Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Intervalo de año de publicación
1.
EMBO J ; 41(2): e105531, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34904718

RESUMEN

Recessive gene mutations underlie many developmental disorders and often lead to disabling neurological problems. Here, we report identification of a homozygous c.170G>A (p.Cys57Tyr or C57Y) mutation in the gene coding for protein disulfide isomerase A3 (PDIA3, also known as ERp57), an enzyme that catalyzes formation of disulfide bonds in the endoplasmic reticulum, to be associated with syndromic intellectual disability. Experiments in zebrafish embryos show that PDIA3C57Y expression is pathogenic and causes developmental defects such as axonal disorganization as well as skeletal abnormalities. Expression of PDIA3C57Y in the mouse hippocampus results in impaired synaptic plasticity and memory consolidation. Proteomic and functional analyses reveal that PDIA3C57Y expression leads to dysregulation of cell adhesion and actin cytoskeleton dynamics, associated with altered integrin biogenesis and reduced neuritogenesis. Biochemical studies show that PDIA3C57Y has decreased catalytic activity and forms disulfide-crosslinked aggregates that abnormally interact with chaperones in the endoplasmic reticulum. Thus, rare disease gene variant can provide insight into how perturbations of neuronal proteostasis can affect the function of the nervous system.


Asunto(s)
Discapacidades del Desarrollo/genética , Retículo Endoplásmico/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteostasis , Adolescente , Adulto , Animales , Axones/metabolismo , Axones/patología , Adhesión Celular , Células Cultivadas , Niño , Citoesqueleto/metabolismo , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Integrinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación Missense , Proyección Neuronal , Plasticidad Neuronal , Linaje , Proteína Disulfuro Isomerasas/metabolismo , Pez Cebra
2.
J Neurosci ; 43(12): 2037-2052, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36948585

RESUMEN

Neuronal polarization is a complex molecular process regulated by intrinsic and extrinsic mechanisms. Nerve cells integrate multiple extracellular cues to generate intracellular messengers that ultimately control cell morphology, metabolism, and gene expression. Therefore, second messengers' local concentration and temporal regulation are crucial elements for acquiring a polarized morphology in neurons. This review article summarizes the main findings and current understanding of how Ca2+, IP3, cAMP, cGMP, and hydrogen peroxide control different aspects of neuronal polarization, and highlights questions that still need to be resolved to fully understand the fascinating cellular processes involved in axodendritic polarization.


Asunto(s)
Neuronas , Sistemas de Mensajero Secundario , Neuronas/fisiología , GMP Cíclico/metabolismo , Polaridad Celular/fisiología
3.
Mol Ther ; 31(7): 2240-2256, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37016577

RESUMEN

Alteration in the buffering capacity of the proteostasis network is an emerging feature of Alzheimer's disease (AD), highlighting the occurrence of endoplasmic reticulum (ER) stress. The unfolded protein response (UPR) is the main adaptive pathway to cope with protein folding stress at the ER. Inositol-requiring enzyme-1 (IRE1) operates as a central ER stress sensor, enabling the establishment of adaptive and repair programs through the control of the expression of the transcription factor X-box binding protein 1 (XBP1). To artificially enforce the adaptive capacity of the UPR in the AD brain, we developed strategies to express the active form of XBP1 in the brain. Overexpression of XBP1 in the nervous system using transgenic mice reduced the load of amyloid deposits and preserved synaptic and cognitive function. Moreover, local delivery of XBP1 into the hippocampus of an 5xFAD mice using adeno-associated vectors improved different AD features. XBP1 expression corrected a large proportion of the proteomic alterations observed in the AD model, restoring the levels of several synaptic proteins and factors involved in actin cytoskeleton regulation and axonal growth. Our results illustrate the therapeutic potential of targeting UPR-dependent gene expression programs as a strategy to ameliorate AD features and sustain synaptic function.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Estrés del Retículo Endoplásmico/genética , Ratones Transgénicos , Proteómica , Proteostasis/genética , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/genética
4.
Alzheimers Dement ; 20(2): 1298-1308, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37985413

RESUMEN

INTRODUCTION: Genome-wide association studies (GWAS) are fundamental for identifying loci associated with diseases. However, they require replication in other ethnicities. METHODS: We performed GWAS on sporadic Alzheimer's disease (AD) including 539 patients and 854 controls from Argentina and Chile. We combined our results with those from the European Alzheimer and Dementia Biobank (EADB) in a meta-analysis and tested their genetic risk score (GRS) performance in this admixed population. RESULTS: We detected apolipoprotein E ε4 as the single genome-wide significant signal (odds ratio  = 2.93 [2.37-3.63], P = 2.6 × 10-23 ). The meta-analysis with EADB summary statistics revealed four new loci reaching GWAS significance. Functional annotations of these loci implicated endosome/lysosomal function. Finally, the AD-GRS presented a similar performance in these populations, despite the score diminished when the Native American ancestry rose. DISCUSSION: We report the first GWAS on AD in a population from South America. It shows shared genetics modulating AD risk between the European and these admixed populations. HIGHLIGHTS: This is the first genome-wide association study on Alzheimer's disease (AD) in a population sample from Argentina and Chile. Trans-ethnic meta-analysis reveals four new loci involving lysosomal function in AD. This is the first independent replication for TREM2L, IGH-gene-cluster, and ADAM17 loci. A genetic risk score (GRS) developed in Europeans performed well in this population. The higher the Native American ancestry the lower the GRS values.


Asunto(s)
Enfermedad de Alzheimer , Azidas , Estudio de Asociación del Genoma Completo , Humanos , Chile , Enfermedad de Alzheimer/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética
5.
J Neurosci ; 41(8): 1636-1649, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33478991

RESUMEN

The acquisition of neuronal polarity is a complex molecular process that depends on changes in cytoskeletal dynamics and directed membrane traffic, regulated by the Rho and Rab families of small GTPases, respectively. However, during axon specification, a molecular link that couples these protein families has yet to be identified. In this paper, we describe a new positive feedback loop between Rab8a and Cdc42, coupled by Tuba, a Cdc42-specific guanine nucleotide-exchange factor (GEF), that ensures a single axon generation in rodent hippocampal neurons from embryos of either sex. Accordingly, Rab8a or Tuba gain-of-function generates neurons with supernumerary axons whereas Rab8a or Tuba loss-of-function abrogated axon specification, phenocopying the well-established effect of Cdc42 on neuronal polarity. Although Rab8 and Tuba do not interact physically, the activity of Rab8 is essential to generate a proximal to distal axonal gradient of Tuba in cultured neurons. Tuba-associated and Rab8a-associated polarity defects are also evidenced in vivo, since dominant negative (DN) Rab8a or Tuba knock-down impairs cortical neuronal migration in mice. Our results suggest that Tuba coordinates directed vesicular traffic and cytoskeleton dynamics during neuronal polarization.SIGNIFICANCE STATEMENT The morphologic, biochemical, and functional differences observed between axon and dendrites, require dramatic structural changes. The extension of an axon that is 1 µm in diameter and grows at rates of up to 500 µm/d, demands the confluence of two cellular processes: directed membrane traffic and fine-tuned cytoskeletal dynamics. In this study, we show that both processes are integrated in a positive feedback loop, mediated by the guanine nucleotide-exchange factor (GEF) Tuba. Tuba connects the activities of the Rab GTPase Rab8a and the Rho GTPase Cdc42, ensuring the generation of a single axon in cultured hippocampal neurons and controlling the migration of cortical neurons in the developing brain. Finally, we provide compelling evidence that Tuba is the GEF that mediates Cdc42 activation during the development of neuronal polarity.


Asunto(s)
Polaridad Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Neurogénesis/fisiología , Neuronas/citología , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Células COS , Movimiento Celular/fisiología , Chlorocebus aethiops , Retroalimentación Fisiológica/fisiología , Femenino , Hipocampo/embriología , Masculino , Ratones , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley
6.
J Cell Sci ; 133(3)2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31932508

RESUMEN

Neurons are highly complex cells that heavily rely on intracellular transport to distribute a range of functionally essential cargoes within the cell. Post-translational modifications of tubulin are emerging as mechanisms for regulating microtubule functions, but their impact on neuronal transport is only marginally understood. Here, we have systematically studied the impact of post-translational polyglutamylation on axonal transport. In cultured hippocampal neurons, deletion of a single deglutamylase, CCP1 (also known as AGTPBP1), is sufficient to induce abnormal accumulation of polyglutamylation, i.e. hyperglutamylation. We next investigated how hyperglutamylation affects axonal transport of a range of functionally different neuronal cargoes: mitochondria, lysosomes, LAMP1 endosomes and BDNF vesicles. Strikingly, we found a reduced motility for all these cargoes, suggesting that polyglutamylation could act as a regulator of cargo transport in neurons. This, together with the recent discovery that hyperglutamylation induces neurodegeneration, makes it likely that perturbed neuronal trafficking could be one of the central molecular causes underlying this novel type of degeneration.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Neuronas , Tubulina (Proteína) , Transporte Axonal , Hipocampo/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo
7.
J Neurochem ; 158(3): 586-588, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33909918

RESUMEN

In Alzheimer's disease (AD), hippocampal hyperactivation is already present at early stages of the disorder, in some cases, even when the individual is still asymptomatic. Neuronal hyperexcitability has been described to occur before the deposition of amyloid beta plaques in mouse models of AD and has been attributed to an imbalance between excitatory and inhibitory activity. In this Editorial Highlight, we discuss the article by Sosulina et al., published in this issue of the Journal of Neurochemistry, which offers novel insights into the possible origins of this neuronal excitability observed during the early pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Hipocampo/patología , Humanos , Neuronas/patología , Ratas , Ratas Transgénicas
8.
J Neurochem ; 158(3): 673-693, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34107066

RESUMEN

Dendritic spines are small, actin-rich protrusions that act as the receiving sites of most excitatory inputs in the central nervous system. The remodeling of the synapse architecture is mediated by actin cytoskeleton dynamics, a process precisely regulated by the small Rho GTPase family. Wnt ligands exert their presynaptic and postsynaptic effects during formation and consolidation of the synaptic structure. Specifically, Wnt5a has been identified as an indispensable synaptogenic factor for the regulation and organization of the postsynaptic side; however, the molecular mechanisms through which Wnt5a induces morphological changes resulting from actin cytoskeleton dynamics within dendritic spines remain unclear. In this work, we employ primary rat hippocampal cultures and HT22 murine hippocampal neuronal cell models, molecular and pharmacological tools, and fluorescence microscopy (laser confocal and epifluorescence) to define the Wnt5a-induced molecular signaling involved in postsynaptic remodeling mediated via the regulation of the small Rho GTPase family. We report that Wnt5a differentially regulates the phosphorylation of Cofilin in neurons through both Ras-related C3 botulinum toxin substrate 1 and cell division cycle 42 depending on the subcellular compartment and the extracellular calcium levels. Additionally, we demonstrate that Wnt5a increases the density of dendritic spines and promotes their maturation via Ras-related C3 botulinum toxin substrate 1. Accordingly, we find that Wnt5a requires the combined activation of small Rho GTPases to increase the levels of filamentous actin, thus promoting the stability of actin filaments. Altogether, these results provide evidence for a new mechanism by which Wnt5a may target actin dynamics, thereby regulating the subsequent morphological changes in dendritic spine architecture.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Proteína Wnt-5a/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Factores Despolimerizantes de la Actina/análisis , Animales , Línea Celular , Células Cultivadas , Espinas Dendríticas/química , Activación Enzimática/fisiología , Femenino , Hipocampo/química , Hipocampo/citología , Neuronas/química , Embarazo , Ratas , Ratas Sprague-Dawley , Proteína Wnt-5a/análisis , Proteínas de Unión al GTP rho/análisis
9.
Alzheimers Dement ; 17(2): 295-313, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33634602

RESUMEN

Across Latin American and Caribbean countries (LACs), the fight against dementia faces pressing challenges, such as heterogeneity, diversity, political instability, and socioeconomic disparities. These can be addressed more effectively in a collaborative setting that fosters open exchange of knowledge. In this work, the Latin American and Caribbean Consortium on Dementia (LAC-CD) proposes an agenda for integration to deliver a Knowledge to Action Framework (KtAF). First, we summarize evidence-based strategies (epidemiology, genetics, biomarkers, clinical trials, nonpharmacological interventions, networking, and translational research) and align them to current global strategies to translate regional knowledge into transformative actions. Then we characterize key sources of complexity (genetic isolates, admixture in populations, environmental factors, and barriers to effective interventions), map them to the above challenges, and provide the basic mosaics of knowledge toward a KtAF. Finally, we describe strategies supporting the knowledge creation stage that underpins the translational impact of KtAF.


Asunto(s)
Demencia/terapia , Práctica Clínica Basada en la Evidencia , Biomarcadores , Demencia/epidemiología , Humanos , América Latina/epidemiología , Factores Socioeconómicos
10.
Semin Cell Dev Biol ; 80: 43-49, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28899716

RESUMEN

Historically, ROS have been considered toxic molecules, especially when their intracellular concentration reaches high values. However, physiological levels of ROS support crucial cellular processes, acting as second messengers able to regulate intrinsic signaling pathways. Specifically, both the central and peripheral nervous systems are especially susceptible to changes in the redox state, developing either a defense or adaptive response depending on the concentration, source and duration of the pro-oxidative stimuli. In this review, we summarize classical and modern concepts regarding ROS physiology, with an emphasis on the role of the NADPH oxidase (NOX) complex, the main enzymatic and regulated source of ROS in the nervous system. We discuss how ROS and redox state contribute to neurogenesis, polarization and maturation of neurons, providing a context for the spatio-temporal conditions in which ROS modulate neural fate, discriminating between "oxidative distress", and "oxidative eustress". Finally, we present a brief discussion about the "physiological range of ROS concentration", and suggest that these values depend on several parameters, including cell type, developmental stage, and the source and type of pro-oxidative molecule.


Asunto(s)
Sistema Nervioso/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Oxidación-Reducción , Transducción de Señal/fisiología
12.
Acta Neuropathol ; 140(5): 737-764, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32642868

RESUMEN

Impaired neuronal proteostasis is a salient feature of many neurodegenerative diseases, highlighting alterations in the function of the endoplasmic reticulum (ER). We previously reported that targeting the transcription factor XBP1, a key mediator of the ER stress response, delays disease progression and reduces protein aggregation in various models of neurodegeneration. To identify disease modifier genes that may explain the neuroprotective effects of XBP1 deficiency, we performed gene expression profiling of brain cortex and striatum of these animals and uncovered insulin-like growth factor 2 (Igf2) as the major upregulated gene. Here, we studied the impact of IGF2 signaling on protein aggregation in models of Huntington's disease (HD) as proof of concept. Cell culture studies revealed that IGF2 treatment decreases the load of intracellular aggregates of mutant huntingtin and a polyglutamine peptide. These results were validated using induced pluripotent stem cells (iPSC)-derived medium spiny neurons from HD patients and spinocerebellar ataxia cases. The reduction in the levels of mutant huntingtin was associated with a decrease in the half-life of the intracellular protein. The decrease in the levels of abnormal protein aggregation triggered by IGF2 was independent of the activity of autophagy and the proteasome pathways, the two main routes for mutant huntingtin clearance. Conversely, IGF2 signaling enhanced the secretion of soluble mutant huntingtin species through exosomes and microvesicles involving changes in actin dynamics. Administration of IGF2 into the brain of HD mice using gene therapy led to a significant decrease in the levels of mutant huntingtin in three different animal models. Moreover, analysis of human postmortem brain tissue and blood samples from HD patients showed a reduction in IGF2 level. This study identifies IGF2 as a relevant factor deregulated in HD, operating as a disease modifier that buffers the accumulation of abnormal protein species.


Asunto(s)
Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Agregación Patológica de Proteínas/metabolismo , Animales , Humanos , Factor II del Crecimiento Similar a la Insulina/farmacología , Ratones , Ratones Transgénicos , Agregado de Proteínas/efectos de los fármacos
13.
BMC Geriatr ; 20(1): 505, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33238908

RESUMEN

BACKGROUND: With the global population aging and life expectancy increasing, dementia has turned a priority in the health care system. In Chile, dementia is one of the most important causes of disability in the elderly and the most rapidly growing cause of death in the last 20 years. Cognitive complaint is considered a predictor for cognitive and functional decline, incident mild cognitive impairment, and incident dementia. The GERO cohort is the Chilean core clinical project of the Geroscience Center for Brain Health and Metabolism (GERO). The objective of the GERO cohort is to analyze the rate of functional decline and progression to clinical dementia and their associated risk factors in a community-dwelling elderly with subjective cognitive complaint, through a population-based study. We also aim to undertake clinical research on brain ageing and dementia disorders, to create data and biobanks with the appropriate infrastructure to conduct other studies and facilitate to the national and international scientific community access to the data and samples for research. METHODS: The GERO cohort aims the recruitment of 300 elderly subjects (> 70 years) from Santiago (Chile), following them up for at least 3 years. Eligible people are adults not diagnosed with dementia with subjective cognitive complaint, which are reported either by the participant, a proxy or both. Participants are identified through a household census. The protocol for evaluation is based on a multidimensional approach including socio-demographic, biomedical, psychosocial, neuropsychological, neuropsychiatric and motor assessments. Neuroimaging, blood and stool samples are also obtained. This multidimensional evaluation is carried out in a baseline and 2 follow-ups assessments, at 18 and 36 months. In addition, in months 6, 12, 24, and 30, a telephone interview is performed in order to keep contact with the participants and to assess general well-being. DISCUSSION: Our work will allow us to determine multidimensional risks factors associated with functional decline and conversion to dementia in elderly with subjective cognitive complain. The aim of our GERO group is to establish the capacity to foster cutting edge and multidisciplinary research on aging in Chile including basic and clinical research. TRIAL REGISTRATION: NCT04265482 in ClinicalTrials.gov. Registration Date: February 11, 2020. Retrospectively Registered.


Asunto(s)
Alcoholismo , Disfunción Cognitiva , Actividades Cotidianas , Anciano , Chile/epidemiología , Cognición , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/epidemiología , Estudios de Cohortes , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Glicoproteínas de Membrana , Pruebas Neuropsicológicas , Proyectos Piloto , Calidad de Vida , Receptores Inmunológicos
14.
J Neurosci ; 37(4): 790-806, 2017 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28123016

RESUMEN

Neurons communicate with each other through their axons and dendrites. However, a full characterization of the molecular mechanisms involved in axon and dendrite formation is still incomplete. Neurite outgrowth requires the supply of membrane components for surface expansion. Two membrane sources for axon outgrowth are suggested: Golgi secretary vesicles and endocytic recycling endosomes. In non-neuronal cells, trafficking of secretary vesicles from Golgi is regulated by Rab8, a member of Rab small GTPases, and that of recycling endosomes is by Rab11, another member of Rabs. However, whether these vesicles are coordinately or independently transported in growing axons is unknown. Herein, we find that GRAB, a guanine nucleotide exchange factor for Rab8, is a novel regulator of axon outgrowth. Knockdown of GRAB suppressed axon outgrowth of cultured mouse brain cortical neurons. GRAB mediates the interaction between Rab11A and Rab8A, and this activity is regulated by phosphorylation at Ser169 and Ser180 by Cdk5-p35. The nonphosphorylatable GRAB mutant S169/180A promoted axonal outgrowth to a greater extent than did the phosphomimetic GRAB mutant S169/180D. Phosphorylation of GRAB suppressed its guanine nucleotide exchange factor activity and its ability to recruit Rab8A- to Rab11A-positive endosomes. In vivo function of GRAB and its Cdk5-phophorylation were shown in migration and process formation of developing neurons in embryonic mouse brains. These results indicate that GRAB regulates axonal outgrowth via activation and recruitment of Rab8A- to Rab11A-positive endosomes in a Cdk5-dependent manner. SIGNIFICANCE STATEMENT: While axon outgrowth requires membrane supply for surface expansion, the molecular mechanisms regulating the membrane transport in growing axons remain unclear. Here, we demonstrate that GRAB, a guanine nucleotide exchange factor for Rab8, is a novel regulator of axon outgrowth. GRAB promotes the axonal membrane transport by mediating the interaction between Rab11 and Rab8 in neurons. The activity of GRAB is regulated by phosphorylation with Cdk5. We describe an in vivo role for GRAB and its Cdk5 phosphorylation during neuronal migration and process formation in embryonic brains. Thus, the membrane supply for axonal outgrowth is regulated by Cdk5 through the Rab11-GRAB-Rab8 cascade.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Proyección Neuronal/fisiología , Fosfotransferasas/fisiología , Proteínas de Unión al GTP rab/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Femenino , Ratones , Ratones Endogámicos ICR , Fosforilación , Embarazo , Ratas , Transducción de Señal/fisiología
15.
Cell Mol Life Sci ; 74(1): 153-172, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506619

RESUMEN

CDK5 is a serine/threonine kinase that is involved in the normal function of the adult brain and plays a role in neurotransmission and synaptic plasticity. However, its over-regulation has been associated with Tau hyperphosphorylation and cognitive deficits. Our previous studies have demonstrated that CDK5 targeting using shRNA-miR provides neuroprotection and prevents cognitive deficits. Dendritic spine morphogenesis and forms of long-term synaptic plasticity-such as long-term potentiation (LTP)-have been proposed as essential processes of neuroplasticity. However, whether CDK5 participates in these processes remains controversial and depends on the experimental model. Using wild-type mice that received injections of CDK5 shRNA-miR in CA1 showed an increased LTP and recovered the PPF in deficient LTP of APPswe/PS1Δ9 transgenic mice. On mature hippocampal neurons CDK5, shRNA-miR for 12 days induced increased dendritic protrusion morphogenesis, which was dependent on Rac activity. In addition, silencing of CDK5 increased BDNF expression, temporarily increased phosphorylation of CaMKII, ERK, and CREB; and facilitated calcium signaling in neurites. Together, our data suggest that CDK5 downregulation induces synaptic plasticity in mature neurons involving Ca2+ signaling and BDNF/CREB activation.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/genética , Regulación hacia Abajo , Hipocampo/citología , Plasticidad Neuronal , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Señalización del Calcio , Células Cultivadas , Quinasa 5 Dependiente de la Ciclina/metabolismo , Femenino , Silenciador del Gen , Hipocampo/fisiología , Potenciación a Largo Plazo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuritas/metabolismo , Fosforilación , Ratas Wistar , Transducción de Señal , Regulación hacia Arriba
16.
J Neurosci ; 36(27): 7298-313, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27383602

RESUMEN

UNLABELLED: Rab35 is a key protein for cargo loading in the recycling endosome. In neuronal immortalized cells, Rab35 promotes neurite differentiation. Here we describe that Rab35 favors axon elongation in rat primary neurons in an activity-dependent manner. In addition, we show that the p53-related protein kinase (PRPK) negatively regulates axonal elongation by reducing Rab35 protein levels through the ubiquitin-proteasome degradation pathway. PRPK-induced Rab35 degradation is regulated by its interaction with microtubule-associated protein 1B (MAP1B), a microtubule stabilizing binding protein essential for axon elongation. Consistently, axon defects found in MAP1B knock-out neurons were reversed by Rab35 overexpression or PRPK inactivation suggesting an epistatic relationship among these proteins. These results define a novel mechanism to support axonal elongation, by which MAP1B prevents PRPK-induced Rab35 degradation. Such a mechanism allows Rab35-mediated axonal elongation and connects the regulation of actin dynamics with membrane trafficking. In addition, our study reveals for the first time that the ubiquitin-proteasome degradation pathway regulates a Rab GTPase. SIGNIFICANCE STATEMENT: Rab35 is required for axonal outgrowth. We define that its protein levels are negatively regulated by p53-related protein kinase (PRPK). We show that microtubule-associated protein 1B (MAP1B) interacts with PRPK, preventing PRPK-dependent Rab35 proteasome degradation. We demonstrate that Rab35 regulates Cdc42 activity in neurons. This is the first evidence showing that a Rab protein is regulated by degradation dependent on the ubiquitin-proteasome system.


Asunto(s)
Axones/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Línea Celular Transformada , Células Cultivadas , Chlorocebus aethiops , Embrión de Mamíferos , Regulación de la Expresión Génica/genética , Hipocampo/citología , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Ratas , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rab/genética
17.
J Neurosci ; 36(43): 11107-11119, 2016 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-27798190

RESUMEN

Physiological levels of ROS support neurite outgrowth and axonal specification, but the mechanisms by which ROS are able to shape neurons remain unknown. Ca2+, a broad intracellular second messenger, promotes both Rac1 activation and neurite extension. Ca2+ release from the endoplasmic reticulum, mediated by both the IP3R1 and ryanodine receptor (RyR) channels, requires physiological ROS levels that are mainly sustained by the NADPH oxidase (NOX) complex. In this work, we explore the contribution of the link between NOX and RyR-mediated Ca2+ release toward axonal specification of rat hippocampal neurons. Using genetic approaches, we find that NOX activation promotes both axonal development and Rac1 activation through a RyR-mediated mechanism, which in turn activates NOX through Rac1, one of the NOX subunits. Collectively, these data suggest a feedforward mechanism that integrates both NOX activity and RyR-mediated Ca2+ release to support cellular mechanisms involved in axon development. SIGNIFICANCE STATEMENT: High levels of ROS are frequently associated with oxidative stress and disease. In contrast, physiological levels of ROS, mainly sustained by the NADPH oxidase (NOX) complex, promote neuronal development and axonal growth. However, the mechanisms by which ROS shape neurons have not been described. Our work suggests that NOX-derived ROS promote axonal growth by regulating Rac1 activity, a molecular determinant of axonal growth, through a ryanodine receptor (RyR)-mediated Ca2+ release mechanism. In addition, Rac1, one of the NOX subunits, was activated after RyR-mediated Ca2+ release, suggesting a feedforward mechanism between NOX and RyR. Collectively, our data suggest a novel mechanism that is instrumental in sustaining physiological levels of ROS required for axonal growth of hippocampal neurons.


Asunto(s)
Orientación del Axón/fisiología , Señalización del Calcio/fisiología , Retroalimentación Fisiológica/fisiología , NADPH Oxidasas/metabolismo , Neuronas/fisiología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/fisiología , Hipocampo/ultraestructura , Masculino , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
18.
J Neurochem ; 142(1): 140-152, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28266714

RESUMEN

Alzheimer's disease (AD) is characterized by extracellular senile plaques, intracellular neurofibrillary tangles, and neuronal death. Aggregated amyloid-ß (Aß) induces inflammation and oxidative stress, which have pivotal roles in the pathogenesis of AD. Hepcidin is a key regulator of systemic iron homeostasis. Recently, an anti-inflammatory response to hepcidin was reported in macrophages. Under the hypothesis that hepcidin mediates anti-inflammatory response in the brain, in this study, we evaluated the putative anti-inflammatory role of hepcidin on Aß-activated astrocytes and microglia. Primary culture of astrocytes and microglia were treated with Aß, with or without hepcidin, and cytokine levels were then evaluated. In addition, the toxicity of Aß-treated astrocyte- or microglia-conditioned media was tested on neurons, evaluating cellular death and oxidative stress generation. Finally, mice were injected in the right lateral ventricle with Aß, with or without hepcidin, and hippocampus glial activation and oxidative stress were evaluated. Pre-treatment with hepcidin reduced the expression and secretion of TNF-α and IL-6 in astrocytes and microglia treated with Aß. Hepcidin also reduced neurotoxicity and oxidative damage triggered by conditioned media obtained from astrocytes and microglia treated with Aß. Stereotaxic intracerebral injection of hepcidin reduced glial activation and oxidative damage triggered by Aß injection in mice. Overall, these results are consistent with the hypothesis that in astrocytes and microglia hepcidin down-regulates the inflammatory and pro-oxidant processes induced by Aß, thus protecting neighboring neurons. This is a newly described property of hepcidin in the central nervous system, which may be relevant for the development of strategies to prevent the neurodegenerative process associated with AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Antioxidantes/farmacología , Astrocitos/efectos de los fármacos , Hepcidinas/farmacología , Inflamación/inducido químicamente , Inflamación/prevención & control , Microglía/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Animales , Medios de Cultivo Condicionados , Hipocampo/patología , Inflamación/metabolismo , Inyecciones Intraventriculares , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Factor de Necrosis Tumoral alfa/metabolismo
19.
EMBO J ; 32(16): 2287-99, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23881099

RESUMEN

The microtubule-associated protein 1B (MAP1B) plays critical roles in neurite growth and synapse maturation during brain development. This protein is well expressed in the adult brain. However, its function in mature neurons remains unknown. We have used a genetically modified mouse model and shRNA techniques to assess the role of MAP1B at established synapses, bypassing MAP1B functions during neuronal development. Under these conditions, we found that MAP1B deficiency alters synaptic plasticity by specifically impairing long-term depression (LTD) expression. Interestingly, this is due to a failure to trigger AMPA receptor endocytosis and spine shrinkage during LTD. These defects are accompanied by an impaired targeting of the Rac1 activator Tiam1 at synaptic compartments. Accordingly, LTD and AMPA receptor endocytosis are restored in MAP1B-deficient neurons by providing additional Rac1. Therefore, these results indicate that the MAP1B-Tiam1-Rac1 relay is essential for spine structural plasticity and removal of AMPA receptors from synapses during LTD. This work highlights the importance of MAPs as signalling hubs controlling the actin cytoskeleton and receptor trafficking during plasticity in mature neurons.


Asunto(s)
Endocitosis/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Plasticidad Neuronal/fisiología , Receptores AMPA/metabolismo , Sinapsis/fisiología , Animales , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hipocampo/citología , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/deficiencia , Neuropéptidos , Técnicas de Placa-Clamp , ARN Interferente Pequeño/genética , Columna Vertebral/citología , Estadísticas no Paramétricas , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteína de Unión al GTP rac1
20.
J Cell Sci ; 128(16): 2989-95, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26101350

RESUMEN

Reactive oxygen species (ROS) produced by the NADPH oxidase (NOX) complex play important physiological and pathological roles in neurotransmission and neurodegeneration, respectively. However, the contribution of ROS to the molecular mechanisms involved in neuronal polarity and axon elongation is not well understood. In this work, we found that loss of NOX complex function altered neuronal polarization and decreased axonal length by a mechanism that involves actin cytoskeleton dynamics. These results indicate that physiological levels of ROS produced by the NOX complex modulate hippocampal neuronal polarity and axonal growth in vitro.


Asunto(s)
Citoesqueleto de Actina/genética , Axones/metabolismo , NADPH Oxidasas/biosíntesis , Neuronas/metabolismo , Citoesqueleto de Actina/metabolismo , Polaridad Celular/genética , Regulación del Desarrollo de la Expresión Génica , Hipocampo/metabolismo , Hipocampo/patología , Humanos , NADPH Oxidasas/genética , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA