Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(4): 114005, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38551961

RESUMEN

The retina is exquisitely patterned, with neuronal somata positioned at regular intervals to completely sample the visual field. Here, we show that phosphatase and tensin homolog (Pten) controls starburst amacrine cell spacing by modulating vesicular trafficking of cell adhesion molecules and Wnt proteins. Single-cell transcriptomics and double-mutant analyses revealed that Pten and Down syndrome cell adhesion molecule Dscam) are co-expressed and function additively to pattern starburst amacrine cell mosaics. Mechanistically, Pten loss accelerates the endocytic trafficking of DSCAM, FAT3, and MEGF10 off the cell membrane and into endocytic vesicles in amacrine cells. Accordingly, the vesicular proteome, a molecular signature of the cell of origin, is enriched in exocytosis, vesicle-mediated transport, and receptor internalization proteins in Pten conditional knockout (PtencKO) retinas. Wnt signaling molecules are also enriched in PtencKO retinal vesicles, and the genetic or pharmacological disruption of Wnt signaling phenocopies amacrine cell patterning defects. Pten thus controls vesicular trafficking of cell adhesion and signaling molecules to establish retinal amacrine cell mosaics.


Asunto(s)
Células Amacrinas , Adhesión Celular , Endocitosis , Fosfohidrolasa PTEN , Retina , Vía de Señalización Wnt , Animales , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/genética , Retina/metabolismo , Ratones , Células Amacrinas/metabolismo , Ratones Noqueados , Transporte de Proteínas , Proteínas Wnt/metabolismo , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/genética
2.
Front Neurosci ; 16: 917071, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36061596

RESUMEN

Direct neuronal reprogramming, the process whereby a terminally differentiated cell is converted into an induced neuron without traversing a pluripotent state, has tremendous therapeutic potential for a host of neurodegenerative diseases. While there is strong evidence for astrocyte-to-neuron conversion in vitro, in vivo studies in the adult brain are less supportive or controversial. Here, we set out to enhance the efficacy of neuronal conversion of adult astrocytes in vivo by optimizing the neurogenic capacity of a driver transcription factor encoded by the proneural gene Ascl1. Specifically, we mutated six serine phospho-acceptor sites in Ascl1 to alanines (Ascl1 SA 6) to prevent phosphorylation by proline-directed serine/threonine kinases. Native Ascl1 or Ascl1 SA 6 were expressed in adult, murine cortical astrocytes under the control of a glial fibrillary acidic protein (GFAP) promoter using adeno-associated viruses (AAVs). When targeted to the cerebral cortex in vivo, mCherry+ cells transduced with AAV8-GFAP-Ascl1 SA 6-mCherry or AAV8-GFAP-Ascl1-mCherry expressed neuronal markers within 14 days post-transduction, with Ascl1 SA 6 promoting the formation of more mature dendritic arbors compared to Ascl1. However, mCherry expression disappeared by 2-months post-transduction of the AAV8-GFAP-mCherry control-vector. To circumvent reporter issues, AAV-GFAP-iCre (control) and AAV-GFAP-Ascl1 (or Ascl1 SA 6)-iCre constructs were generated and injected into the cerebral cortex of Rosa reporter mice. In all comparisons of AAV capsids (AAV5 and AAV8), GFAP promoters (long and short), and reporter mice (Rosa-zsGreen and Rosa-tdtomato), Ascl1 SA 6 transduced cells more frequently expressed early- (Dcx) and late- (NeuN) neuronal markers. Furthermore, Ascl1 SA 6 repressed the expression of astrocytic markers Sox9 and GFAP more efficiently than Ascl1. Finally, we co-transduced an AAV expressing ChR2-(H134R)-YFP, an optogenetic actuator. After channelrhodopsin photostimulation, we found that Ascl1 SA 6 co-transduced astrocytes exhibited a significantly faster decay of evoked potentials to baseline, a neuronal feature, when compared to iCre control cells. Taken together, our findings support an enhanced neuronal conversion efficiency of Ascl1 SA 6 vs. Ascl1, and position Ascl1 SA 6 as a critical transcription factor for future studies aimed at converting adult brain astrocytes to mature neurons to treat disease.

3.
Neuron ; 109(18): 2847-2863.e11, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34407390

RESUMEN

Asymmetric neuronal expansion is thought to drive evolutionary transitions between lissencephalic and gyrencephalic cerebral cortices. We report that Neurog2 and Ascl1 proneural genes together sustain neurogenic continuity and lissencephaly in rodent cortices. Using transgenic reporter mice and human cerebral organoids, we found that Neurog2 and Ascl1 expression defines a continuum of four lineage-biased neural progenitor cell (NPC) pools. Double+ NPCs, at the hierarchical apex, are least lineage restricted due to Neurog2-Ascl1 cross-repression and display unique features of multipotency (more open chromatin, complex gene regulatory network, G2 pausing). Strikingly, selectively eliminating double+ NPCs by crossing Neurog2-Ascl1 split-Cre mice with diphtheria toxin-dependent "deleter" strains locally disrupts Notch signaling, perturbs neurogenic symmetry, and triggers cortical folding. In support of our discovery that double+ NPCs are Notch-ligand-expressing "niche" cells that control neurogenic periodicity and cortical folding, NEUROG2, ASCL1, and HES1 transcript distribution is modular (adjacent high/low zones) in gyrencephalic macaque cortices, prefiguring future folds.


Asunto(s)
Diferenciación Celular/fisiología , Neocórtex/embriología , Neocórtex/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Neocórtex/citología , Embarazo , Imagen de Lapso de Tiempo/métodos
4.
Curr Alzheimer Res ; 17(5): 446-459, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32579500

RESUMEN

BACKGROUND: Amyloid Precursor Protein (APP)-Binding Protein 1 (APP-BP1) is a crucial regulator of many key signaling pathways and functions mainly as a scaffold protein to enhance molecular interactions and facilitate catalytic reactions. The interaction of APP-BP1 with Amyloid Precursor Protein (APP) plays a role in cell cycle transit control, which determines the mechanism behind the loss of cell cycle regulation in Alzheimer's Disease (AD). In contrast, neddylation, a posttranslational modification mediated by conjugation of ubiquitin-like protein neural precursor cell expressed developmentally downregulated protein 8 (NEDD8), is activated by a heterodimer composed of APP-BP1 and NEDD8-activating enzyme E1 catalytic subunit (Uba3). NEDD8 controls vital biological events, and along with APP-BP1, its levels are deregulated in AD. OBJECTIVE: The present study investigated the role of melatonin in regulating the APP-BP1 pathway under both physiological and pathological conditions to develop an understanding of the underlying mechanisms. METHODS: Therefore, human SH-SY5Y neuroblastoma cells were treated with various concentrations of Aß42 to induce neurotoxic conditions comparable to AD. RESULTS: The results are the first to demonstrate that melatonin prevents Aß42-induced enhancement of APP-BP1 protein expression and alteration in the cellular localization of NEDD8. Moreover, using MLN4924 (APP-BP1 pathway blocker), we also verified the components of the downstream effector cascade of the APP-BP1 pathway, including tau, APP-cleaving secretases, ß-catenin and p53. CONCLUSION: These findings indicate that melatonin regulates the interplay of molecular signaling associated with the APP-BP1 pathway and might preclude the pathogenic mechanisms occurring during disease development, thus providing a propitious therapeutic strategy for preventing AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Antioxidantes/farmacología , Melatonina/farmacología , Proteína NEDD8/metabolismo , Fragmentos de Péptidos/toxicidad , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/biosíntesis , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Neuroblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
5.
Ann N Y Acad Sci ; 1443(1): 75-96, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30756405

RESUMEN

Neurodegenerative diseases are typified by neuronal loss associated with progressive dysfunction and clinical presentation. Neurodegenerative diseases are characterized by the intra- and extracellular conglomeration of misfolded proteins that occur because of abnormal protein dynamics and genetic manipulations; these trigger processes of cell death in these disorders. The disrupted signaling mechanisms involved are oxidative stress-mediated mitochondrial and calcium signaling deregulation, alterations in immune and inflammatory signaling, disruption of autophagic integrity, proteostasis dysfunction, and anomalies in the insulin, Notch, and Wnt/ß-catenin signaling pathways. Herein, we accentuate some of the contemporary translational approaches made in characterizing the underlying mechanisms of neurodegeneration. Melatonin-induced cognitive enhancement and inhibition of oxidative signaling substantiates the efficacy of melatonin in combating neurodegenerative processes. Our review considers in detail the possible roles of melatonin in understanding the synergistic pathogenic mechanisms between aggregated proteins and in regulating, modulating, and preventing the altered signaling mechanisms discovered in cellular and animal models along with clinical evaluations pertaining to neurodegeneration. Furthermore, this review showcases the therapeutic potential of melatonin in preventing and treating neurodegenerative diseases with optimum prognosis.


Asunto(s)
Melatonina/fisiología , Enfermedades Neurodegenerativas/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis , Calcio/metabolismo , Homeostasis , Humanos , Insulina/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Proteostasis/fisiología
6.
J Pineal Res ; 64(4): e12470, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29352484

RESUMEN

Melatonin is involved in the physiological regulation of the ß-amyloid precursor protein (ßAPP)-cleaving secretases which are responsible for generation of the neurotoxic amyloid beta (Aß) peptide, one of the hallmarks of Alzheimer's disease (AD) pathology. In this study, we aimed to determine the underlying mechanisms of this regulation under pathological conditions. We establish that melatonin prevents Aß42 -induced downregulation of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) as well as upregulation of ß-site APP-cleaving enzyme 1 (BACE1) and presenilin 1 (PS1) in SH-SY5Y cell cultures. We also demonstrate that the intrinsic mechanisms of the observed effects occurred via regulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and glycogen synthase kinase (GSK)-3ß as melatonin reversed Aß42 -induced upregulation and nuclear translocation of NF-κBp65 as well as activation of GSK3ß via its receptor activation. Furthermore, specific blocking of the NF-κB and GSK3ß pathways partially abrogated the Aß42 -induced reduction in the BACE1 and PS1 levels. In addition, GSK3ß blockage affected α-secretase cleavage and modulated nuclear translocation of NF-κB. Importantly, our study for the first time shows that peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) is a crucial target of melatonin. The compromised levels and/or genetic variation of Pin1 are associated with age-dependent tau and Aß pathologies and neuronal degeneration. Interestingly, melatonin alleviated the Aß42 -induced reduction of nuclear Pin1 levels and preserved the functional integrity of this isomerase. Our findings illustrate that melatonin attenuates Aß42 -induced alterations of ßAPP-cleaving secretases possibly via the Pin1/GSK3ß/NF-κB pathway.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Melatonina/farmacología , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/efectos de los fármacos , Péptidos beta-Amiloides/efectos de los fármacos , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/efectos de los fármacos , Línea Celular , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , FN-kappa B/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Neuronas/metabolismo , Receptores de Melatonina/metabolismo
7.
Alzheimers Res Ther ; 7(1): 44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26045718

RESUMEN

In order to understand and find therapeutic strategies for neurological disorders, disease models that recapitulate the connectivity and circuitry of patients' brain are needed. Owing to many limitations of animal disease models, in vitro neuronal models using patient-derived stem cells are currently being developed. However, prior to employing neurons as a model in a dish, they need to be evaluated for their electrophysiological properties, including both passive and active membrane properties, dynamics of neurotransmitter release, and capacity to undergo synaptic plasticity. In this review, we survey recent attempts to study these issues in human induced pluripotent stem cell-derived neurons. Although progress has been made, there are still many hurdles to overcome before human induced pluripotent stem cell-derived neurons can fully recapitulate all of the above physiological properties of adult mature neurons. Moreover, proper integration of neurons into pre-existing circuitry still needs to be achieved. Nevertheless, in vitro neuronal stem cell-derived models hold great promise for clinical application in neurological diseases in the future.

8.
Am J Physiol Cell Physiol ; 308(3): C209-19, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25394470

RESUMEN

Production and isolation of forebrain interneuron progenitors are essential for understanding cortical development and developing cell-based therapies for developmental and neurodegenerative disorders. We demonstrate production of a population of putative calretinin-positive bipolar interneurons that express markers consistent with caudal ganglionic eminence identities. Using serum-free embryoid bodies (SFEBs) generated from human inducible pluripotent stem cells (iPSCs), we demonstrate that these interneuron progenitors exhibit morphological, immunocytochemical, and electrophysiological hallmarks of developing cortical interneurons. Finally, we develop a fluorescence-activated cell-sorting strategy to isolate interneuron progenitors from SFEBs to allow development of a purified population of these cells. Identification of this critical neuronal cell type within iPSC-derived SFEBs is an important and novel step in describing cortical development in this iPSC preparation.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/fisiología , Cuerpos Embrioides/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Interneuronas/fisiología , Animales , Células Cultivadas , Fibroblastos/fisiología , Humanos , Ratones , Ratones Noqueados
9.
PLoS One ; 9(7): e103418, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25072157

RESUMEN

Many protocols have been designed to differentiate human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs) into neurons. Despite the relevance of electrophysiological properties for proper neuronal function, little is known about the evolution over time of important neuronal electrophysiological parameters in iPSC-derived neurons. Yet, understanding the development of basic electrophysiological characteristics of iPSC-derived neurons is critical for evaluating their usefulness in basic and translational research. Therefore, we analyzed the basic electrophysiological parameters of forebrain neurons differentiated from human iPSCs, from day 31 to day 55 after the initiation of neuronal differentiation. We assayed the developmental progression of various properties, including resting membrane potential, action potential, sodium and potassium channel currents, somatic calcium transients and synaptic activity. During the maturation of iPSC-derived neurons, the resting membrane potential became more negative, the expression of voltage-gated sodium channels increased, the membrane became capable of generating action potentials following adequate depolarization and, at day 48-55, 50% of the cells were capable of firing action potentials in response to a prolonged depolarizing current step, of which 30% produced multiple action potentials. The percentage of cells exhibiting miniature excitatory post-synaptic currents increased over time with a significant increase in their frequency and amplitude. These changes were associated with an increase of Ca2+ transient frequency. Co-culturing iPSC-derived neurons with mouse glial cells enhanced the development of electrophysiological parameters as compared to pure iPSC-derived neuronal cultures. This study demonstrates the importance of properly evaluating the electrophysiological status of the newly generated neurons when using stem cell technology, as electrophysiological properties of iPSC-derived neurons mature over time.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Neuronas/fisiología , Animales , Calcio/metabolismo , Técnicas de Cocultivo , Fenómenos Electrofisiológicos , Humanos , Inmunofenotipificación , Ratones , Neuroglía , Técnicas de Placa-Clamp , Potenciales Sinápticos , Transmisión Sináptica , Factores de Tiempo
10.
Neurosci Lett ; 556: 20-5, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24120429

RESUMEN

The pathogenesis of Alzheimer's disease involves an amyloid ß-peptide (Aß)-induced cascade of elevated oxidative damage and inflammation. The present study investigates the protective effects and the underlying mechanisms of N-benzylcinnamide (PT-3), purified from Piper submultinerve. Against Aß-induced oxidative stress and inflammation in rat primary cortical cell cultures. Pre-treatment with 10-00nM PT-3 significantly attenuated neuronal cell death induced by 10µM Aß1-42. PT-3 was found to enhance cell viability through a significant reduction in the level of reactive oxygen species, down-regulated expression of pro-apoptotic activated caspase-3 and Bax, increased expression of anti-apoptotic Bcl-2, and mitigation of Aß-induced morphological alterations. Regarding its effects on inflammatory responses, PT-3 pre-treatment decreased the expression of pro-inflammatory cytokines IL-1ß and IL-6. The mechanisms of PT-3 neuronal protection against inflammation may be associated with the mitogen-activated protein kinases (MAPK) pathway. Aß1-42-induced phosphorylation of JNK and p38 MAPK was inhibited by pretreatment with PT-3 in a dose-dependent manner. However, phosphorylation of ERK1/2 was not affected by either PT-3 or Aß1-42. PT-3 did not stimulate Akt phosphorylation, which was inhibited by Aß1-42. These findings suggest that PT-3 protects neurons from Aß1-42-induced neurotoxicity through its anti-apoptotic, anti-oxidative, and anti-inflammatory properties with inhibition of JNK and p38 MAPK phosphorylation as the potential underlying mechanism.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Antiinflamatorios no Esteroideos/farmacología , Antioxidantes/farmacología , Cinamatos/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/citología , Fosforilación , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA