Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Cereb Cortex ; 28(2): 538-548, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27999124

RESUMEN

Neuronal survival and morphological maturation depends on the action of the transcription factor calcium responsive element binding protein (CREB), which regulates expression of several target genes in an activity-dependent manner. However, it remains largely unknown whether CREB-mediated transcription could play a role at early stages of neuronal differentiation, prior to the establishment of functional synaptic contacts. Here, we show that CREB is phosphorylated at very early stages of neuronal differentiation in vivo and in vitro, even in the absence of depolarizing agents. Using genetic tools, we also show that inhibition of CREB-signaling affects neuronal growth and survival in vitro without affecting cell proliferation and neurogenesis. Expression of A-CREB or M-CREB, 2 dominant-negative inhibitors of CREB, decreases cell survival and the complexity of neuronal arborization. Similar changes are observed in neurons treated with protein kinase A (PKA) and Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitors, which also show decreased levels of pCREBSer133. Notably, expression of CREB-FY, a Tyr134Phe CREB mutant with a lower Km for phosphorylation, partly rescues the effects of PKA and CaMKII inhibition. Our data indicate that CREB-mediated signaling play important roles at early stages of cortical neuron differentiation, prior to the establishment of fully functional synaptic contacts.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Neuronas/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Embarazo , Inhibidores de Proteínas Quinasas/farmacología
2.
Neurooncol Adv ; 2(1): vdaa106, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33134921

RESUMEN

BACKGROUND: Pediatric high-grade gliomas (pHGGs) are aggressive primary brain tumors with local invasive growth and poor clinical prognosis. Treatment of pHGGs is particularly challenging given the intrinsic resistance to chemotherapy, an absence of novel therapeutics, and the difficulty of drugs to reach the tumor beds. Accumulating evidence suggests that production of reactive oxygen species (ROS) and misfolded proteins, which typically leads to endoplasmic reticulum (ER) stress, is an essential mechanism in cancer cell survival. METHODS: Several cell viability assays were used in 6 patient-derived pHGG cultures to evaluate the effect of the natural compound obtusaquinone (OBT) on cytotoxicity. Orthotopic mouse models were used to determine OBT effects in vivo. Immunoblotting, immunostaining, flow cytometry, and biochemical assays were used to investigate the OBT mechanism of action. RESULTS: OBT significantly inhibited cell survival of patient-derived pHGG cells in culture. OBT inhibited tumor growth and extended survival in 2 different orthotopic xenograft models. Mechanistically, OBT induced ER stress through abnormal ROS accumulation. CONCLUSION: Our data demonstrate the utility and feasibility of OBT as a potential therapeutic option for improving the clinical treatment of pHGGs.

3.
Adv Biosyst ; 4(12): e1900312, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32519463

RESUMEN

Glioblastoma (GBM) is the most common primary malignant brain tumor and despite optimal treatment, long-term survival remains uncommon. GBM can be roughly divided into three different molecular subtypes, each varying in aggressiveness and treatment resistance. Recent evidence shows plasticity between these subtypes in which the proneural (PN) glioma stem-like cells (GSCs) undergo transition into the more aggressive mesenchymal (MES) subtype, leading to therapeutic resistance. Extracellular vesicles (EVs) are membranous structures secreted by nearly every cell and are shown to play a key role in GBM progression by acting as multifunctional signaling complexes. Here, it is shown that EVs derived from MES cells educate PN cells to increase stemness, invasiveness, cell proliferation, migration potential, aggressiveness, and therapeutic resistance by inducing mesenchymal transition through nuclear factor-κB/signal transducer and activator of transcription 3 signaling. The findings could potentially help explore new treatment strategies for GBM and indicate that EVs may also play a role in mesenchymal transition of different tumor types.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Resistencia a Antineoplásicos/fisiología , Transición Epitelial-Mesenquimal/fisiología , Vesículas Extracelulares/metabolismo , Glioblastoma/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Humanos , Ratones , FN-kappa B/metabolismo , Células Madre Neoplásicas , Factor de Transcripción STAT3/metabolismo , Células Tumorales Cultivadas
4.
J Natl Cancer Inst ; 111(3): 283-291, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30257000

RESUMEN

BACKGROUND: The olfactory ensheathing cells (OECs) migrate from the peripheral nervous system to the central nervous system (CNS), a critical process for the development of the olfactory system and axonal extension after injury in neural regeneration. Because of their ability to migrate to the injury site and anti-inflammatory properties, OECs were tested against different neurological pathologies, but were never studied in the context of cancer. Here, we evaluated OEC tropism to gliomas and their potential as a "Trojan horse" to deliver therapeutic transgenes through the nasal pathway, their natural route to CNS. METHODS: OECs were purified from the mouse olfactory bulb and engineered to express a fusion protein between cytosine deaminase and uracil phosphoribosyltransferase (CU), which convert the prodrug 5-fluorocytosine (5-FC) into cytotoxic metabolite 5-fluorouracil, leading to a bystander killing of tumor cells. These cells were injected into the nasal cavity of mice bearing glioblastoma tumors and OEC-mediated gene therapy was monitored by bioluminescence imaging and confirmed with survival and ex vivo histological analysis. All statistical tests were two-sided. RESULTS: OECs migrated from the nasal pathway to the primary glioma site, tracked infiltrative glioma stemlike cells, and delivered therapeutic transgene, leading to a slower tumor growth and increased mice survival. At day 28, bioluminescence imaging revealed that mice treated with a single injection of OEC-expressing CU and 5-FC had tumor-associated photons (mean [SD]) of 1.08E + 08 [9.7E + 07] vs 4.1E + 08 [2.3E + 08] for control group (P < .001), with a median survival of 41 days vs 34 days, respectively (ratio = 0.8293, 95% confidence interval = 0.4323 to 1.226, P < .001) (n = 9 mice per group). CONCLUSIONS: We show for the first time that autologous transplantation of OECs can target and deliver therapeutic transgenes to brain tumors upon intranasal delivery, the natural route of OECs to the CNS, which could be extended to other types of cancer.


Asunto(s)
Citosina Desaminasa/administración & dosificación , Fluorouracilo/metabolismo , Terapia Genética , Glioma/terapia , Bulbo Olfatorio/trasplante , Pentosiltransferasa/administración & dosificación , Transgenes , Administración Intranasal , Animales , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo , Femenino , Flucitosina/metabolismo , Glioma/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Bulbo Olfatorio/citología , Bulbo Olfatorio/fisiología , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Sci Rep ; 6: 26353, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27198044

RESUMEN

We developed a novel approach to assess tumor vascularity using recombinant Gaussia luciferase (rGluc) protein and bioluminescence imaging. Upon intravenous injection of rGluc followed by its substrate coelenterazine, non-invasive visualization of tumor vascularity by bioluminescence imaging was possible. We applied this method for longitudinal monitoring of tumor vascularity in response to the anti-angiogenic drug tivozanib. This simple and sensitive method could be extended to image blood vessels/vasculature in many different fields.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Imidazoles/administración & dosificación , Luciferasas/metabolismo , Sustancias Luminiscentes/administración & dosificación , Neoplasias/irrigación sanguínea , Compuestos de Fenilurea/administración & dosificación , Pirazinas/administración & dosificación , Quinolinas/administración & dosificación , Animales , Línea Celular Tumoral , Copépodos/enzimología , Humanos , Mediciones Luminiscentes , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes/administración & dosificación , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA