Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
FASEB J ; 38(10): e23659, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38733301

RESUMEN

HDAC3 inhibition has been shown to improve memory and reduce amyloid-ß (Aß) in Alzheimer's disease (AD) models, but the underlying mechanisms are unclear. We investigated the molecular effects of HDAC3 inhibition on AD pathology, using in vitro and ex vivo models of AD, based on our finding that HDAC3 expression is increased in AD brains. For this purpose, N2a mouse neuroblastoma cells as well as organotypic brain cultures (OBCSs) of 5XFAD and wild-type mice were incubated with various concentrations of the HDAC3 selective inhibitor RGFP966 (0.1-10 µM) for 24 h. Treatment with RGFP966 or HDAC3 knockdown in N2a cells was associated with an increase on amyloid precursor protein (APP) and mRNA expressions, without alterations in Aß42 secretion. In vitro chromatin immunoprecipitation analysis revealed enriched HDAC3 binding at APP promoter regions. The increase in APP expression was also detected in OBCSs from 5XFAD mice incubated with 1 µM RGFP966, without changes in Aß. In addition, HDAC3 inhibition resulted in a reduction of activated Iba-1-positive microglia and astrocytes in 5XFAD slices, which was not observed in OBCSs from wild-type mice. mRNA sequencing analysis revealed that HDAC3 inhibition modulated neuronal regenerative pathways related to neurogenesis, differentiation, axonogenesis, and dendritic spine density in OBCSs. Our findings highlight the complexity and diversity of the effects of HDAC3 inhibition on AD models and suggest that HDAC3 may have multiple roles in the regulation of APP expression and processing, as well as in the modulation of neuroinflammatory and neuroprotective genes.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Histona Desacetilasas , Animales , Ratones , Acrilamidas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Fenilendiaminas/farmacología
2.
J Neuroinflammation ; 21(1): 91, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38609999

RESUMEN

OBJECTIVE: Soluble CD27 is a promising cerebrospinal fluid inflammatory biomarker in multiple sclerosis. In this study, we investigate relevant immune and neuro-pathological features of soluble CD27 in multiple sclerosis. METHODS: Protein levels of soluble CD27 were correlated to inflammatory cell subpopulations and inflammatory cytokines and chemokines detected in cerebrospinal fluid of 137 patients with multiple sclerosis and 47 patients with inflammatory and non-inflammatory neurological disease from three independent cohorts. Production of soluble CD27 was investigated in cell cultures of activated T and B cells and CD27-knockout T cells. In a study including matched cerebrospinal fluid and post-mortem brain tissues of patients with multiple sclerosis and control cases, levels of soluble CD27 were correlated with perivascular and meningeal infiltrates and with neuropathological features. RESULTS: We demonstrate that soluble CD27 favours the differentiation of interferon-γ-producing T cells and is released through a secretory mechanism activated by TCR engagement and regulated by neutral sphingomyelinase. We also show that the levels of soluble CD27 correlate with the representation of inflammatory T cell subsets in the CSF of patients with relapsing-remitting multiple sclerosis and with the magnitude of perivascular and meningeal CD27 + CD4 + and CD8 + T cell infiltrates in post-mortem central nervous system tissue, defining a subgroup of patients with extensive active inflammatory lesions. INTERPRETATION: Our results demonstrate that soluble CD27 is a biomarker of disease activity, potentially informative for personalized treatment and monitoring of treatment outcomes.


Asunto(s)
Esclerosis Múltiple Recurrente-Remitente , Esclerosis Múltiple , Humanos , Linfocitos T CD8-positivos , Sistema Nervioso Central , Biomarcadores
3.
EMBO J ; 38(13): e101032, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31268609

RESUMEN

The molecular mechanisms discriminating between regenerative failure and success remain elusive. While a regeneration-competent peripheral nerve injury mounts a regenerative gene expression response in bipolar dorsal root ganglia (DRG) sensory neurons, a regeneration-incompetent central spinal cord injury does not. This dichotomic response offers a unique opportunity to investigate the fundamental biological mechanisms underpinning regenerative ability. Following a pharmacological screen with small-molecule inhibitors targeting key epigenetic enzymes in DRG neurons, we identified HDAC3 signalling as a novel candidate brake to axonal regenerative growth. In vivo, we determined that only a regenerative peripheral but not a central spinal injury induces an increase in calcium, which activates protein phosphatase 4 that in turn dephosphorylates HDAC3, thus impairing its activity and enhancing histone acetylation. Bioinformatics analysis of ex vivo H3K9ac ChIPseq and RNAseq from DRG followed by promoter acetylation and protein expression studies implicated HDAC3 in the regulation of multiple regenerative pathways. Finally, genetic or pharmacological HDAC3 inhibition overcame regenerative failure of sensory axons following spinal cord injury. Together, these data indicate that PP4-dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure.


Asunto(s)
Ganglios Espinales/fisiología , Histona Desacetilasas/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Axones , Células Cultivadas , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Femenino , Masculino , Ratones , Regeneración Nerviosa , Fosforilación/efectos de los fármacos , Transducción de Señal
4.
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
5.
Proc Natl Acad Sci U S A ; 109(34): E2284-93, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22826225

RESUMEN

The epigenetic silencing of exogenous transcriptional units integrated into the genome represents a critical problem both for long-term gene therapy efficacy and for the eradication of latent viral infections. We report here that limitation of essential amino acids, such as methionine and cysteine, causes selective up-regulation of exogenous transgene expression in mammalian cells. Prolonged amino acid deprivation led to significant and reversible increase in the expression levels of stably integrated transgenes transcribed by means of viral or human promoters in HeLa cells. This phenomenon was mediated by epigenetic chromatin modifications, because histone deacetylase (HDAC) inhibitors reproduced starvation-induced transgene up-regulation, and transcriptome analysis, ChIP, and pharmacological and RNAi approaches revealed that a specific class II HDAC, namely HDAC4, plays a critical role in maintaining the silencing of exogenous transgenes. This mechanism was also operational in cells chronically infected with HIV-1, the etiological agent of AIDS, in a latency state. Indeed, both amino acid starvation and pharmacological inhibition of HDAC4 promoted reactivation of HIV-1 transcription and reverse transcriptase activity production in HDAC4(+) ACH-2 T-lymphocytic cells but not in HDAC4(-) U1 promonocytic cells. Thus, amino acid deprivation leads to transcriptional derepression of silenced transgenes, including integrated plasmids and retroviruses, by a process involving inactivation or down-regulation of HDAC4. These findings suggest that selective targeting of HDAC4 might represent a unique strategy for modulating the expression of therapeutic viral vectors, as well as that of integrated HIV-1 proviruses in latent reservoirs without significant cytotoxicity.


Asunto(s)
Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Silenciador del Gen , VIH-1/genética , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Albinismo Ocular/metabolismo , Metilación de ADN , Proteínas del Ojo/metabolismo , Células HeLa , Humanos , Glicoproteínas de Membrana/metabolismo , Regiones Promotoras Genéticas , Provirus/genética , Activación Transcripcional , Transgenes , Factor de Necrosis Tumoral alfa/metabolismo , Tirosina/química
7.
bioRxiv ; 2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38895406

RESUMEN

The in vivo three-dimensional genomic architecture of adult mature neurons at homeostasis and after medically relevant perturbations such as axonal injury remains elusive. Here we address this knowledge gap by mapping the three-dimensional chromatin architecture and gene expression programme at homeostasis and after sciatic nerve injury in wild-type and cohesin-deficient mouse sensory dorsal root ganglia neurons via combinatorial Hi-C and RNA-seq. We find that cohesin is required for the full induction of the regenerative transcriptional program, by organising 3D genomic domains required for the activation of regenerative genes. Importantly, loss of cohesin results in disruption of chromatin architecture at regenerative genes and severely impaired nerve regeneration. Together, these data provide an original three-dimensional chromatin map of adult sensory neurons in vivo and demonstrate a role for cohesin-dependent chromatin interactions in neuronal regeneration.

8.
Methods Mol Biol ; 2636: 101-144, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36881298

RESUMEN

RNA sequencing (RNA-seq), chromatin immunoprecipitation sequencing (ChIP-seq), and assay for transposase-accessible chromatin sequencing (ATAC-seq) are genome-wide techniques that provide information relative to gene expression, chromatin binding sites, and chromatin accessibility, respectively. Here we describe RNA-seq, H3K9ac, H3K27ac and H3K27me3 ChIP-seq, and ATAC-seq in dorsal root ganglia (DRG) after sciatic nerve or dorsal column axotomy, to characterize the transcriptional and epigenetic signatures of DRG upon regenerative vs non-regenerative axonal lesion.


Asunto(s)
Epigenómica , Ganglios Espinales , Axones , Axotomía , Cromatina
9.
Cell Metab ; 35(12): 2153-2164.e4, 2023 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-37951214

RESUMEN

Nerve injuries cause permanent neurological disability due to limited axonal regeneration. Injury-dependent and -independent mechanisms have provided important insight into neuronal regeneration, however, common denominators underpinning regeneration remain elusive. A comparative analysis of transcriptomic datasets associated with neuronal regenerative ability revealed circadian rhythms as the most significantly enriched pathway. Subsequently, we demonstrated that sensory neurons possess an endogenous clock and that their regenerative ability displays diurnal oscillations in a murine model of sciatic nerve injury. Consistently, transcriptomic analysis showed a time-of-day-dependent enrichment for processes associated with axonal regeneration and the circadian clock. Conditional deletion experiments demonstrated that Bmal1 is required for neuronal intrinsic circadian regeneration and target re-innervation. Lastly, lithium enhanced nerve regeneration in wild-type but not in clock-deficient mice. Together, these findings demonstrate that the molecular clock fine-tunes the regenerative ability of sensory neurons and propose compounds affecting clock pathways as a novel approach to nerve repair.


Asunto(s)
Relojes Circadianos , Ratones , Animales , Relojes Circadianos/genética , Ritmo Circadiano , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales , Factores de Transcripción ARNTL/genética
10.
Science ; 376(6594): eabd5926, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35549409

RESUMEN

Aging is associated with increased prevalence of axonal injuries characterized by poor regeneration and disability. However, the underlying mechanisms remain unclear. In our experiments, RNA sequencing of sciatic dorsal root ganglia (DRG) revealed significant aging-dependent enrichment in T cell signaling both before and after sciatic nerve injury (SNI) in mice. Lymphotoxin activated the transcription factor NF-κB, which induced expression of the chemokine CXCL13 by neurons. This in turn recruited CXCR5+CD8+ T cells to injured DRG neurons overexpressing major histocompatibility complex class I. CD8+ T cells repressed the axonal regeneration of DRG neurons via caspase 3 activation. CXCL13 neutralization prevented CXCR5+CD8+ T cell recruitment to the DRG and reversed aging-dependent regenerative decline, thereby promoting neurological recovery after SNI. Thus, axonal regeneration can be facilitated by antagonizing cross-talk between immune cells and neurons.


Asunto(s)
Envejecimiento , Axones , Linfocitos T CD8-positivos , Ganglios Espinales , Regeneración Nerviosa , Neuronas , Nervio Ciático , Envejecimiento/metabolismo , Animales , Axones/fisiología , Linfocitos T CD8-positivos/metabolismo , Ganglios Espinales/metabolismo , Ratones , Neuronas/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/fisiología
11.
Hum Mol Genet ; 17(22): 3487-501, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18697795

RESUMEN

The protein product of the ocular albinism type 1 gene, named OA1, is a pigment cell-specific G protein-coupled receptor exclusively localized to intracellular organelles, namely lysosomes and melanosomes. Loss of OA1 function leads to the formation of macromelanosomes, suggesting that this receptor is implicated in organelle biogenesis, however the mechanism involved in the pathogenesis of the disease remains obscure. We report here the identification of an unexpected abnormality in melanosome distribution both in retinal pigment epithelium (RPE) and skin melanocytes of Oa1-knock-out (KO) mice, consisting in a displacement of the organelles from the central cytoplasm towards the cell periphery. Despite their depletion from the microtubule (MT)-enriched perinuclear region, Oa1-KO melanosomes were able to aggregate at the centrosome upon disruption of the actin cytoskeleton or expression of a dominant-negative construct of myosin Va. Consistently, quantification of organelle transport in living cells revealed that Oa1-KO melanosomes displayed a severe reduction in MT-based motility; however, this defect was rescued to normal following inhibition of actin-dependent capture at the cell periphery. Together, these data point to a defective regulation of organelle transport in the absence of OA1 and imply that the cytoskeleton might represent a downstream effector of this receptor. Furthermore, our results enlighten a novel function for OA1 in pigment cells and suggest that ocular albinism type 1 might result from a different pathogenetic mechanism than previously thought, based on an organelle-autonomous signalling pathway implicated in the regulation of both membrane traffic and transport.


Asunto(s)
Proteínas del Ojo/metabolismo , Melanocitos/metabolismo , Melanosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Albinismo Ocular/genética , Albinismo Ocular/metabolismo , Animales , Citoesqueleto/fisiología , Proteínas del Ojo/genética , Humanos , Melanocitos/patología , Melanocitos/ultraestructura , Melanosomas/genética , Melanosomas/ultraestructura , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Microscopía Electrónica , Epitelio Pigmentado Ocular/citología , Receptores Acoplados a Proteínas G/genética
12.
Nat Metab ; 2(9): 918-933, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32778834

RESUMEN

Regeneration after injury occurs in axons that lie in the peripheral nervous system but fails in the central nervous system, thereby limiting functional recovery. Differences in axonal signalling in response to injury that might underpin this differential regenerative ability are poorly characterized. Combining axoplasmic proteomics from peripheral sciatic or central projecting dorsal root ganglion (DRG) axons with cell body RNA-seq, we uncover injury-dependent signalling pathways that are uniquely represented in peripheral versus central projecting sciatic DRG axons. We identify AMPK as a crucial regulator of axonal regenerative signalling that is specifically downregulated in injured peripheral, but not central, axons. We find that AMPK in DRG interacts with the 26S proteasome and its CaMKIIα-dependent regulatory subunit PSMC5 to promote AMPKα proteasomal degradation following sciatic axotomy. Conditional deletion of AMPKα1 promotes multiple regenerative signalling pathways after central axonal injury and stimulates robust axonal growth across the spinal cord injury site, suggesting inhibition of AMPK as a therapeutic strategy to enhance regeneration following spinal cord injury.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Axones , Ganglios Espinales/metabolismo , Regeneración Nerviosa , Células Receptoras Sensoriales/metabolismo , Traumatismos de la Médula Espinal/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Animales , Transporte Axonal , Axotomía , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Femenino , Ganglios Espinales/patología , Ratones , Ratones Endogámicos C57BL , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Nervio Ciático/metabolismo , Nervio Ciático/patología , Células Receptoras Sensoriales/patología , Traumatismos de la Médula Espinal/patología
13.
Nat Commun ; 11(1): 6425, 2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33349630

RESUMEN

Overcoming the restricted axonal regenerative ability that limits functional repair following a central nervous system injury remains a challenge. Here we report a regenerative paradigm that we call enriched conditioning, which combines environmental enrichment (EE) followed by a conditioning sciatic nerve axotomy that precedes a spinal cord injury (SCI). Enriched conditioning significantly increases the regenerative ability of dorsal root ganglia (DRG) sensory neurons compared to EE or a conditioning injury alone, propelling axon growth well beyond the spinal injury site. Mechanistically, we established that enriched conditioning relies on the unique neuronal intrinsic signaling axis PKC-STAT3-NADPH oxidase 2 (NOX2), enhancing redox signaling as shown by redox proteomics in DRG. Finally, NOX2 conditional deletion or overexpression respectively blocked or phenocopied enriched conditioning-dependent axon regeneration after SCI leading to improved functional recovery. These studies provide a paradigm that drives the regenerative ability of sensory neurons offering a potential redox-dependent regenerative model for mechanistic and therapeutic discoveries.


Asunto(s)
Regeneración Nerviosa , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología , Transducción de Señal , Traumatismos de la Médula Espinal/fisiopatología , Animales , Axones/patología , Axotomía , Ganglios Espinales/patología , Ratones Endogámicos C57BL , NADPH Oxidasa 2/metabolismo , Proyección Neuronal , Plasticidad Neuronal , Oxidación-Reducción , Fosforilación , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/metabolismo , Subunidades de Proteína/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/metabolismo , Nervio Ciático/fisiopatología , Regulación hacia Arriba
14.
Sci Transl Med ; 12(551)2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641489

RESUMEN

Cyclin-dependent-like kinase 5 (CDKL5) gene mutations lead to an X-linked disorder that is characterized by infantile epileptic encephalopathy, developmental delay, and hypotonia. However, we found that a substantial percentage of these patients also report a previously unrecognized anamnestic deficiency in pain perception. Consistent with a role in nociception, we found that CDKL5 is expressed selectively in nociceptive dorsal root ganglia (DRG) neurons in mice and in induced pluripotent stem cell (iPS)-derived human nociceptors. CDKL5-deficient mice display defective epidermal innervation, and conditional deletion of CDKL5 in DRG sensory neurons impairs nociception, phenocopying CDKL5 deficiency disorder in patients. Mechanistically, CDKL5 interacts with calcium/calmodulin-dependent protein kinase II α (CaMKIIα) to control outgrowth and transient receptor potential cation channel subfamily V member 1 (TRPV1)-dependent signaling, which are disrupted in both CDKL5 mutant murine DRG and human iPS-derived nociceptors. Together, these findings unveil a previously unrecognized role for CDKL5 in nociception, proposing an original regulatory mechanism for pain perception with implications for future therapeutics in CDKL5 deficiency disorder.


Asunto(s)
Células Receptoras Sensoriales , Transducción de Señal , Animales , Ciclinas , Modelos Animales de Enfermedad , Humanos , Ratones , Dolor , Proteínas Serina-Treonina Quinasas/genética
15.
Nat Neurosci ; 22(11): 1913-1924, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591560

RESUMEN

Axonal injury results in regenerative success or failure, depending on whether the axon lies in the peripheral or the CNS, respectively. The present study addresses whether epigenetic signatures in dorsal root ganglia discriminate between regenerative and non-regenerative axonal injury. Chromatin immunoprecipitation for the histone 3 (H3) post-translational modifications H3K9ac, H3K27ac and H3K27me3; an assay for transposase-accessible chromatin; and RNA sequencing were performed in dorsal root ganglia after sciatic nerve or dorsal column axotomy. Distinct histone acetylation and chromatin accessibility signatures correlated with gene expression after peripheral, but not central, axonal injury. DNA-footprinting analyses revealed new transcriptional regulators associated with regenerative ability. Machine-learning algorithms inferred the direction of most of the gene expression changes. Neuronal conditional deletion of the chromatin remodeler CCCTC-binding factor impaired nerve regeneration, implicating chromatin organization in the regenerative competence. Altogether, the present study offers the first epigenomic map providing insight into the transcriptional response to injury and the differential regenerative ability of sensory neurons.


Asunto(s)
Axones/fisiología , Epigenómica , Ganglios Espinales/fisiología , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales/fisiología , Acetilación , Algoritmos , Animales , Factor de Unión a CCCTC/genética , Cromatina/metabolismo , Femenino , Ganglios Espinales/lesiones , Expresión Génica , Histonas/metabolismo , Aprendizaje Automático , Masculino , Ratones , Ratones Transgénicos , Nervio Ciático/lesiones , Análisis de Secuencia de ARN
16.
Sci Transl Med ; 11(487)2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971452

RESUMEN

After a spinal cord injury, axons fail to regenerate in the adult mammalian central nervous system, leading to permanent deficits in sensory and motor functions. Increasing neuronal activity after an injury using electrical stimulation or rehabilitation can enhance neuronal plasticity and result in some degree of recovery; however, the underlying mechanisms remain poorly understood. We found that placing mice in an enriched environment before an injury enhanced the activity of proprioceptive dorsal root ganglion neurons, leading to a lasting increase in their regenerative potential. This effect was dependent on Creb-binding protein (Cbp)-mediated histone acetylation, which increased the expression of genes associated with the regenerative program. Intraperitoneal delivery of a small-molecule activator of Cbp at clinically relevant times promoted regeneration and sprouting of sensory and motor axons, as well as recovery of sensory and motor functions in both the mouse and rat model of spinal cord injury. Our findings showed that the increased regenerative capacity induced by enhancing neuronal activity is mediated by epigenetic reprogramming in rodent models of spinal cord injury. Understanding the mechanisms underlying activity-dependent neuronal plasticity led to the identification of potential molecular targets for improving recovery after spinal cord injury.


Asunto(s)
Axones/fisiología , Proteína de Unión a CREB/metabolismo , Ambiente , Histonas/metabolismo , Regeneración Nerviosa , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Acetilación , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Proteína p300 Asociada a E1A/metabolismo , Ganglios Espinales/patología , Ganglios Espinales/fisiopatología , Ratones , Neuronas Motoras/patología , Propiocepción , Recuperación de la Función , Células Receptoras Sensoriales/patología , Transducción de Señal , Traumatismos de la Médula Espinal/patología
17.
Neurotherapeutics ; 15(3): 529-540, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29948919

RESUMEN

Axonal regeneration relies on the expression of regenerative associated genes within a coordinated transcriptional programme, which is finely tuned as a result of the activation of several regenerative signalling pathways. In mammals, this chain of events occurs in neurons following peripheral axonal injury, however it fails upon axonal injury in the central nervous system, such as in the spinal cord and the brain. Accumulating evidence has been suggesting that epigenetic control is a key factor to initiate and sustain the regenerative transcriptional response and that it might contribute to regenerative success versus failure. This review will discuss experimental evidence so far showing a role for epigenetic regulation in models of peripheral and central nervous system axonal injury. It will also propose future directions to fill key knowledge gaps and to test whether epigenetic control might indeed discriminate between regenerative success and failure.


Asunto(s)
Epigénesis Genética/fisiología , Epigenómica/métodos , Regeneración Nerviosa/fisiología , Animales , Humanos , Mamíferos
18.
PLoS One ; 13(7): e0200783, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30020994

RESUMEN

In a variety of species, reduced food intake, and in particular protein or amino acid (AA) restriction, extends lifespan and healthspan. However, the underlying epigenetic and/or transcriptional mechanisms are largely unknown, and dissection of specific pathways in cultured cells may contribute to filling this gap. We have previously shown that, in mammalian cells, deprivation of essential AAs (methionine/cysteine or tyrosine) leads to the transcriptional reactivation of integrated silenced transgenes, including plasmid and retroviral vectors and latent HIV-1 provirus, by a process involving epigenetic chromatic remodeling and histone acetylation. Here we show that the deprivation of methionine/cysteine also leads to the transcriptional upregulation of endogenous retroviruses, suggesting that essential AA starvation affects the expression not only of exogenous non-native DNA sequences, but also of endogenous anciently-integrated and silenced parasitic elements of the genome. Moreover, we show that the transgene reactivation response is highly conserved in different mammalian cell types, and it is reproducible with deprivation of most essential AAs. The General Control Non-derepressible 2 (GCN2) kinase and the downstream integrated stress response represent the best candidates mediating this process; however, by pharmacological approaches, RNA interference and genomic editing, we demonstrate that they are not implicated. Instead, the response requires MEK/ERK and/or JNK activity and is reproduced by ribosomal inhibitors, suggesting that it is triggered by a novel nutrient-sensing and signaling pathway, initiated by translational block at the ribosome, and independent of mTOR and GCN2. Overall, these findings point to a general transcriptional response to essential AA deprivation, which affects the expression of non-native genomic sequences, with relevant implications for the epigenetic/transcriptional effects of AA restriction in health and disease.


Asunto(s)
Aminoácidos Esenciales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Aminoácidos Esenciales/deficiencia , Animales , Western Blotting , Sistemas CRISPR-Cas , Línea Celular , Edición Génica , Células HeLa , Células Hep G2 , Humanos , Ratones , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Activación Transcripcional/genética , Activación Transcripcional/fisiología
19.
Nat Cell Biol ; 20(3): 307-319, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29434374

RESUMEN

Reactive oxygen species (ROS) contribute to tissue damage and remodelling mediated by the inflammatory response after injury. Here we show that ROS, which promote axonal dieback and degeneration after injury, are also required for axonal regeneration and functional recovery after spinal injury. We find that ROS production in the injured sciatic nerve and dorsal root ganglia requires CX3CR1-dependent recruitment of inflammatory cells. Next, exosomes containing functional NADPH oxidase 2 complexes are released from macrophages and incorporated into injured axons via endocytosis. Once in axonal endosomes, active NOX2 is retrogradely transported to the cell body through an importin-ß1-dynein-dependent mechanism. Endosomal NOX2 oxidizes PTEN, which leads to its inactivation, thus stimulating PI3K-phosporylated (p-)Akt signalling and regenerative outgrowth. Challenging the view that ROS are exclusively involved in nerve degeneration, we propose a previously unrecognized role of ROS in mammalian axonal regeneration through a NOX2-PI3K-p-Akt signalling pathway.


Asunto(s)
Axones/enzimología , Exosomas/enzimología , Ganglios Espinales/enzimología , NADPH Oxidasa 2/metabolismo , Degeneración Nerviosa , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/enzimología , Especies Reactivas de Oxígeno/metabolismo , Nervio Ciático/enzimología , Traumatismos de la Médula Espinal/enzimología , Animales , Axones/patología , Receptor 1 de Quimiocinas CX3C/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Dineínas/metabolismo , Endocitosis , Endosomas/enzimología , Endosomas/patología , Exosomas/patología , Ganglios Espinales/lesiones , Ganglios Espinales/patología , Macrófagos/enzimología , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2/deficiencia , NADPH Oxidasa 2/genética , Proteínas Nucleares/metabolismo , Fosfohidrolasa PTEN/metabolismo , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/patología , Traumatismos de los Nervios Periféricos/fisiopatología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Transducción de Señal , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , beta Carioferinas
20.
Nat Cell Biol ; 20(9): 1098, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29520084

RESUMEN

In the version of this Article originally published, the affiliations for Roland A. Fleck and José Antonio Del Río were incorrect due to a technical error that resulted in affiliations 8 and 9 being switched. The correct affiliations are: Roland A. Fleck: 8Centre for Ultrastructural Imaging, Kings College London, London, UK. José Antonio Del Río: 2Cellular and Molecular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain; 9Department of Cell Biology, Physiology and Immunology, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; 10Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain. This has now been amended in all online versions of the Article.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA