Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 14(4): 296, 2023 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-37120445

RESUMEN

The diffuse nature of Glioblastoma (GBM) tumors poses a challenge to current therapeutic options. We have previously shown that Acyl-CoA Binding Protein (ACBP, also known as DBI) regulates lipid metabolism in GBM cells, favoring fatty acid oxidation (FAO). Here we show that ACBP downregulation results in wide transcriptional changes affecting invasion-related genes. In vivo experiments using patient-derived xenografts combined with in vitro models demonstrated that ACBP sustains GBM invasion via binding to fatty acyl-CoAs. Blocking FAO mimics ACBPKD-induced immobility, a cellular phenotype that can be rescued by increasing FAO rates. Further investigation into ACBP-downstream pathways served to identify Integrin beta-1, a gene downregulated upon inhibition of either ACBP expression or FAO rates, as a mediator for ACBP's role in GBM invasion. Altogether, our findings highlight a role for FAO in GBM invasion and reveal ACBP as a therapeutic vulnerability to stall FAO and subsequent cell invasion in GBM tumors.


Asunto(s)
Proteínas Portadoras , Glioblastoma , Humanos , Proteínas Portadoras/metabolismo , Glioblastoma/genética , Inhibidor de la Unión a Diazepam/metabolismo , Metabolismo de los Lípidos , Ácidos Grasos/metabolismo
2.
Cell Rep ; 33(2): 108256, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33053356

RESUMEN

Angiogenesis and neurogenesis are tightly coupled during embryonic brain development. However, little is known about how these two processes interact. We show that nascent blood vessels actively contact dividing neural stem cells by endothelial filopodia in the ventricular zone (VZ) of the murine ventral telencephalon; this association is conserved in the human ventral VZ. Using mouse mutants with altered vascular filopodia density, we show that this interaction leads to prolonged cell cycle of apical neural progenitors (ANPs) and favors early neuronal differentiation. Interestingly, pharmacological experiments reveal that ANPs induce vascular filopodia formation by upregulating vascular endothelial growth factor (VEGF)-A in a cell-cycle-dependent manner. This mutual relationship between vascular filopodia and ANPs works as a self-regulatory system that senses ANP proliferation rates and rapidly adjusts neuronal production levels. Our findings indicate a function of vascular filopodia in fine-tuning neural stem cell behavior, which is the basis for proper brain development.


Asunto(s)
Células-Madre Neurales/metabolismo , Neurogénesis , Seudópodos/metabolismo , Telencéfalo/irrigación sanguínea , Animales , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Endotelio Vascular/metabolismo , Humanos , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Neuronas/citología , Seudópodos/ultraestructura , Telencéfalo/ultraestructura , Imagen de Lapso de Tiempo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Cell Metab ; 30(2): 274-289.e5, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31056285

RESUMEN

Glioblastoma multiforme (GBM) undergoes metabolic reprogramming to meet the high ATP and anabolic demands of the tumor cells. However, the role of fatty acid oxidation (FAO) and its regulators in the GBM context has been largely unknown. Here, we show that the neural stem cell pro-proliferative factor acyl-CoA-binding protein (ACBP, also known as DBI) is highly expressed in GBM, and by binding to acyl-CoAs, it cell-autonomously maintains high proliferation rates, promoting tumor growth and poor survival in several preclinical models. Mechanistic experiments using ACBP-acyl-CoA binding affinity variants and pharmacological FAO modulators suggest that ACBP supports tumor growth by controlling the availability of long-chain fatty acyl-CoAs to mitochondria, promoting FAO in GBM. Thus, our findings uncover a critical link between lipid metabolism and GBM progression established by ACBP and offer a potential therapeutic strategy for an effective anti-proliferative metabolic management of GBM.


Asunto(s)
Inhibidor de la Unión a Diazepam/metabolismo , Ácidos Grasos/metabolismo , Glioblastoma/metabolismo , Animales , Células Cultivadas , Femenino , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Oxidación-Reducción
4.
J Neurosci ; 35(40): 13659-72, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26446219

RESUMEN

Neuroblast migration is a highly orchestrated process that ensures the proper integration of newborn neurons into complex neuronal circuits. In the postnatal rodent brain, neuroblasts migrate long distances from the subependymal zone of the lateral ventricles to the olfactory bulb (OB) within the rostral migratory stream (RMS). They first migrate tangentially in close contact to each other and later radially as single cells until they reach their final destination in the OB. Sphingosine 1-phosphate (S1P) is a bioactive lipid that interacts with cell-surface receptors to exert different cellular responses. Although well studied in other systems and a target for the treatment of multiple sclerosis, little is known about S1P in the postnatal brain. Here, we report that the S1P receptor 1 (S1P1) is expressed in neuroblasts migrating in the RMS. Using in vivo and in vitro gain- and loss-of-function approaches in both wild-type and transgenic mice, we found that the activation of S1P1 by its natural ligand S1P, acting as a paracrine signal, contributes to maintain neuroblasts attached to each other while they migrate in chains within the RMS. Once in the OB, neuroblasts cease to express S1P1, which results in cell detachment and initiation of radial migration, likely via downregulation of NCAM1 and ß1 integrin. Our results reveal a novel physiological function for S1P1 in the postnatal brain, directing the path followed by newborn neurons in the neurogenic niche. SIGNIFICANCE STATEMENT: The function of each neuron is highly determined by the position it occupies within a neuronal circuit. Frequently, newborn neurons must travel long distances from their birthplace to their predetermined final location and, to do so, they use different modes of migration. In this study, we identify the sphingosine 1-phosphate (S1P) receptor 1 (S1P1) as one of the key players that govern the switch from tangential to radial migration of postnatally generated neuroblasts in the olfactory bulb. Of interest is the evidence that the ligand, S1P, is provided by nearby astrocytes. Finally, we also propose adhesion molecules that act downstream of S1P1 and initiate the transition from tangential chain migration to individual radial migration outside of the stream.


Asunto(s)
Movimiento Celular/genética , Regulación hacia Abajo/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Bulbo Olfatorio/citología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Animales Recién Nacidos , Antígeno CD56/genética , Antígeno CD56/metabolismo , Caspasa 3/metabolismo , Proteínas de Dominio Doblecortina , Células HEK293 , Humanos , Técnicas In Vitro , Integrina beta1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuropéptidos/metabolismo , Técnicas de Cultivo de Órganos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ácidos Siálicos/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
5.
J Neurosci ; 34(25): 8630-45, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24948817

RESUMEN

Correlative evidence suggests that GABAergic signaling plays an important role in the regulation of activity-dependent hippocampal neurogenesis and emotional behavior in adult mice. However, whether these are causally linked at the molecular level remains elusive. Nuclear factor of activated T cell (NFAT) proteins are activity-dependent transcription factors that respond to environmental stimuli in different cell types, including hippocampal newborn neurons. Here, we identify NFATc4 as a key activity-dependent transcriptional regulator of GABA signaling in hippocampal progenitor cells via an unbiased high-throughput genome-wide study. Next, we demonstrate that GABAA receptor (GABAAR) signaling modulates hippocampal neurogenesis through NFATc4 activity, which in turn regulates GABRA2 and GABRA4 subunit expression via binding to specific promoter responsive elements, as assessed by ChIP and luciferase assays. Furthermore, we show that selective pharmacological enhancement of GABAAR activity promotes hippocampal neurogenesis via the calcineurin/NFATc4 axis. Importantly, the NFATc4-dependent increase in hippocampal neurogenesis after GABAAR stimulation is required for the suppression of the anxiety response in mice. Together, these data provide a novel molecular insight into the regulation of the anxiety response in mice, suggesting that the GABAAR/NFATc4 axis is a druggable target for the therapy of emotional disorders.


Asunto(s)
Ansiedad/metabolismo , Ansiedad/prevención & control , Factores de Transcripción NFATC/metabolismo , Neurogénesis/fisiología , Receptores de GABA-A/fisiología , Transducción de Señal/fisiología , Animales , Ansiedad/patología , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...