Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 607(7919): 585-592, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35732737

RESUMEN

The regenerative potential of mammalian peripheral nervous system neurons after injury is critically limited by their slow axonal regenerative rate1. Regenerative ability is influenced by both injury-dependent and injury-independent mechanisms2. Among the latter, environmental factors such as exercise and environmental enrichment have been shown to affect signalling pathways that promote axonal regeneration3. Several of these pathways, including modifications in gene transcription and protein synthesis, mitochondrial metabolism and the release of neurotrophins, can be activated by intermittent fasting (IF)4,5. However, whether IF influences the axonal regenerative ability remains to be investigated. Here we show that IF promotes axonal regeneration after sciatic nerve crush in mice through an unexpected mechanism that relies on the gram-positive gut microbiome and an increase in the gut bacteria-derived metabolite indole-3-propionic acid (IPA) in the serum. IPA production by Clostridium sporogenes is required for efficient axonal regeneration, and delivery of IPA after sciatic injury significantly enhances axonal regeneration, accelerating the recovery of sensory function. Mechanistically, RNA sequencing analysis from sciatic dorsal root ganglia suggested a role for neutrophil chemotaxis in the IPA-dependent regenerative phenotype, which was confirmed by inhibition of neutrophil chemotaxis. Our results demonstrate the ability of a microbiome-derived metabolite, such as IPA, to facilitate regeneration and functional recovery of sensory axons through an immune-mediated mechanism.


Asunto(s)
Indoles , Regeneración Nerviosa , Propionatos , Cicatrización de Heridas , Animales , Ratones , Axones/efectos de los fármacos , Axones/fisiología , Quimiotaxis de Leucocito , Clostridium/metabolismo , Ayuno , Ganglios Espinales/metabolismo , Microbioma Gastrointestinal , Indoles/sangre , Indoles/metabolismo , Indoles/farmacología , Compresión Nerviosa , Factores de Crecimiento Nervioso/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Neutrófilos/citología , Neutrófilos/inmunología , Propionatos/sangre , Propionatos/metabolismo , Propionatos/farmacología , Recuperación de la Función , Nervio Ciático/lesiones , Análisis de Secuencia de ARN , Cicatrización de Heridas/efectos de los fármacos
2.
Proc Natl Acad Sci U S A ; 121(38): e2402518121, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39254997

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 program at homeostasis and after sciatic nerve injury in wild-type and cohesin-deficient mouse sensory dorsal root ganglia neurons via combinatorial Hi-C, promoter-capture Hi-C, CUT&Tag for H3K27ac and RNA-seq. We find that genes involved in axonal regeneration form long-range, complex chromatin loops, and that cohesin is required for the full induction of the regenerative transcriptional program. Importantly, loss of cohesin results in disruption of chromatin architecture and severely impaired nerve regeneration. Complex enhancer-promoter loops are also enriched in the human fetal cortical plate, where the axonal growth potential is highest, and are lost in mature adult neurons. Together, these data provide an original three-dimensional chromatin map of adult sensory neurons in vivo and demonstrate a role for cohesin-dependent long-range promoter interactions in nerve regeneration.


Asunto(s)
Axones , Cromatina , Cohesinas , Regeneración Nerviosa , Regiones Promotoras Genéticas , Células Receptoras Sensoriales , Animales , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Ratones , Regiones Promotoras Genéticas/genética , Cromatina/metabolismo , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Axones/metabolismo , Axones/fisiología , Humanos , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Elementos de Facilitación Genéticos/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ganglios Espinales/metabolismo , Ganglios Espinales/citología , Nervio Ciático/metabolismo
3.
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
4.
PLoS Biol ; 20(9): e3001310, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36126035

RESUMEN

The interruption of spinal circuitry following spinal cord injury (SCI) disrupts neural activity and is followed by a failure to mount an effective regenerative response resulting in permanent neurological disability. Functional recovery requires the enhancement of axonal and synaptic plasticity of spared as well as injured fibres, which need to sprout and/or regenerate to form new connections. Here, we have investigated whether the epigenetic stimulation of the regenerative gene expression program can overcome the current inability to promote neurological recovery in chronic SCI with severe disability. We delivered the CBP/p300 activator CSP-TTK21 or vehicle CSP weekly between week 12 and 22 following a transection model of SCI in mice housed in an enriched environment. Data analysis showed that CSP-TTK21 enhanced classical regenerative signalling in dorsal root ganglia sensory but not cortical motor neurons, stimulated motor and sensory axon growth, sprouting, and synaptic plasticity, but failed to promote neurological sensorimotor recovery. This work provides direct evidence that clinically suitable pharmacological CBP/p300 activation can promote the expression of regeneration-associated genes and axonal growth in a chronic SCI with severe neurological disability.


Asunto(s)
Regeneración Nerviosa , Traumatismos de la Médula Espinal , Animales , Axones/metabolismo , Ratones , Regeneración Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo
5.
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
6.
Medicina (Kaunas) ; 59(5)2023 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-37241169

RESUMEN

Objective: The aim of this study is to show our experience with the correct management of patients suffering from odontogenic sinusitis with oroantral communication and fistula. Methods: According to the inclusion criteria, 41 patients were enrolled in this retrospective study with a diagnosis of odontogenic sinusitis with oroantral communication and fistula; 1 patient with pre-implantological complication, 14 with implantological complications, and 26 with classical complications. Results: Two patients were treated with a fractioned combined approach, 13 patients were treated with an oral approach only, and 26 patients were treated with a combination. There was a complete resolution of the symptoms and closure of the fistula in all the patients enrolled. Conclusions: In our study, in all 41 patients, there was a surgical success. The best option is to use a multidisciplinary approach for patients suffering from odontogenic sinusitis.


Asunto(s)
Sinusitis Maxilar , Sinusitis , Humanos , Sinusitis Maxilar/cirugía , Sinusitis Maxilar/complicaciones , Estudios Retrospectivos , Sinusitis/complicaciones , Fístula Oroantral/etiología , Fístula Oroantral/cirugía
7.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35054943

RESUMEN

While about half of the population experience persistent pain associated with tissue damages during their lifetime, current symptom-based approaches often fail to reduce such pain to a satisfactory level. To provide better patient care, mechanism-based analgesic approaches must be developed, which necessitates a comprehensive understanding of the nociceptive mechanism leading to tissue injury-associated persistent pain. Epigenetic events leading the altered transcription in the nervous system are pivotal in the maintenance of pain in tissue injury. However, the mechanisms through which those events contribute to the persistence of pain are not fully understood. This review provides a summary and critical evaluation of two epigenetic mechanisms, DNA methylation and non-coding RNA expression, on transcriptional modulation in nociceptive pathways during the development of tissue injury-associated pain. We assess the pre-clinical data and their translational implication and evaluate the potential of controlling DNA methylation and non-coding RNA expression as novel analgesic approaches and/or biomarkers of persistent pain.


Asunto(s)
Dolor Crónico/etiología , Metilación de ADN , Epigénesis Genética , ARN no Traducido , Heridas y Lesiones/complicaciones , Adaptación Biológica , Biomarcadores , Dolor Crónico/diagnóstico , Dolor Crónico/metabolismo , Dolor Crónico/terapia , Islas de CpG , Diagnóstico Diferencial , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos
8.
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
9.
Brain ; 138(Pt 7): 1843-62, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25981963

RESUMEN

Regeneration of injured central nervous system axons is highly restricted, causing neurological impairment. To date, although the lack of intrinsic regenerative potential is well described, a key regulatory molecular mechanism for the enhancement of both axonal regrowth and functional recovery after central nervous system injury remains elusive. While ubiquitin ligases coordinate neuronal morphogenesis and connectivity during development as well as after axonal injury, their role specifically in axonal regeneration is unknown. Following a bioinformatics network analysis combining ubiquitin ligases with previously defined axonal regenerative proteins, we found a triad composed of the ubiquitin ligases MDM4, MDM2 and the transcription factor p53 (encoded by TP53) as a putative central signalling complex restricting the regeneration program. Indeed, conditional deletion of MDM4 or pharmacological inhibition of MDM2/p53 interaction in the eye and spinal cord promote axonal regeneration and sprouting of the optic nerve after crush and of supraspinal tracts after spinal cord injury. The double conditional deletion of MDM4-p53 as well as MDM2 inhibition in p53-deficient mice blocks this regenerative phenotype, showing its dependence upon p53. Genome-wide gene expression analysis from ex vivo fluorescence-activated cell sorting in MDM4-deficient retinal ganglion cells identifies the downstream target IGF1R, whose activity and expression was found to be required for the regeneration elicited by MDM4 deletion. Importantly, we demonstrate that pharmacological enhancement of the MDM2/p53-IGF1R axis enhances axonal sprouting as well as functional recovery after spinal cord injury. Thus, our results show MDM4-MDM2/p53-IGF1R as an original regulatory mechanism for CNS regeneration and offer novel targets to enhance neurological recovery.media-1vid110.1093/brain/awv125_video_abstractawv125_video_abstract.


Asunto(s)
Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/metabolismo , Recuperación de la Función/fisiología , Transducción de Señal/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Axones/metabolismo , Axones/patología , Biología Computacional , Modelos Animales de Enfermedad , Citometría de Flujo , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Compresión Nerviosa , Traumatismos del Nervio Óptico/patología , Traumatismos del Nervio Óptico/fisiopatología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Transcriptoma , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
10.
J Neurosci ; 34(25): 8630-45, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24948817

RESUMEN

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


Asunto(s)
Ansiedad/metabolismo , Ansiedad/prevención & control , Factores de Transcripción NFATC/metabolismo , Neurogénesis/fisiología , Receptores de GABA-A/fisiología , Transducción de Señal/fisiología , Animales , Ansiedad/patología , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Proc Natl Acad Sci U S A ; 109(38): E2523-32, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22927399

RESUMEN

Aberrant mitochondrial function, morphology, and transport are main features of neurodegenerative diseases. To date, mitochondrial transport within neurons is thought to rely mainly on microtubules, whereas actin might mediate short-range movements and mitochondrial anchoring. Here, we analyzed the impact of actin on neuronal mitochondrial size and localization. F-actin enhanced mitochondrial size and mitochondrial number in neurites and growth cones. In contrast, raising G-actin resulted in mitochondrial fragmentation and decreased mitochondrial abundance. Cellular F-actin/G-actin levels also regulate serum response factor (SRF)-mediated gene regulation, suggesting a possible link between SRF and mitochondrial dynamics. Indeed, SRF-deficient neurons display neurodegenerative hallmarks of mitochondria, including disrupted morphology, fragmentation, and impaired mitochondrial motility, as well as ATP energy metabolism. Conversely, constitutively active SRF-VP16 induced formation of mitochondrial networks and rescued huntingtin (HTT)-impaired mitochondrial dynamics. Finally, SRF and actin dynamics are connected via the actin severing protein cofilin and its slingshot phosphatase to modulate neuronal mitochondrial dynamics. In summary, our data suggest that the SRF-cofilin-actin signaling axis modulates neuronal mitochondrial function.


Asunto(s)
Actinas/metabolismo , Cofilina 1/metabolismo , Mitocondrias/metabolismo , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Adenosina Trifosfato/metabolismo , Animales , Proteína Vmw65 de Virus del Herpes Simple/metabolismo , Hipocampo/metabolismo , Proteína Huntingtina , Ratones , Ratones Transgénicos , Microtúbulos/metabolismo , Modelos Biológicos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Distribución Tisular
12.
Proc Natl Acad Sci U S A ; 109(23): E1499-508, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22586092

RESUMEN

New neurons generated in the adult dentate gyrus are constantly integrated into the hippocampal circuitry and activated during encoding and recall of new memories. Despite identification of extracellular signals that regulate survival and integration of adult-born neurons such as neurotrophins and neurotransmitters, the nature of the intracellular modulators required to transduce those signals remains elusive. Here, we provide evidence of the expression and transcriptional activity of nuclear factor of activated T cell c4 (NFATc4) in hippocampal progenitor cells. We show that NFATc4 calcineurin-dependent activity is required selectively for survival of adult-born neurons in response to BDNF signaling. Indeed, cyclosporin A injection and stereotaxic delivery of the BDNF scavenger TrkB-Fc in the mouse dentate gyrus reduce the survival of hippocampal adult-born neurons in wild-type but not in NFATc4(-/-) mice and do not affect the net rate of neural precursor proliferation and their fate commitment. Furthermore, associated with the reduced survival of adult-born neurons, the absence of NFATc4 leads to selective defects in LTP and in the encoding of hippocampal-dependent spatial memories. Thus, our data demonstrate that NFATc4 is essential in the regulation of adult hippocampal neurogenesis and identify NFATc4 as a central player of BDNF-driven prosurvival signaling in hippocampal adult-born neurons.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Supervivencia Celular/fisiología , Hipocampo/citología , Memoria/fisiología , Factores de Transcripción NFATC/fisiología , Neuronas/fisiología , Percepción Espacial/fisiología , Análisis de Varianza , Animales , Western Blotting , Técnicas de Cultivo de Célula , Condicionamiento Psicológico/fisiología , Cartilla de ADN/genética , Potenciales Evocados/fisiología , Inmunohistoquímica , Luciferasas , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Neurosci ; 33(36): 14318-30, 2013 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-24005285

RESUMEN

Mounting evidence points to a role for endogenous reactive oxygen species (ROS) in cell signaling, including in the control of cell proliferation, differentiation, and fate. However, the function of ROS and their molecular regulation in embryonic mouse neural progenitor cells (eNPCs) has not yet been clarified. Here, we describe that physiological ROS are required for appropriate timing of neurogenesis in the developing telencephalon in vivo and in cultured NPCs, and that the tumor suppressor p53 plays a key role in the regulation of ROS-dependent neurogenesis. p53 loss of function leads to elevated ROS and early neurogenesis, while restoration of p53 and antioxidant treatment partially reverse the phenotype associated with premature neurogenesis. Furthermore, we describe that the expression of a number of neurogenic and oxidative stress genes relies on p53 and that both p53 and ROS-dependent induction of neurogenesis depend on PI3 kinase/phospho-Akt signaling. Our results suggest that p53 fine-tunes endogenous ROS levels to ensure the appropriate timing of neurogenesis in eNPCs. This may also have implications for the generation of tumors of neurodevelopmental origin.


Asunto(s)
Células-Madre Neurales/metabolismo , Neurogénesis , Fosfatidilinositol 3-Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Cultivadas , Ratones , Células-Madre Neurales/citología , Estrés Oxidativo/genética , Telencéfalo/citología , Telencéfalo/embriología , Telencéfalo/metabolismo , Proteína p53 Supresora de Tumor/genética
14.
J Neurosci ; 33(48): 18836-48, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24285890

RESUMEN

Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.


Asunto(s)
Factores Despolimerizantes de la Actina/fisiología , Axones/efectos de los fármacos , Citoplasma/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Factor de Respuesta Sérica/farmacología , Actinas/metabolismo , Animales , Axotomía , Citoplasma/efectos de los fármacos , Nervio Facial/fisiología , Femenino , Proteínas Fluorescentes Verdes , Masculino , Ratones , Nervios Periféricos/citología , Nervios Periféricos/efectos de los fármacos , Fosforilación , Reacción en Cadena de la Polimerasa , Fracciones Subcelulares/metabolismo
15.
Cell Mol Life Sci ; 70(6): 993-1007, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22899311

RESUMEN

In the last several years, relevant progress has been made in our understanding of the transcriptional machinery regulating CNS repair after acute injury, such as following trauma or stroke. In order to survive and functionally reconnect to the synaptic network, injured neurons activate an intrinsic rescue program aimed to increase their plasticity. Perhaps, in the attempt to switch back to a plastic and growth-competent state, post-mitotic neurons wake up and re-express a set of transcription factors that are also critical for the regulation of their younger brothers, the neural stem cells. Here, we review and discuss the transcriptional pathways regulating both axonal regeneration and neurogenesis highlighting the connection between the two. Clarification of their common molecular substrate may help simultaneous targeting of both neurogenesis and axonal regeneration with the hope to enhance functional recovery following CNS injury.


Asunto(s)
Axones/fisiología , Sistema Nervioso Central/citología , Regulación de la Expresión Génica/fisiología , Mamíferos/fisiología , Regeneración Nerviosa/fisiología , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Factores de Transcripción/metabolismo , Animales , Sistema Nervioso Central/lesiones , Humanos , Modelos Biológicos
16.
J Clin Med ; 13(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38337476

RESUMEN

BACKGROUND: With promising outcomes, platelet-rich plasma (PRP) has recently been suggested as a treatment for olfactory dysfunction (OD). METHODS: Clinical studies utilizing PRP in OD caused by COVID-19, trauma, anesthetic exposure, viral infection, and chronic rhinosinusitis were included in a systematic review. RESULTS: Ten clinical studies were qualitatively analyzed. Six of these studies used the PRP for OD caused by COVID-19, one on OD after functional endoscopic sinus surgery, and three on post-infectious or post-trauma OD. The population included 531 patients, ranging in age from 15 to 63. CONCLUSION: The use of PRP may be a risk-free and efficient therapeutic option with very encouraging outcomes. Indeed, it enhances olfactory perception in patients who not only exhibit COVID-19 infection aftereffects, but also in those who have lost their sense of smell due to trauma, rhinosinusitis, rhinitis, or even surgery. To evaluate the PRP's therapeutic benefits in OD patients and to compare the efficacy of different therapeutic protocols with regard to treatment schedules, there is an urgent need for focused controlled trials.

17.
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.

18.
J Neurosci ; 32(40): 13956-70, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23035104

RESUMEN

Following spinal trauma, the limited physiological axonal sprouting that contributes to partial recovery of function is dependent upon the intrinsic properties of neurons as well as the inhibitory glial environment. The transcription factor p53 is involved in DNA repair, cell cycle, cell survival, and axonal outgrowth, suggesting p53 as key modifier of axonal and glial responses influencing functional recovery following spinal injury. Indeed, in a spinal cord dorsal hemisection injury model, we observed a significant impairment in locomotor recovery in p53(-/-) versus wild-type mice. p53(-/-) spinal cords showed an increased number of activated microglia/macrophages and a larger scar at the lesion site. Loss- and gain-of-function experiments suggested p53 as a direct regulator of microglia/macrophages proliferation. At the axonal level, p53(-/-) mice showed a more pronounced dieback of the corticospinal tract (CST) and a decreased sprouting capacity of both CST and spinal serotoninergic fibers. In vivo expression of p53 in the sensorimotor cortex rescued and enhanced the sprouting potential of the CST in p53(-/-) mice, while, similarly, p53 expression in p53(-/-) cultured cortical neurons rescued a defect in neurite outgrowth, suggesting a direct role for p53 in regulating the intrinsic sprouting ability of CNS neurons. In conclusion, we show that p53 plays an important regulatory role at both extrinsic and intrinsic levels affecting the recovery of motor function following spinal cord injury. Therefore, we propose p53 as a novel potential multilevel therapeutic target for spinal cord injury.


Asunto(s)
Locomoción/fisiología , Neuronas/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Regeneración de la Medula Espinal/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Células Cultivadas , Cicatriz/patología , Cordotomía , Conducta Exploratoria/fisiología , Genes p53 , Calor , Cojera Animal/etiología , Cojera Animal/fisiopatología , Activación de Macrófagos , Masculino , Ratones , Ratones Noqueados , Microglía/patología , Plasticidad Neuronal/fisiología , Tractos Piramidales/patología , Recuperación de la Función , Degeneración Retrógrada , Umbral Sensorial , Neuronas Serotoninérgicas/fisiología , Traumatismos de la Médula Espinal/genética , Regeneración de la Medula Espinal/genética , Proteína p53 Supresora de Tumor/deficiencia
19.
Curr Urol Rep ; 14(5): 476-87, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23824516

RESUMEN

When sterile culture techniques of mammalian cells first became state of the art, there was tremendous anticipation that such cells could be eventually applied for therapeutic purposes. The discovery of adult human stem or progenitor cells further motivated scientists to pursue research in cell-based therapies. Although evidence from animal studies suggests that application of cells yields measurable benefits, in urology and many other disciplines, progenitor-cell-based therapies are not yet routinely clinically available. Stress urinary incontinence (SUI) is a condition affecting a large number of patients. The etiology of SUI includes, but is not limited to, degeneration of the urinary sphincter muscle tissue and loss of innervation, as well as anatomical and biomechanical causes. Therefore, different regimens were developed to treat SUI. However, at present, a curative functional treatment is not at hand. A progenitor-cell-based therapy that can tackle the etiology of incontinence, rather than the consequences, is a promising strategy. Therefore, several research teams have intensified their efforts to develop such a therapy for incontinence. Here, we introduce candidate stem and progenitor cells suitable for SUI treatment, show how the functional homogeneity and state of maturity of differentiated cells crucial for proper tissue integration can be assessed electrophysiologically prior to their clinical application, and discuss the trophic potential of adult mesenchymal stromal (or stem) cells in regeneration of neuronal function.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Recuperación de la Función , Vejiga Urinaria/fisiopatología , Incontinencia Urinaria de Esfuerzo/cirugía , Micción/fisiología , Animales , Humanos , Trasplante de Células Madre/métodos , Resultado del Tratamiento , Incontinencia Urinaria de Esfuerzo/fisiopatología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA