Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Neurooncol ; 126(2): 225-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26518542

RESUMEN

Chemotherapies active in preclinical studies frequently fail in the clinic due to lack of efficacy, which limits progress for rare cancers since only small numbers of patients are available for clinical trials. Thus, a preclinical drug development pipeline was developed to prioritize potentially active regimens for pediatric brain tumors spanning from in vitro drug screening, through intracranial and intra-tumoral pharmacokinetics to in vivo efficacy studies. Here, as an example of the pipeline, data are presented for the combination of 5-fluoro-2'-deoxycytidine and tetrahydrouridine in three pediatric brain tumor models. The in vitro activity of nine novel therapies was tested against tumor spheres derived from faithful mouse models of Group 3 medulloblastoma, ependymoma, and choroid plexus carcinoma. Agents with the greatest in vitro potency were then subjected to a comprehensive series of in vivo pharmacokinetic (PK) and pharmacodynamic (PD) studies culminating in preclinical efficacy trials in mice harboring brain tumors. The nucleoside analog 5-fluoro-2'-deoxycytidine (FdCyd) markedly reduced the proliferation in vitro of all three brain tumor cell types at nanomolar concentrations. Detailed intracranial PK studies confirmed that systemically administered FdCyd exceeded concentrations in brain tumors necessary to inhibit tumor cell proliferation, but no tumor displayed a significant in vivo therapeutic response. Despite promising in vitro activity and in vivo PK properties, FdCyd is unlikely to be an effective treatment of pediatric brain tumors, and therefore was deprioritized for the clinic. Our comprehensive and integrated preclinical drug development pipeline should reduce the attrition of drugs in clinical trials.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Desoxicitidina/análogos & derivados , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Tetrahidrouridina/administración & dosificación , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/sangre , Desoxicitidina/farmacocinética , Desoxicitidina/uso terapéutico , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Ratones , Ratones Desnudos , Tetrahidrouridina/sangre , Tetrahidrouridina/farmacocinética , Tetrahidrouridina/uso terapéutico
2.
Cereb Cortex ; 23(7): 1742-55, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22700645

RESUMEN

During the pre- and neonatal period, the cerebral cortex reveals distinct patterns of spontaneous synchronized activity, which is critically involved in the formation of early networks and in the regulation of neuronal survival and programmed cell death (apoptosis). During this period, the cortex is also highly vulnerable to inflammation and in humans prenatal infection may have a profound impact on neurodevelopment causing long-term neurological deficits. Using in vitro and in vivo multi-electrode array recordings and quantification of caspase-3 (casp-3)-dependent apoptosis, we demonstrate that lipopolysaccharide-induced inflammation causes rapid alterations in the pattern of spontaneous burst activities, which subsequently leads to an increase in apoptosis. We show that these inflammatory effects are specifically initiated by the microglia-derived pro-inflammatory cytokine tumor necrosis factor α and the chemokine macrophage inflammatory protein 2. Our data demonstrate that inflammation-induced modifications in spontaneous network activities influence casp-3-dependent cell death in the developing cerebral cortex.


Asunto(s)
Apoptosis/fisiología , Corteza Cerebral/fisiopatología , Inflamación/fisiopatología , Microglía/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Animales Recién Nacidos , Western Blotting , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Electrofisiología , Inflamación/inducido químicamente , Inflamación/patología , Lipopolisacáridos/toxicidad , Neuronas/patología , Ratas , Ratas Wistar
3.
Cereb Cortex ; 21(5): 1192-202, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20966045

RESUMEN

Electrical activity and sufficient supply with survival factors play a major role in the control of apoptosis in the developing cortex. Coherent high-frequency neuronal activity, which efficiently releases neurotrophins, is essential for the survival of immature neurons. We studied the influence of neuronal activity on apoptosis in the developing cortex. Dissociated cultures of the newborn mouse cerebral cortex were grown on multielectrode arrays to determine the activity patterns that promote neuronal survival. Cultures were transfected with a plasmid coding for a caspase-3-sensitive fluorescent protein allowing real-time analysis of caspase-3-dependent apoptosis in individual neurons. Elevated extracellular potassium concentrations (5 and 8 mM), application of 4-aminopyridine or the γ-aminobutyric acid-A receptor antagonist Gabazine induced a shift in the frequency distribution of activity toward high-frequency bursts. Under these conditions, a reduction or delay in caspase-3 activation and an overall increase in neuronal survival could be observed. This effect was dependent on the activity of phosphatidylinositol-3 kinase, as blockade of this enzyme abolished the survival-promoting effect of high extracellular potassium concentrations. Our data indicate that increased network activity can prevent apoptosis in developing cortical neurons.


Asunto(s)
Potenciales de Acción/fisiología , Apoptosis/fisiología , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Neuronas/citología , Neuronas/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/citología , Red Nerviosa/efectos de los fármacos , Red Nerviosa/crecimiento & desarrollo , Neuronas/efectos de los fármacos , Piridazinas/farmacología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
4.
Autophagy ; 14(7): 1256-1266, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29999454

RESUMEN

Macroautophagy/autophagy is an evolutionarily conserved catabolic pathway whose modulation has been linked to diverse disease states, including age-associated disorders. Conventional and conditional whole-body knockout mouse models of key autophagy genes display perinatal death and lethal neurotoxicity, respectively, limiting their applications for in vivo studies. Here, we have developed an inducible shRNA mouse model targeting Atg5, allowing us to dynamically inhibit autophagy in vivo, termed ATG5i mice. The lack of brain-associated shRNA expression in this model circumvents the lethal phenotypes associated with complete autophagy knockouts. We show that ATG5i mice recapitulate many of the previously described phenotypes of tissue-specific knockouts. While restoration of autophagy in the liver rescues hepatomegaly and other pathologies associated with autophagy deficiency, this coincides with the development of hepatic fibrosis. These results highlight the need to consider the potential side effects of systemic anti-autophagy therapies.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/metabolismo , Autofagia , ARN Interferente Pequeño/metabolismo , Animales , Animales Recién Nacidos , Proteína 5 Relacionada con la Autofagia/genética , Regulación hacia Abajo/genética , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Modelos Animales , Fenotipo , Factores de Tiempo
5.
Clin Cancer Res ; 24(7): 1654-1666, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29301833

RESUMEN

Purpose: Curing all children with brain tumors will require an understanding of how each subtype responds to conventional treatments and how best to combine existing and novel therapies. It is extremely challenging to acquire this knowledge in the clinic alone, especially among patients with rare tumors. Therefore, we developed a preclinical brain tumor platform to test combinations of conventional and novel therapies in a manner that closely recapitulates clinic trials.Experimental Design: A multidisciplinary team was established to design and conduct neurosurgical, fractionated radiotherapy and chemotherapy studies, alone or in combination, in accurate mouse models of supratentorial ependymoma (SEP) subtypes and choroid plexus carcinoma (CPC). Extensive drug repurposing screens, pharmacokinetic, pharmacodynamic, and efficacy studies were used to triage active compounds for combination preclinical trials with "standard-of-care" surgery and radiotherapy.Results: Mouse models displayed distinct patterns of response to surgery, irradiation, and chemotherapy that varied with tumor subtype. Repurposing screens identified 3-hour infusions of gemcitabine as a relatively nontoxic and efficacious treatment of SEP and CPC. Combination neurosurgery, fractionated irradiation, and gemcitabine proved significantly more effective than surgery and irradiation alone, curing one half of all animals with aggressive forms of SEP.Conclusions: We report a comprehensive preclinical trial platform to assess the therapeutic activity of conventional and novel treatments among rare brain tumor subtypes. It also enables the development of complex, combination treatment regimens that should deliver optimal trial designs for clinical testing. Postirradiation gemcitabine infusion should be tested as new treatments of SEP and CPC. Clin Cancer Res; 24(7); 1654-66. ©2018 AACR.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ratones , Ratones Desnudos , Resultado del Tratamiento , Gemcitabina
6.
Cancer Cell ; 27(5): 712-27, 2015 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-25965574

RESUMEN

Choroid plexus carcinomas (CPCs) are poorly understood and frequently lethal brain tumors with few treatment options. Using a mouse model of the disease and a large cohort of human CPCs, we performed a cross-species, genome-wide search for oncogenes within syntenic regions of chromosome gain. TAF12, NFYC, and RAD54L co-located on human chromosome 1p32-35.3 and mouse chromosome 4qD1-D3 were identified as oncogenes that are gained in tumors in both species and required for disease initiation and progression. TAF12 and NFYC are transcription factors that regulate the epigenome, whereas RAD54L plays a central role in DNA repair. Our data identify a group of concurrently gained oncogenes that cooperate in the formation of CPC and reveal potential avenues for therapy.


Asunto(s)
Factor de Unión a CCAAT/genética , Carcinoma/genética , Neoplasias del Plexo Coroideo/genética , ADN Helicasas/genética , Genómica , Proteínas Nucleares/genética , Oncogenes , Factores Asociados con la Proteína de Unión a TATA/genética , Animales , Secuencia de Bases , Carcinoma/patología , Proliferación Celular/genética , Neoplasias del Plexo Coroideo/patología , Mapeo Cromosómico , Cartilla de ADN , Proteínas de Unión al ADN , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie
7.
Nat Genet ; 47(8): 878-87, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26075792

RESUMEN

Cancers are characterized by non-random chromosome copy number alterations that presumably contain oncogenes and tumor-suppressor genes (TSGs). The affected loci are often large, making it difficult to pinpoint which genes are driving the cancer. Here we report a cross-species in vivo screen of 84 candidate oncogenes and 39 candidate TSGs, located within 28 recurrent chromosomal alterations in ependymoma. Through a series of mouse models, we validate eight new ependymoma oncogenes and ten new ependymoma TSGs that converge on a small number of cell functions, including vesicle trafficking, DNA modification and cholesterol biosynthesis, identifying these as potential new therapeutic targets.


Asunto(s)
Ependimoma/genética , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad/genética , Oncogenes/genética , Animales , Células Cultivadas , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN , Ependimoma/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones Desnudos , Ratones Transgénicos , Microscopía Confocal , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
8.
Neuroreport ; 22(10): 509-13, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-21666516

RESUMEN

To investigate whether glycine receptors influence radial migration in the neocortex, we analyzed the effect of glycine and the glycinergic antagonist strychnine, on the distribution of 5-bromo-2'deoxyuridine-labeled neurons in organotypic slice cultures from embryonic mice cortices. Application of glycine impeded radial migration only in the presence of the glycine-transport blockers, ALX-5407 and ALX-1393. This effect was blocked by the specific glycine receptor antagonist strychnine, whereas application of strychnine in the absence of glycine was without effect. We conclude from these observations that an activation of glycine receptors can impede radial migration, but that the glycinergic system is not directly implicated in the regulation of radial migration in organotypic slice cultures.


Asunto(s)
Movimiento Celular/fisiología , Neocórtex/citología , Neuronas/fisiología , Receptores de Glicina/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Recuento de Células , Movimiento Celular/efectos de los fármacos , Interacciones Farmacológicas , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicina/farmacología , Glicinérgicos/farmacología , Ratones , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fosfopiruvato Hidratasa/metabolismo , Sarcosina/análogos & derivados , Sarcosina/farmacología , Estricnina/farmacología
9.
Brain Res Bull ; 80(6): 428-32, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19682559

RESUMEN

Cyclic nucleotides mediate transient as well as plastic cellular responses. The most ultimate response is cell death. In the present study, we propose that an increase of intracellular cyclic guanosine monophosphate (cGMP) for at least 1h promotes cell death in the murine microglial cell line, BV-2 cells, as well as in primary murine microglia. Cells were exposed to ammonium, the guanylyl cyclase inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), and to the membrane-permeable cGMP analogue, 8-Bromo-cGMP (8-Br-cGMP), respectively. Cell death was estimated using DAPI labelling and annexin-V labelling of exposed phosphatidylserine, and cGMP level was quantified by an immunoassay. Ammonium not only increased the number of apoptotic cells but also promoted a moderate increase in intracellular cGMP. Addition of ODQ suppressed ammonium-induced apoptosis. Furthermore, we found that 8-Br-cGMP significantly increased the number of BV-2 cells and primary microglia, respectively, containing nuclei with condensed chromatin accumulated at the nuclear periphery. Similarly, cells exposed to 8-Br-cGMP showed significantly more cells with exposed phosphatidylserine compared to control cells. Thus, according to the nuclear structure as well as to changes in the plasma membrane, chronic elevation of cGMP induces apoptosis in microglia.


Asunto(s)
Apoptosis/fisiología , GMP Cíclico/metabolismo , Microglía/fisiología , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/fisiología , Células Cultivadas , Cromatina/efectos de los fármacos , Cromatina/fisiología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Inhibidores Enzimáticos/farmacología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Nitritos/metabolismo , Oxadiazoles/farmacología , Fosfatidilserinas/metabolismo , Compuestos de Amonio Cuaternario/toxicidad , Quinoxalinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA