CDK6 and prevents proliferation of glioblastoma and medulloblastoma cells in vitro. We examined the effects of miR-124 overexpression on medulloblastoma cells both in vitro and in vivo and compared cell lines that have low and high CDK6 expression. MiR-124 overexpression inhibits the proliferation of medulloblastoma cells, and this effect is mediated mostly through the action of miR-124 upon CDK6. We further show that induced expression of miR-124 potently inhibits growth of medulloblastoma xenograft tumors in rodents. Further testing of miR-124 will help define the ultimate therapeutic potential of preclinical models of medulloblastoma in conjunction with various delivery strategies for treatment.


Asunto(s)
Ciclo Celular , Proliferación Celular , Neoplasias Cerebelosas/prevención & control , Meduloblastoma/prevención & control , MicroARNs/genética , Animales , Apoptosis , Western Blotting , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Neuro Oncol ; 14(7): 859-69, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22591662

RESUMEN

Medulloblastomas account for 20% of pediatric brain tumors. With an overall survival of 40%-70%, their treatment is still a challenge. The majority of medulloblastomas lack p53 mutations, but even in cancers retaining wild-type p53, the tumor surveillance function of p53 is inhibited by the oncoprotein MDM2. Deregulation of the MDM2/p53 balance leads to malignant transformation. Here, we analyzed MDM2 mRNA and protein expression in primary medulloblastomas and normal cerebellum and assessed the mutational status of p53 and MDM2 expression in 6 medulloblastoma cell lines. MDM2 expression was elevated in medulloblastomas, compared with cerebellum. Four of 6 medulloblastoma cell lines expressed wild-type p53 and high levels of MDM2. The tumor-promoting p53-MDM2 interaction can be inhibited by the small molecule, nutlin-3, restoring p53 function. Targeting the p53-MDM2 axis using nutlin-3 significantly reduced cell viability and induced either cell cycle arrest or apoptosis and expression of the p53 target gene p21 in these 4 cell lines. In contrast, DAOY and UW-228 cells harboring TP53 mutations were almost unaffected by nutlin-3 treatment. MDM2 knockdown in medulloblastoma cells by siRNA mimicked nutlin-3 treatment, whereas expression of dominant negative p53 abrogated nutlin-3 effects. Oral nutlin-3 treatment of mice with established medulloblastoma xenografts inhibited tumor growth and significantly increased survival. Thus, nutlin-3 reduced medulloblastoma cell viability in vitro and in vivo by re-activating p53 function. We suggest that inhibition of the MDM2-p53 interaction with nutlin-3 is a promising therapeutic option for medulloblastomas with functional p53 that should be further evaluated in clinical trials.


Asunto(s)
Neoplasias Cerebelosas/prevención & control , Imidazoles/farmacología , Meduloblastoma/prevención & control , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Estudios de Casos y Controles , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Cerebelo/metabolismo , Femenino , Humanos , Técnicas para Inmunoenzimas , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Desnudos , Mutación/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Análisis de Matrices Tisulares , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 108(41): 17123-8, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21969549

RESUMEN

Over half of all human cancers, of a wide variety of types, sustain mutations in the p53 tumor suppressor gene. Although p53 limits tumorigenesis through the induction of apoptosis or cell cycle arrest, its molecular mechanism of action in tumor suppression has been elusive. The best-characterized p53 activity in vitro is as a transcriptional activator, but the identification of numerous additional p53 biochemical activities in vitro has made it unclear which mechanism accounts for tumor suppression. Here, we assess the importance of transcriptional activation for p53 tumor suppression function in vivo in several tissues, using a knock-in mouse strain expressing a p53 mutant compromised for transcriptional activation, p53(25,26). p53(25,26) is severely impaired for the transactivation of numerous classical p53 target genes, including p21, Noxa, and Puma, but it retains the ability to activate a small subset of p53 target genes, including Bax. Surprisingly, p53(25,26) can nonetheless suppress tumor growth in cancers derived from the epithelial, mesenchymal, central nervous system, and lymphoid lineages. Therefore, full transactivation of most p53 target genes is dispensable for p53 tumor suppressor function in a range of tissue types. In contrast, a transcriptional activation mutant that is completely defective for transactivation, p53(25,26,53,54), fails to suppress tumor development. These findings demonstrate that transcriptional activation is indeed broadly critical for p53 tumor suppressor function, although this requirement reflects the limited transcriptional activity observed with p53(25,26) rather than robust transactivation of a full complement of p53 target genes.


Asunto(s)
Genes p53 , Neoplasias/genética , Neoplasias/prevención & control , Animales , Linaje de la Célula/genética , Técnicas de Sustitución del Gen , Humanos , Linfoma de Células B/genética , Linfoma de Células B/prevención & control , Meduloblastoma/genética , Meduloblastoma/prevención & control , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Activación Transcripcional
11.
Biochem Biophys Res Commun ; 403(3-4): 264-9, 2010 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-21073863

RESUMEN

Medulloblastoma (MB) is the most devastating and common pediatric brain tumor. Tumor cells invading into surrounding tissue and disseminating through cerebrospinal fluid make treatment extremely difficult. Identifying the mechanisms of MB cells is therefore imperative for the development of novel treatments. A research group demonstrated recently that the multifunctional signaling protein neurotrophin receptor p75(NTR) is a central regulator for glioma invasion. γ-secretase mediated processing of the p75(NTR) is a major contributor to the highly invasive nature of malignant gliomas. In this study we examine the p75(NTR) expression and processing in medulloblastoma cells. Results show that p75(NTR) is a critical regulator of medulloblastoma spinal metastasis. γ-secretase inhibitor, which blocks p75(NTR) proteolytic processing, significantly abrogates p75(NTR) induced medulloblastoma migration and invasion in vitro and in vivo. This data suggests that p75(NTR) is also an important therapeutic target for MB. γ-secretase inhibitor may be a potentially effective clinical application for the treatment of medulloblastoma spinal metastasis.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Neoplasias Cerebelosas/patología , Inhibidores Enzimáticos/uso terapéutico , Meduloblastoma/prevención & control , Meduloblastoma/secundario , Receptor de Factor de Crecimiento Nervioso/metabolismo , Neoplasias de la Columna Vertebral/prevención & control , Neoplasias de la Columna Vertebral/secundario , Animales , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia
12.
Proc Natl Acad Sci U S A ; 107(30): 13432-7, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20624968

RESUMEN

Aberrant Hedgehog (Hh) pathway activation has been implicated in cancers of diverse tissues and organs, and the tumor growth-inhibiting effects of pathway antagonists in animal models have stimulated efforts to develop pathway antagonists for human therapeutic purposes. These efforts have focused largely on cyclopamine derivatives or other compounds that mimic cyclopamine action in binding to and antagonizing Smoothened, a membrane transductory component. We report here that arsenicals, in contrast, antagonize the Hh pathway by targeting Gli transcriptional effectors; in the short term, arsenic blocks Hh-induced ciliary accumulation of Gli2, the primary activator of Hh-dependent transcription, and with prolonged incubation arsenic reduces steady-state levels of Gli2. Arsenicals active in Hh pathway antagonism include arsenic trioxide (ATO), a curative agent in clinical use for acute promyelocytic leukemia (APL); in our studies, ATO inhibited growth of Hh pathway-driven medulloblastoma allografts derived from Ptch+/-p53-/- mice within a range of serum levels comparable to those achieved in treatment of human APL. Arsenic thus could be tested rapidly as a therapeutic agent in malignant diseases associated with Hh pathway activation and could be particularly useful in such diseases that are inherently resistant or have acquired resistance to cyclopamine mimics.


Asunto(s)
Arsenicales/farmacología , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Trióxido de Arsénico , Arsenitos/farmacología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/prevención & control , Relación Dosis-Respuesta a Droga , Proteínas Hedgehog/genética , Factores de Transcripción de Tipo Kruppel/genética , Luciferasas/genética , Luciferasas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Meduloblastoma/genética , Meduloblastoma/patología , Meduloblastoma/prevención & control , Ratones , Ratones Noqueados , Ratones Desnudos , Células 3T3 NIH , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Óxidos/farmacología , Estabilidad Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Compuestos de Sodio/farmacología , Transfección , Trasplante Homólogo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína Gli2 con Dedos de Zinc
13.
Int J Cancer ; 127(12): 2749-57, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21351254

RESUMEN

Medulloblastoma (MB) is the most common pediatric tumor of the CNS, representing ∼20% of all childhood CNS tumors. Although in recent years many molecular mechanisms that control MB development have been clarified, the effects of biological factors such as sex on this tumor remain to be explained. Epidemiological data, in fact, indicate a significant difference in the incidence of MB between the 2 sexes, with considerably higher susceptibility of males than females. Besides this different susceptibility, female sex is also a significant favorable prognostic factor in MB, with girls having a much better outcome. Despite these literature data, there has been little investigation into estrogen influence on MB development. In our study, we evaluated how hormone deficiency resulting from ovariectomy and hormone replacement influences the development of early and advanced MB stages in Patched1 heterozygous mice, a well-characterized mouse model of radiation-induced MB. Susceptibility to MB development was significantly increased in ovariectomized Ptch1(+/-) females and restored to levels observed in control mice after estrogen replacement. We next investigated the molecular mechanisms by which estrogen might influence tumor progression and show that ERß, but not ERα, is involved in modulation of MB development by estrogens. Finally, our study shows that a functional interaction between estrogen- and IGF-I-mediated pathways may be responsible for the effects observed.


Asunto(s)
Neoplasias Cerebelosas/prevención & control , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Meduloblastoma/prevención & control , Receptores de Superficie Celular/fisiología , Animales , Western Blotting , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Receptor beta de Estrógeno/metabolismo , Femenino , Heterocigoto , Técnicas para Inmunoenzimas , Factor I del Crecimiento Similar a la Insulina/metabolismo , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Noqueados , Ovariectomía , Receptores Patched , Receptor Patched-1 , ARN Mensajero/genética , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Irradiación Corporal Total
14.
Science ; 326(5958): 1424-7, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19965762

RESUMEN

Granule neuron precursors (GNPs) are the most actively proliferating cells in the postnatal nervous system, and mutations in pathways that control the GNP cell cycle can result in medulloblastoma. The transcription factor Atoh1 has been suspected to contribute to GNP proliferation, but its role in normal and neoplastic postnatal cerebellar development remains unexplored. We show that Atoh1 regulates the signal transduction pathway of Sonic Hedgehog, an extracellular factor that is essential for GNP proliferation, and demonstrate that deletion of Atoh1 prevents cerebellar neoplasia in a mouse model of medulloblastoma. Our data shed light on the function of Atoh1 in postnatal cerebellar development and identify a new mechanism that can be targeted to regulate medulloblastoma formation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Neoplasias Cerebelosas/prevención & control , Cerebelo/metabolismo , Proteínas Hedgehog/metabolismo , Meduloblastoma/prevención & control , Neuronas/citología , Animales , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Neoplasias Cerebelosas/etiología , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Regulación hacia Abajo , Eliminación de Gen , Técnicas de Sustitución del Gen , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Meduloblastoma/etiología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal , Receptor Smoothened , Proteína Gli2 con Dedos de Zinc
15.
Proc Natl Acad Sci U S A ; 106(40): 17037-42, 2009 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-19805107

RESUMEN

Elevated expression of HSP90 is observed in many tumor types and is associated with a limited clinical response. Targeting HSP90 using inhibitors such as 17-DMAG (17-desmethoxy-17-N,N-dimethylaminoethylaminogeldanamycin) has shown limited therapeutic success. HSP90 regulates the function of several proteins implicated in tumorigenesis although the precise mechanism through which 17-DMAG regulates tumor cell survival remains unclear. We observed a requirement for p53 in mediating 17-DMAG-induced cell death. The sensitivity of primary mouse embryonic fibroblasts and tumor cells to 17-DMAG-induced apoptosis depended on the p53 status. Wild-type MEFs underwent 17-DMAG-induced caspase-dependent cell death, whilst those lacking p53 failed to do so. Interestingly p53-dependent cell death occurred independently of Atm or Arf. Primary tumor cells derived from two models of murine medulloblastoma (Ptch1(+/-);Ink4c(-/-) and p53(FL/FL);Nestin-Cre(+); Ink4c(-/-)) that retain and lack p53 function, respectively, displayed a dependence on functional p53 to engage 17-DMAG-induced apoptosis. Strikingly, 17-DMAG treatment in an allograft model of Ptch1(+/-);Ink4c(-/-) but not p53(FL/FL);Nestin-Cre(+); Ink4c(-/-) tumor cells prevented tumor growth in vivo. Our data suggest that p53 status is a likely predictor of the sensitivity of tumors to 17-DMAG.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/metabolismo , Lactamas Macrocíclicas/farmacología , Meduloblastoma/prevención & control , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Células Cultivadas , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Citometría de Flujo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Humanos , Immunoblotting , Inmunohistoquímica , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Receptores Patched , Receptor Patched-1 , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
16.
Mol Cancer Res ; 7(1): 33-40, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19147535

RESUMEN

Two cyclin-dependent kinase inhibitors, p18(Ink4c) and p27(Kip1), are required for proper cerebellar development. Loss of either of these proteins conferred a proliferative advantage to granule neuron progenitors, although inactivation of Kip1 exerted a greater effect. Mice heterozygous for Patched-1 (Ptc1+/-) that are either heterozygous or nullizygous for Kip1 developed medulloblastoma rapidly and with high penetrance. All tumors from Ptc1+/-;Kip1+/- or Ptc1+/-;Kip1-/- mice failed to express the wild-type Ptc1 allele, consistent with its role as a canonical "two-hit" tumor suppressor. In contrast, expression of the wild-type p27(Kip1) protein was invariably maintained in medulloblastomas arising in Ptc1+/-;Kip1+/- mice, indicating that Kip1 is haploinsufficient for tumor suppression. Although medulloblastomas occurring in Ptc1+/- mice were histopathologically heterogeneous and contained intermixed regions of both rapidly proliferating and nondividing more differentiated cells, tumors that also lacked Kip1 were uniformly less differentiated, more highly proliferative, and invasive. Molecular analysis showed that the latter medulloblastomas exhibited constitutive activation of the Sonic hedgehog signaling pathway without loss of functional p53. Apart from gains or losses of single chromosomes, with gain of chromosome 6 being the most frequent, no other chromosomal anomalies were identified by spectral karyotyping, and half of the medulloblastomas so examined retained a normal karyotype. In this respect, this mouse medulloblastoma model recapitulates the vast majority of human medulloblastomas that do not sustain TP53 mutations and are not aneuploid.


Asunto(s)
Neoplasias Cerebelosas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Meduloblastoma/genética , Receptores de Superficie Celular/genética , Animales , Ciclo Celular , Neoplasias Cerebelosas/prevención & control , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Aberraciones Cromosómicas , Cromosomas Humanos Par 6 , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Tamización de Portadores Genéticos , Genotipo , Humanos , Cariotipificación , Meduloblastoma/prevención & control , Ratones , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/deficiencia
17.
J Cell Biol ; 183(3): 385-91, 2008 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-18955550

RESUMEN

The Sonic Hedgehog (Shh) pathway plays important roles in embryogenesis, stem cell maintenance, tissue repair, and tumorigenesis. Haploinsufficiency of Patched-1, a gene that encodes a repressor of the Shh pathway, dysregulates the Shh pathway and increases genomic instability and the development of spontaneous and ionizing radiation (IR)-induced tumors by an unknown mechanism. Here we show that Ptc1(+/-) mice have a defect in the IR-induced activation of the ATR-Chk1 checkpoint signaling pathway. Likewise, transient expression of Gli1, a downstream target of Shh signaling, disrupts Chk1 activation in human cells by preventing the interaction of Chk1 with Claspin, a Chk1 adaptor protein that is required for Chk1 activation. These results suggest that inappropriate Shh pathway activation promotes tumorigenesis by disabling a key signaling pathway that helps maintain genomic stability and inhibits tumorigenesis.


Asunto(s)
Inestabilidad Genómica/genética , Proteínas Hedgehog/fisiología , Radiación Ionizante , Receptores de Superficie Celular/genética , Animales , Neoplasias Cerebelosas/epidemiología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/prevención & control , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Daño del ADN/genética , Daño del ADN/efectos de la radiación , ADN de Cadena Simple/genética , ADN de Cadena Simple/efectos de la radiación , Inestabilidad Genómica/fisiología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Meduloblastoma/epidemiología , Meduloblastoma/genética , Meduloblastoma/prevención & control , Ratones , Ratones Noqueados , Neoplasias/genética , Receptores Patched , Receptor Patched-1 , Proteínas Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Superficie Celular/deficiencia , Proteína con Dedos de Zinc GLI1
18.
Carcinogenesis ; 29(10): 1911-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18660545

RESUMEN

The patched (Ptc1) protein is a negative regulator of sonic hedgehog signaling, a genetic pathway whose perturbation causes developmental defects and predisposition to specific malignant tumors. Humans and mice with mutated Ptc1 are prone to medulloblastoma and basal cell carcinoma (BCC), both tumors showing dependence on radiation damage for rapid onset and high penetrance. Poly(ADP-ribose) polymerase (PARP-1) is a nuclear enzyme that plays a multifunctional role in DNA damage signaling and repair. In healthy and fertile PARP-1-null mice, radiation exposure reveals an extreme sensitivity and a high genomic instability. To test for interactions between PARP-1 and sonic hedgehog signaling, PARP-1-null mice were crossed to Ptc1 heterozygous mice. PARP-1 deletion further accelerated medulloblastoma development in irradiated Ptc1(+/-) mice, showing that PARP-1 inactivation sensitizes cerebellar cells to radiation tumorigenic effects. In addition to increased formation and slowed down kinetics of disappearance of gamma-H2AX foci, we observed increased apoptosis in PARP-1-deficient granule cell progenitors after irradiation. Double-mutant mice were also strikingly more susceptible to BCC, with >50% of animals developing multiple, large, infiltrative tumors within 30 weeks of age. The results provide genetic evidence that PARP-1 function suppresses sonic hedgehog pathway-associated tumors arising in response to environmental stress.


Asunto(s)
Carcinoma Basocelular/prevención & control , Meduloblastoma/prevención & control , Poli(ADP-Ribosa) Polimerasas/fisiología , Receptores de Superficie Celular/fisiología , Animales , Apoptosis , Carcinoma Basocelular/etiología , Carcinoma Basocelular/patología , Cerebelo/patología , Cerebelo/efectos de la radiación , Daño del ADN , Inestabilidad Genómica , Histonas/análisis , Meduloblastoma/etiología , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Lesiones Precancerosas/etiología , Receptores de Superficie Celular/genética
19.
Genes Dev ; 22(6): 722-7, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18347090

RESUMEN

Bone morphogenic proteins 2 and 4 (BMP2 and BMP4) inhibit proliferation and induce differentiation of cerebellar granule neuron progenitors (GNPs) and primary GNP-like medulloblastoma (MB) cells. This occurs through rapid proteasome-mediated degradation of Math1 (Atoh1), a transcription factor expressed in proliferating GNPs. Ectopic expression of Atoh1, but not of Sonic hedgehog (Shh)-regulated Gli1 or Mycn, cancels these BMP-mediated effects and restores Shh-dependent proliferation of GNPs and MB cells in vitro and in vivo. Genes regulating the BMP signaling pathway are down-regulated in mouse MBs. Thus, BMPs are potent inhibitors of MB and should be considered as novel therapeutic agents.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Neoplasias Cerebelosas/prevención & control , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/prevención & control , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Neoplasias Cerebelosas/metabolismo , Regulación hacia Abajo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Técnicas para Inmunoenzimas , Factores de Transcripción de Tipo Kruppel/metabolismo , Meduloblastoma/metabolismo , Ratones , Proteína Proto-Oncogénica N-Myc , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína con Dedos de Zinc GLI1
20.
FASEB J ; 21(9): 2215-25, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17371797

RESUMEN

Medulloblastoma, the most common brain tumor in childhood, appears to originate from cerebellar granule cell precursors (GCPs), located in the external granular layer (EGL) of the cerebellum. The antiproliferative gene PC3 (Tis21/BTG2) promotes cerebellar neurogenesis by inducing GCPs to shift from proliferation to differentiation. To assess whether PC3 can prevent the neoplastic transformation of GCPs and medulloblastoma development, we crossed transgenic mice conditionally expressing PC3 (TgPC3) in GCPs with Patched1 heterozygous mice (Ptc(+/-)), a model of medulloblastoma pathogenesis characterized by hyperactivation of the Sonic Hedgehog pathway. Perinatal up-regulation of PC3 in Ptc(+/-)/TgPC3 mice results in a decrease of medulloblastoma incidence of approximately 40% and in a marked reduction of preneoplastic abnormalities, such as hyperplastic EGL areas and lesions. Moreover, overexpression of cyclin D1, hyperproliferation, and defective differentiation--observed in Ptc(+/-) GCPs--are restored to normality in Ptc(+/-)/TgPC3 mice. The PC3-mediated inhibition of cyclin D1 expression correlates with recruitment of PC3 to the cyclin D1 promoter, which is accompanied by histone deacetylation. Remarkably, down-regulation of PC3 is observed in preneoplastic lesions, as well as in human and murine medulloblastomas. As a whole, this indicates that PC3 may prevent medulloblastoma development by controlling cell cycle and promoting differentiation of GCPs.


Asunto(s)
Neoplasias Cerebelosas/prevención & control , Genes Supresores de Tumor , Proteínas Inmediatas-Precoces/fisiología , Meduloblastoma/prevención & control , Acetilación , Animales , Síndrome del Nevo Basocelular/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , División Celular/genética , División Celular/fisiología , Transformación Celular Neoplásica/genética , Corteza Cerebelosa/anomalías , Corteza Cerebelosa/embriología , Neoplasias Cerebelosas/genética , Coristoma/genética , Inmunoprecipitación de Cromatina , Ciclina D , Ciclinas/biosíntesis , Ciclinas/genética , Proteínas Hedgehog/fisiología , Heterocigoto , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Meduloblastoma/genética , Ratones , Ratones Transgénicos , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/prevención & control , Neuronas/patología , Células PC12/química , Receptores Patched , Receptor Patched-1 , Lesiones Precancerosas/genética , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , ARN Neoplásico/genética , Ratas , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA