Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664711

RESUMEN

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Asunto(s)
Factor de Transcripción Activador 3 , Axones , Roturas del ADN de Doble Cadena , Ganglios Espinales , Células Madre Mesenquimatosas , Mitocondrias , Regeneración Nerviosa , Especies Reactivas de Oxígeno , Nervio Ciático , Regulación hacia Arriba , Animales , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Mitocondrias/metabolismo , Mitocondrias/genética , Especies Reactivas de Oxígeno/metabolismo , Axones/metabolismo , Regeneración Nerviosa/genética , Regulación hacia Arriba/genética , Ratones , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Nervio Ciático/lesiones , Nervio Ciático/patología , Ganglios Espinales/metabolismo , Ratones Endogámicos C57BL , Masculino
2.
Biol Pharm Bull ; 47(2): 366-372, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38325853

RESUMEN

Neuronal regrowth after traumatic injury is strongly inhibited in the central nervous system (CNS) of adult mammals. Cell-intrinsic and extrinsic factors limit the regulation of axonal growth and regrowth of fibers is minimal despite nearly all neurons surviving. Developing medical drugs to promote neurological recovery is crucial since neuronal injuries have few palliative cares and no pharmacological interventions. Herein, we developed a novel in vitro axonal regeneration assay system to screen the chemical reagents using human-induced pluripotent stem cell (hiPSC)-derived neurons. These neurons were cultured in a 96-well plate to form a monolayer and were scraped using a floating metal pin tool for axotomy. The cell number and plate coating conditions were optimized to score the regenerating axon. Treatment using the Rho-associated kinase (ROCK) inhibitor Y-27632 enhanced axonal regeneration in this regeneration assay system with hiPSC-derived neurons. Therefore, our novel screening method is suitable for drug screening to identify the chemical compounds that promote axonal regeneration after axotomy under in vitro conditions.


Asunto(s)
Axones , Células Madre Pluripotentes Inducidas , Humanos , Animales , Regeneración Nerviosa , Neuronas/fisiología , Sistema Nervioso Central , Mamíferos
3.
J Nanobiotechnology ; 22(1): 399, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970101

RESUMEN

Spinal cord injury (SCI) represents a profound central nervous system affliction, resulting in irreversibly compromised daily activities and disabilities. SCI involves excessive inflammatory responses, which are characterized by the existence of high levels of proinflammatory M1 macrophages, and neuronal mitochondrial energy deficit, exacerbating secondary damage and impeding axon regeneration. This study delves into the mechanistic intricacies of SCI, offering insights from the perspectives of neuroimmune regulation and mitochondrial function, leading to a pro-fibrotic macrophage phenotype and energy-supplying deficit. To address these challenges, we developed a smart scaffold incorporating enzyme mimicry nanoparticle-ceriumoxide (COPs) into nanofibers (NS@COP), which aims to pioneer a targeted neuroimmune repair strategy, rescuing CGRP receptor on macrophage and concurrently remodeling mitochondrial function. Our findings indicate that the integrated COPs restore the responsiveness of pro-inflammatory macrophages to calcitonin gene-related peptide (CGRP) signal by up-regulating receptor activity modifying protein 1 (RAMP1), a vital component of the CGRP receptor. This promotes macrophage fate commitment to an anti-inflammatory pro-resolution M2 phenotype, then alleviating glial scar formation. In addition, NS@COP implantation also protected neuronal mitochondrial function. Collectively, our results suggest that the strategy of integrating nanozyme COP nanoparticles into a nanofiber scaffold provides a promising therapeutic candidate for spinal cord trauma via rational regulation of neuroimmune communication and mitochondrial function.


Asunto(s)
Axones , Macrófagos , Nanofibras , Regeneración Nerviosa , Traumatismos de la Médula Espinal , Animales , Axones/metabolismo , Nanofibras/química , Regeneración Nerviosa/efectos de los fármacos , Ratones , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Ratas , Andamios del Tejido/química , Nanopartículas/química , Ratas Sprague-Dawley , Péptido Relacionado con Gen de Calcitonina/metabolismo , Femenino , Ratones Endogámicos C57BL
4.
J Neurophysiol ; 130(5): 1265-1281, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37820016

RESUMEN

After rostral spinal cord injury (SCI) of lampreys, the descending axons of injured (axotomized) reticulospinal (RS) neurons regenerate and locomotor function gradually recovers. Our previous studies indicated that relative to uninjured lamprey RS neurons, injured RS neurons display several dramatic changes in their biophysical properties, called the "injury phenotype." In the present study, at the onset of applied depolarizing current pulses for membrane potentials below as well as above threshold for action potentials (APs), injured RS neurons displayed a transient depolarization consisting of an initial depolarizing component followed by a delayed repolarizing component. In contrast, for uninjured neurons the transient depolarization was mostly only evident at suprathreshold voltages when APs were blocked. For injured RS neurons, the delayed repolarizing component resisted depolarization to threshold and made these neurons less excitable than uninjured RS neurons. After block of voltage-gated sodium and calcium channels for injured RS neurons, the transient depolarization was still present. After a further block of voltage-gated potassium channels, the delayed repolarizing component was abolished or significantly reduced, with little or no effect on the initial depolarizing component. Voltage-clamp experiments indicated that the delayed repolarizing component was due to a noninactivating outward-rectifying potassium channel whose conductance (gK) was significantly larger for injured RS neurons compared to that for uninjured neurons. Thus, SCI results in an increase in gK and other changes in the biophysical properties of injured lamprey RS neurons that lead to a reduction in excitability, which is proposed to create an intracellular environment that supports axonal regeneration.NEW & NOTEWORTHY After spinal cord injury (SCI), lamprey reticulospinal (RS) neurons responded to subthreshold depolarizing current pulses with a transient depolarization, which included an initial depolarization that was due to passive channels followed by a delayed repolarization that was mediated by voltage-gated potassium channels. The conductance of these channels (gK) was significantly increased for RS neurons after SCI and contributed to a reduction in excitability, which is expected to provide supportive conditions for subsequent axonal regeneration.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Traumatismos de la Médula Espinal , Animales , Canales de Potasio/fisiología , Neuronas/fisiología , Potenciales de la Membrana/fisiología , Lampreas , Médula Espinal
5.
J Transl Med ; 21(1): 511, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507810

RESUMEN

BACKGROUND: Preinjury of peripheral nerves triggers dorsal root ganglia (DRG) axon regeneration, a biological change that is more pronounced in young mice than in old mice, but the complex mechanism has not been clearly explained. Here, we aim to gain insight into the mechanisms of axon regeneration after conditioning lesion in different age groups of mice, thereby providing effective therapeutic targets for central nervous system (CNS) injury. METHODS: The microarray GSE58982 and GSE96051 were downloaded and analyzed to identify differentially expressed genes (DEGs). The protein-protein interaction (PPI) network, the miRNA-TF-target gene network, and the drug-hub gene network of conditioning lesion were constructed. The L4 and L5 DRGs, which were previously axotomized by the sciatic nerve conditioning lesions, were harvested for qRT-PCR. Furthermore, histological and behavioral tests were performed to assess the therapeutic effects of the candidate drug telmisartan in spinal cord injury (SCI). RESULTS: A total of 693 and 885 DEGs were screened in the old and young mice, respectively. Functional enrichment indicates that shared DEGs are involved in the inflammatory response, innate immune response, and ion transport. QRT-PCR results showed that in DRGs with preinjury of peripheral nerve, Timp1, P2ry6, Nckap1l, Csf1, Ccl9, Anxa1, and C3 were upregulated, while Agtr1a was downregulated. Based on the bioinformatics analysis of DRG after conditioning lesion, Agtr1a was selected as a potential therapeutic target for the SCI treatment. In vivo experiments showed that telmisartan promoted axonal regeneration after SCI by downregulating AGTR1 expression. CONCLUSION: This study provides a comprehensive map of transcriptional changes that discriminate between young and old DRGs in response to injury. The hub genes and their related drugs that may affect the axonal regeneration program after conditioning lesion were identified. These findings revealed the speculative pathogenic mechanism involved in conditioning-dependent regenerative growth and may have translational significance for the development of CNS injury treatment in the future.


Asunto(s)
MicroARNs , Traumatismos de la Médula Espinal , Ratones , Animales , Axones/metabolismo , Axones/patología , Regeneración Nerviosa/genética , Telmisartán/metabolismo , Telmisartán/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Médula Espinal
6.
Int J Mol Sci ; 24(16)2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37629041

RESUMEN

One of the most prevalent causes of olfactory loss includes traumatic brain injury with subsequent shearing of olfactory axons at the level of the cribriform plate (anterior skull base). Scar tissue at this level may prevent axonal regrowth toward the olfactory bulb. Currently, there is no cure for this debilitating and often permanent condition. One promising therapeutic concept is to implant a synthetic scaffold with growth factors through the cribriform plate/scar tissue to induce neuroregeneration. The first step toward this goal is to investigate the optimum conditions (growth factors, extracellular matrix proteins) to boost this regeneration. However, the lack of a specifically tailored in vitro model and an automated procedure for quantifying axonal length limits our ability to address this issue. The aim of this study is to create an automated quantification tool to measure axonal length and to determine the ideal growth factors and extracellular proteins to enhance axonal regrowth of olfactory sensory neurons in a mouse organotypic 2D model. We harvested olfactory epithelium (OE) of C57BL/6 mice and cultured them during 15 days on coverslips coated with various extracellular matrix proteins (Fibronectin, Collagen IV, Laminin, none) and different growth factors: fibroblast growth factor 2 (FGF2), brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), retinoic acid (RA), transforming growth factor ß (TGFß), and none. We measured the attachment rate on coverslips, the presence of cellular and axonal outgrowth, and finally, the total axonal length with a newly developed automated high-throughput quantification tool. Whereas the coatings did not influence attachment and neuronal outgrowth rates, the total axonal length was enhanced on fibronectin and collagen IV (p = 0.001). The optimum growth factor supplementation media to culture OE compared to the control condition were as follows: FGF2 alone and FGF2 from day 0 to 7 followed by FGF2 in combination with NGF from day 7 to 15 (p < 0.0001). The automated quantification tool to measure axonal length outperformed the standard Neuron J application by reducing the average analysis time from 22 to 3 min per specimen. In conclusion, robust regeneration of murine olfactory neurons in vitro can be induced, controlled, and efficiently measured using an automated quantification tool. These results will help advance the therapeutic concept closer toward preclinical studies.


Asunto(s)
Neuronas Receptoras Olfatorias , Animales , Ratones , Ratones Endogámicos C57BL , Fibronectinas , Cicatriz , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor de Crecimiento Nervioso , Axones , Proteínas de la Matriz Extracelular , Colágeno Tipo IV , Medios de Cultivo
7.
J Cell Physiol ; 237(3): 1818-1832, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34825376

RESUMEN

Even though astrocytes have been widely reported to support several brain functions, studies have emerged that they exert deleterious effects on the brain after ischemia and reperfusion (I/R) injury. The present study investigated the neuroprotective effects of melatonin on the processes of reactive astrogliosis and glial scar formation, as well as axonal regeneration after transient middle cerebral artery occlusion. Male Wistar rats were randomly divided into four groups: sham-operated, I/R, I/R treated with melatonin, and I/R treated with edaravone. All drugs were administered via intraperitoneal injection at the onset of reperfusion and were continued until the rats were sacrificed on Day 7 or 14 after the surgery. Melatonin presented long-term benefits on cerebral damage after I/R injury, as demonstrated by a decreased infarct volume, histopathological changes, and reduced neuronal cell death. We also found that melatonin attenuated reactive astrogliosis and glial scar formation and, consequently, enhanced axonal regeneration and promoted neurobehavioral recovery. Furthermore, glycogen synthase kinase-3 beta (GSK-3ß) and receptor-interacting serine/threonine-protein 1 kinase (RIP1K), which had previously been revealed as proteins involved in astrocyte responses, were significantly reduced after melatonin administration. Taken together, melatonin effectively counteracted the deleterious effects due to astrocyte responses and improved axonal regeneration to promote functional recovery during the chronic phase of cerebral I/R injury by inhibiting GSK-3ß and RIP1K activities.


Asunto(s)
Isquemia Encefálica , Melatonina , Fármacos Neuroprotectores , Daño por Reperfusión , Animales , Isquemia Encefálica/metabolismo , Gliosis/tratamiento farmacológico , Gliosis/patología , Glucógeno Sintasa Quinasa 3 beta , Inflamación , Masculino , Melatonina/farmacología , Melatonina/uso terapéutico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Wistar , Daño por Reperfusión/patología
8.
Glia ; 70(1): 20-34, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34519370

RESUMEN

The phenotypic plasticity of Schwann cells (SCs) has contributed to the regenerative potential of the peripheral nervous system (PNS), but also pathological processes. This double-sided effect has led to an increasing attention to the role of extracellular vesicles (EVs) or exosomes in SCs to examine the intercellular communication between SCs and their surroundings. Here, we first describe the current knowledge of SC and EV biology, which forms the basis for the updates on advances in SC-derived exosomes research. We seek to explore in-depth the exosome-mediated molecular mechanisms involved in the regulation of SCs and their microenvironment. This review concludes with potential applications of SC-derived exosomes as delivery vehicles for therapeutics and biomarkers. The goal of this review is to emphasize the crucial role of SC-derived exosomes in the functional integration of the PNS, highlighting an emerging area in which there is much to explore and re-explore.


Asunto(s)
Exosomas , Vesículas Extracelulares , Comunicación Celular , Regeneración Nerviosa/fisiología , Células de Schwann/patología
9.
Neurobiol Dis ; 172: 105811, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35809764

RESUMEN

Glaucoma is an optic neuropathy and is currently one of the most common diseases that leads to irreversible blindness. The axonal degeneration that occurs before retinal ganglion neuronal loss is suggested to be involved in the pathogenesis of glaucoma. G protein-coupled receptor 3 (GPR3) belongs to the class A rhodopsin-type GPCR family and is highly expressed in various neurons. GPR3 is unique in its ability to constitutively activate the Gαs protein without a ligand, which elevates the basal intracellular cAMP level. Our earlier reports suggested that GPR3 enhances both neurite outgrowth and neuronal survival. However, the potential role of GPR3 in axonal regeneration after neuronal injury has not been elucidated. Herein, we investigated retinal GPR3 expression and its possible involvement in axonal regeneration after retinal injury in mice. GPR3 was relatively highly expressed in retinal ganglion cells (RGCs). Surprisingly, RGCs in GPR3 knockout mice were vulnerable to neural death during aging without affecting high intraocular pressure (IOP) and under ischemic conditions. Primary cultured neurons from the retina showed that GPR3 expression was correlated with neurite outgrowth and neuronal survival. Evaluation of the effect of GPR3 on axonal regeneration using GPR3 knockout mice revealed that GPR3 in RGCs participates in axonal regeneration after optic nerve crush (ONC) under zymosan stimulation. In addition, regenerating axons were further stimulated when GPR3 was upregulated in RGCs, and the effect was further augmented when combined with zymosan treatment. These results suggest that GPR3 expression in RGCs helps maintain neuronal survival and accelerates axonal regeneration after ONC in mice.


Asunto(s)
Glaucoma , Traumatismos del Nervio Óptico , Animales , Axones/patología , Glaucoma/metabolismo , Ratones , Ratones Noqueados , Compresión Nerviosa , Regeneración Nerviosa/fisiología , Nervio Óptico , Traumatismos del Nervio Óptico/patología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Ganglionares de la Retina/metabolismo , Zimosan/metabolismo , Zimosan/farmacología
10.
J Cell Sci ; 133(12)2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32409566

RESUMEN

Functional recovery after peripheral nerve damage is dependent on the reprogramming of differentiated Schwann cells (dSCs) into repair Schwann cells (rSCs), which promotes axonal regeneration and tissue homeostasis. Transition into a repair phenotype requires expression of c-Jun and Sox2, which transcriptionally mediates inhibition of the dSC program of myelination and activates a non-cell-autonomous repair program, characterized by the secretion of neuronal survival and regenerative molecules, formation of a cellular scaffold to guide regenerating axons and activation of an innate immune response. Moreover, rSCs release exosomes that are internalized by peripheral neurons, promoting axonal regeneration. Here, we demonstrate that reprogramming of Schwann cells (SCs) is accompanied by a shift in the capacity of their secreted exosomes to promote neurite growth, which is dependent on the expression of c-Jun (also known as Jun) and Sox2 by rSCs. Furthermore, increased expression of miRNA-21 is responsible for the pro-regenerative capacity of rSC exosomes, which is associated with PTEN downregulation and PI3-kinase activation in neurons. We propose that modification of exosomal cargo constitutes another important feature of the repair program of SCs, contributing to axonal regeneration and functional recovery after nerve injury.


Asunto(s)
Exosomas , MicroARNs , Axones , Reprogramación Celular , Exosomas/genética , MicroARNs/genética , Regeneración Nerviosa/genética , Células de Schwann
11.
Biochem Biophys Res Commun ; 607: 158-165, 2022 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-35367829

RESUMEN

Schwann cells play an important role in peripheral myelination, and dysfunction of these cells leads to axonal damage. Schwann cells degenerate following peripheral nerve injury. Immature Schwann cells proliferate, differentiate, and support axonal regeneration and extension during recovery. There are a lot of intracellular signals involved in the myelination process. Although serum- and glucocorticoid-inducible kinase (SGK1) in Schwann cells is supposedly involved in developmental myelination, its significance during peripheral nerve injury and repair remains unknown. In this study, we examined the dynamics of SGK1 during peripheral nerve repair and the potential role of SGK in the process. Axonal crush injury was first generated in the right sciatic nerve under anesthesia in mice, which exhibited apparent paralysis and subsequent recovery of the injured hindlimbs. Immunohistochemical analysis revealed the appearance of glial fibrillary acidic protein (GFAP)-positive immature Schwann cells around injured nerves, and SGK1 was present in these cells. Next, we employed S16 cells, a Schwann cell line, to explore the impact of SGK1 on Schwann cells. Administration of the SGK inhibitor gsk650394 decreased cell proliferation and increased cell size. SGK inhibition did not cause cellular injury, suggesting that it suppresses proliferation and enlarges Schwann cells without causing cell death. Furthermore, quantitative PCR and immunoblotting revealed that SGK inhibition upregulated the gene expression of BDNF, MBP, and Krox20, which are facilitating factors for myelination and neural regeneration, and downregulated that of Sox10. Taken together, these findings indicate that SGK1 inactivation in Schwann cells diverts cell fate from proliferation to differentiation.


Asunto(s)
Traumatismos de los Nervios Periféricos , Animales , Axones/metabolismo , Ratones , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Células de Schwann/metabolismo , Nervio Ciático/metabolismo
12.
J Neurosci Res ; 100(11): 2044-2054, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35986577

RESUMEN

Human spinal cord injury (SCI) is characterized by permanent loss of damaged axons, resulting in chronic disability. In contrast, zebrafish can regenerate axonal projections following central nervous system injury and re-establish synaptic contacts with distant targets; elucidation of the underlying molecular events is an important goal with translational potential for improving outcomes in SCI patients. We generated transgenic zebrafish with GFP-labeled axons and transected their spinal cords at 10 days post-fertilization. Intravital confocal microscopy revealed robust axonal regeneration following the procedure, with abundant axons bridging the transection site by 48 h post-injury. In order to analyze neurological function in this model, we developed and validated new open-source software to measure zebrafish lateral trunk curvature during propulsive and turning movements at high temporal resolution. Immediately following spinal cord transection, axial movements were dramatically decreased caudal to the lesion site, but preserved rostral to the injury, suggesting the induction of motor paralysis below the transection level. Over the subsequent 96 h, the magnitude of movements caudal to the lesion recovered to baseline, but the rate of change of truncal curvature did not fully recover, suggesting incomplete restoration of caudal strength over this time course. Quantification of both morphological and functional recovery following SCI will be important for the analysis of axonal regeneration and downstream events necessary for restoration of motor function. An extensive array of genetic and pharmacological interventions can be deployed in the larval zebrafish model to investigate the underlying molecular mechanisms.


Asunto(s)
Traumatismos de la Médula Espinal , Pez Cebra , Animales , Axones/patología , Humanos , Larva , Regeneración Nerviosa/fisiología , Recuperación de la Función/fisiología , Médula Espinal/patología , Traumatismos de la Médula Espinal/patología
13.
RNA ; 26(11): 1637-1653, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32747606

RESUMEN

Programmed cell death 4 (PDCD4) protein is a tumor suppressor that inhibits translation through the mTOR-dependent initiation factor EIF4A, but its functional role and mRNA targets in neurons remain largely unknown. Our work identified that PDCD4 is highly expressed in axons and dendrites of CNS and PNS neurons. Using loss- and gain-of-function experiments in cortical and dorsal root ganglia primary neurons, we demonstrated the capacity of PDCD4 to negatively control axonal growth. To explore PDCD4 transcriptome and translatome targets, we used Ribo-seq and uncovered a list of potential targets with known functions as axon/neurite outgrowth regulators. In addition, we observed that PDCD4 can be locally synthesized in adult axons in vivo, and its levels decrease at the site of peripheral nerve injury and before nerve regeneration. Overall, our findings demonstrate that PDCD4 can act as a new regulator of axonal growth via the selective control of translation, providing a target mechanism for axon regeneration and neuronal plasticity processes in neurons.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Axones/metabolismo , Dendritas/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Células Cultivadas , Mutación con Ganancia de Función , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Mutación con Pérdida de Función , Masculino , Ratones , Células PC12 , Cultivo Primario de Células , Biosíntesis de Proteínas , Proteínas de Unión al ARN/genética , Ratas , Regulación hacia Arriba
14.
J Surg Res ; 280: 379-388, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36037615

RESUMEN

INTRODUCTION: Two-stage free functional muscle transfers for long-standing facial palsy can yield unpredictable results. Earlier studies have demonstrated incomplete regeneration across neurorrhaphies in native nerve and higher donor axonal counts correlating with improved outcomes but axonal count in nerve grafts have not been as thoroughly reviewed. To investigate the impact of varying axonal counts in autologous grafts on functional outcomes of repair. MATERIALS AND METHODS: Animals were allocated into three groups: Direct Nerve Repair (DNR, n = 50), Small Nerve Graft (SNG, n = 50), and Large Nerve Graft (LNG, n = 50). All grafts were inset into the Posterior Auricular Nerve with ear movement recovery (EMR) monitored as functional outcome. At various postoperative weeks (POWs), excised specimens were imaged with electron microscopy. Axonal counts were measured proximal to, distal (DAC) to, and within grafts. Total Success Ratio (TSR) was calculated. RESULTS: In DNR, DAC was significantly lower than proximal axonal counts at all POWs, with maximum TSR of 80%. TSR for LNG and SNG were significantly lower at all POWs when compared to DNR, with maximums of 56% and 38%, respectively. LNG had a significantly larger DAC than SNG at POW12 and beyond. A direct relationship was present between DAC and EMR for all values. CONCLUSIONS: Higher native axonal count of autologous nerve grafts resulted in higher percentage of regeneration across neurorrhaphies.


Asunto(s)
Parálisis Facial , Regeneración Nerviosa , Animales , Axones/fisiología , Nervio Facial , Procedimientos Neuroquirúrgicos/métodos
15.
Int J Mol Sci ; 24(1)2022 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-36613945

RESUMEN

Spinal cord injuries result in the loss of motor and sensory functions controlled by neurons located at the site of the lesion and below. We hypothesized that experimentally enhanced remyelination supports axon preservation and/or growth in the total spinal cord transection in rats. Multifocal demyelination was induced by injection of ethidium bromide (EB), either at the time of transection or twice during transection and at 5 days post-injury. We demonstrated that the number of oligodendrocyte progenitor cells (OPCs) significantly increased 14 days after demyelination. Most OPCs differentiated into mature oligodendrocytes by 60-90 dpi in double-EB-injected rats; however, most axons were remyelinated by Schwann cells. A significant number of axons passed the injury epicenter and entered the distant segments of the spinal cord in the double-EB-injected rats. Moreover, some serotoninergic fibers, not detected in control animals, grew caudally through the injury site. Behavioral tests performed at 60-90 dpi revealed significant improvement in locomotor function recovery in double-EB-injected rats, which was impaired by the blockade of serotonin receptors, confirming the important role of restored serotonergic fibers in functional recovery. Our findings indicate that enhanced remyelination per se, without substantial inhibition of glial scar formation, is an important component of spinal cord injury regeneration.


Asunto(s)
Enfermedades Desmielinizantes , Remielinización , Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Ratas , Animales , Axones/patología , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/patología , Médula Espinal/patología , Etidio , Enfermedades Desmielinizantes/patología
16.
Int J Mol Sci ; 23(24)2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36555556

RESUMEN

The existing studies by our team demonstrated the pro-recovery effect of 3-Acetyl-11-keto-beta-boswellic acid (AKBA) on a sciatic nerve injury. To further investigate the role of AKBA in peripheral nerve injury repair, The TMT quantitative proteomics technique was used to obtain differentially significant proteins in a Sham group, Model group, and AKBA group. After that, three time points (5, 14, and 28 d) and four groups (Sham + AKBA, Sham, Model, and AKBA) were set up, and immunoblotting, immunofluorescence, and cellular assays were applied to investigate the expression of CDC42, Rac1, RhoA, and Rictor in the sciatic nerve at different time points for each group in more depth. The results showed that AKBA enriched the cellular components of the myelin sheath and axon regeneration after a sciatic nerve injury and that AKBA upregulated CDC42 and Rac1 and downregulated RhoA expression 5 d after a sciatic nerve injury, promoting axon regeneration and improving the repair of a sciatic nerve injury in rats. Rictor is regulated by AKBA and upregulated in PC12 cells after AKBA action. Our findings provide a new basis for AKBA treatment of a peripheral nerve injury.


Asunto(s)
Traumatismos de los Nervios Periféricos , Neuropatía Ciática , Animales , Ratas , Axones , Regeneración Nerviosa/fisiología , Proteína Asociada al mTOR Insensible a la Rapamicina , Nervio Ciático/lesiones , Neuropatía Ciática/tratamiento farmacológico
17.
Arch Ital Biol ; 159(3-4): 159-177, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35077573

RESUMEN

Due to the complexity of pathological processes in spinal cord injury (SCI), it is increasingly recognized that combined strategies are more effective than single treatments. The aim of the present study was to enhance neural tissue regeneration and axon regrowth using Rho-A-kinase inhibitor (Y-27632) in a rat SCI model (Th9 compression) and to bridge the lesion with a chitosan/collagen porous scaffold (ChC-PS) applied two weeks after SCI. In addition, to see the synergic effect of Y-27632 and ChC-PS, we combined these single therapeutic strategies to enhance the regenerative capacity of injured spinal cord tissue. The animals survived eight weeks. Application of Y-27632 modulated the inhibitory milieu by specifically targeting gray and white matter integrity, glial fibrillary acidic protein (GFAP)-immunoreactivity, and the outgrowth of neurofilaments and growth-associated protein-43 (GAP-43) immunoreactive axons across the lesion sites, leading to significant positive functional outcome from day 20 to 56. Compared to single treatments, combined Y-27632/ChC-PS therapy was more effective in neurofilaments and GAP-43 expression and GFAP immunoreactivity in the perilesional area of dorsal, lateral and ventral columns, and in enhancing the gray and white matter integrity throughout the cranio-caudal extent. The findings indicate that combined therapy provides a supportive environment for endogenous regenerative processes.


Asunto(s)
Quitosano , Traumatismos de la Médula Espinal , Animales , Colágeno , Regeneración Nerviosa , Porosidad , Ratas , Médula Espinal , Traumatismos de la Médula Espinal/tratamiento farmacológico
18.
Medicina (Kaunas) ; 58(4)2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35454380

RESUMEN

Unlike peripheral nerves, axonal regeneration is limited following injury to the spinal cord. While there may be reduced regenerative potential of injured neurons, the central nervous system (CNS) white matter environment appears to be more significant in limiting regrowth. Several factors may inhibit regeneration, and their neutralization can modestly enhance regrowth. However, most investigations have not considered the cytoarchitecture of spinal cord white matter. Several lines of investigation demonstrate that axonal regeneration is enhanced by maintaining, repairing, or reconstituting the parallel geometry of the spinal cord white matter. In this review, we focus on environmental factors that have been implicated as putative inhibitors of axonal regeneration and the evidence that their organization may be an important determinant in whether they inhibit or promote regeneration. Consideration of tissue geometry may be important for developing successful strategies to promote spinal cord regeneration.


Asunto(s)
Traumatismos de la Médula Espinal , Regeneración de la Medula Espinal , Axones/fisiología , Humanos , Regeneración Nerviosa/fisiología , Neuronas/fisiología , Médula Espinal , Traumatismos de la Médula Espinal/terapia
19.
Glia ; 69(6): 1444-1463, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33502042

RESUMEN

Neurodegenerative disorders, characterized by progressive neuronal loss, eventually lead to functional impairment in the adult mammalian central nervous system (CNS). Importantly, these deteriorations are irreversible, due to the very limited regenerative potential of these CNS neurons. Stimulating and redirecting neuroinflammation was recently put forward as an important approach to induce axonal regeneration, but it remains elusive how inflammatory processes and CNS repair are intertwined. To gain more insight into these interactions, we investigated how immunomodulation affects the regenerative outcome after optic nerve crush (ONC) in the spontaneously regenerating zebrafish. First, inducing intraocular inflammation using zymosan resulted in an acute inflammatory response, characterized by an increased infiltration and proliferation of innate blood-borne immune cells, reactivation of Müller glia, and altered retinal cytokine expression. Strikingly, inflammatory stimulation also accelerated axonal regrowth after optic nerve injury. Second, we demonstrated that acute depletion of both microglia and macrophages in the retina, using pharmacological treatments with both the CSF1R inhibitor PLX3397 and clodronate liposomes, compromised optic nerve regeneration. Moreover, we observed that csf1ra/b double mutant fish, lacking microglia in both retina and brain, displayed accelerated RGC axonal regrowth after ONC, which was accompanied with unusual Müller glia proliferative gliosis. Altogether, our results highlight the importance of altered glial cell interactions in the axonal regeneration process after ONC in adult zebrafish. Unraveling the relative contribution of the different cell types, as well as the signaling pathways involved, may pinpoint new targets to stimulate repair in the vertebrate CNS.


Asunto(s)
Regeneración Nerviosa , Pez Cebra , Animales , Macrófagos , Neuroglía , Enfermedades Neuroinflamatorias , Retina
20.
Neurobiol Dis ; 150: 105259, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33434618

RESUMEN

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/farmacología , Regeneración Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Nervio Óptico/efectos de los fármacos , Animales , Anticolesterolemiantes/farmacología , Axones/efectos de los fármacos , Axones/patología , Supervivencia Celular , Embrión de Pollo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Vaina de Mielina , Neuronas/metabolismo , Nervio Óptico/metabolismo , Nervio Óptico/patología , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/patología , Células Fotorreceptoras , Prenilación , Profármacos , Ratas , Retina , Retinitis Pigmentosa , Diclorhidrato de trans-1,4-Bis(2-clorobenzaminometil)ciclohexano/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA