Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Neurobiol Dis ; 184: 106190, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37290578

RESUMEN

Embryonic and early postnatal deletion of the gene phosphatase and tensin homolog (PTEN) results in neuronal hypertrophy, formation of aberrant neural networks and spontaneous seizures. Our previous studies document that deletion of PTEN in mature neurons also causes growth of cortical neuron cell bodies and dendrites, but it is unknown how this growth alters connectivity in mature circuits. Here, we explore consequences of deleting PTEN in a focal area of the dentate gyrus in adult male and female mice. PTEN deletion was accomplished by injecting AAV-Cre unilaterally into the dentate gyrus of double transgenic mice with lox-P sites flanking exon 5 of the PTEN gene and stop/flox tdTomato in the Rosa locus (PTENf/f/RosatdTomato). Focal deletion led to progressive increases in the size of the dentate gyrus at the injection site, enlargement of granule cell bodies, and increases in dendritic length and caliber. Quantitative analysis of dendrites by Golgi staining revealed dramatic increases in spine numbers throughout the proximo-distal extent of the dendritic tree, suggesting that dendritic growth is sufficient to induce new synapse formation by input neurons with intact PTEN expression. Tract tracing of input pathways to the dentate gyrus from the ipsilateral entorhinal cortex and commissural/associational system revealed that laminar specificity of termination of inputs is maintained. Mossy fiber axons from PTEN-deleted granule cells expanded their terminal field in CA3 where PTEN expression was intact and supra-granular mossy fibers developed in some mice. These findings document that persistent activation of mTOR via PTEN deletion in fully mature neurons re-initiates a state of robust cell-intrinsic growth, upending connectional homeostasis in fully mature hippocampal circuits.


Asunto(s)
Cuerpo Celular , Fibras Musgosas del Hipocampo , Ratones , Animales , Fibras Musgosas del Hipocampo/fisiología , Hipocampo/fisiología , Ratones Transgénicos , Dendritas , Giro Dentado
3.
J Neurosci ; 41(37): 7712-7726, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34326146

RESUMEN

Injury responses require communication between different cell types in the skin. Sensory neurons contribute to inflammation and can secrete signaling molecules that affect non-neuronal cells. Despite the pervasive role of translational regulation in nociception, the contribution of activity-dependent protein synthesis to inflammation is not well understood. To address this problem, we examined the landscape of nascent translation in murine dorsal root ganglion (DRG) neurons treated with inflammatory mediators using ribosome profiling. We identified the activity-dependent gene, Arc, as a target of translation in vitro and in vivo Inflammatory cues promote local translation of Arc in the skin. Arc-deficient male mice display exaggerated paw temperatures and vasodilation in response to an inflammatory challenge. Since Arc has recently been shown to be released from neurons in extracellular vesicles (EVs), we hypothesized that intercellular Arc signaling regulates the inflammatory response in skin. We found that the excessive thermal responses and vasodilation observed in Arc defective mice are rescued by injection of Arc-containing EVs into the skin. Our findings suggest that activity-dependent production of Arc in afferent fibers regulates neurogenic inflammation potentially through intercellular signaling.SIGNIFICANCE STATEMENT Nociceptors play prominent roles in pain and inflammation. We examined rapid changes in the landscape of nascent translation in cultured dorsal root ganglia (DRGs) treated with a combination of inflammatory mediators using ribosome profiling. We identified several hundred transcripts subject to rapid preferential translation. Among them is the immediate early gene (IEG) Arc. We provide evidence that Arc is translated in afferent fibers in the skin. Arc-deficient mice display several signs of exaggerated inflammation which is normalized on injection of Arc containing extracellular vesicles (EVs). Our work suggests that noxious cues can trigger Arc production by nociceptors which in turn constrains neurogenic inflammation in the skin.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Ganglios Espinales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Vasodilatación/fisiología , Animales , Proteínas del Citoesqueleto/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Nocicepción/fisiología , Nociceptores/fisiología , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/fisiopatología
4.
Cereb Cortex ; 31(5): 2322-2344, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33350438

RESUMEN

Rostro-caudal specificity of corticospinal tract (CST) projections from different areas of the cortex was assessed by retrograde labeling with fluorogold and retrograde transfection following retro-AAV/Cre injection into the spinal cord of tdT reporter mice. Injections at C5 led to retrograde labeling of neurons throughout forelimb area of the sensorimotor cortex and a region in the dorsolateral cortex near the barrel field (S2). Injections at L2 led to retrograde labeling of neurons in the posterior sensorimotor cortex (hindlimb area) but not the dorsolateral cortex. With injections of biotinylated dextran amine (BDA) into the main sensorimotor cortex (forelimb region), labeled axons terminated selectively at cervical levels. With BDA injections into caudal sensorimotor cortex (hindlimb region), labeled axons passed through cervical levels without sending collaterals into the gray matter and then elaborated terminal arbors at thoracic sacral levels. With BDA injections into the dorsolateral cortex near the barrel field, labeled axons terminated at high cervical levels. Axons from medial sensorimotor cortex terminated primarily in intermediate laminae and axons from lateral sensorimotor cortex terminated primarily in laminae III-V of the dorsal horn. One of the descending pathways seen in rats (the ventral CST) was not observed in most mice.


Asunto(s)
Corteza Motora/fisiología , Neuronas/patología , Tractos Piramidales/fisiología , Médula Espinal/fisiología , Animales , Axones/fisiología , Miembro Posterior/patología , Miembro Posterior/fisiología , Masculino , Ratones Endogámicos BALB C , Corteza Motora/patología , Neuronas/fisiología , Tractos Piramidales/patología , Médula Espinal/patología
5.
Neurobiol Dis ; 140: 104868, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32276110

RESUMEN

Multiple sclerosis (MS) is a chronic, inflammatory autoimmune disease that affects the central nervous system (CNS) for which there is no cure. In MS, encephalitogenic T cells infiltrate the CNS causing demyelination and neuroinflammation; however, little is known about the role of regulatory T cells (Tregs) in CNS tissue repair. Transplantation of neural stem and progenitor cells (NSCs and NPCs) is a promising therapeutic strategy to promote repair through cell replacement, although recent findings suggest transplanted NSCs also instruct endogenous repair mechanisms. We have recently described that dampened neuroinflammation and increased remyelination is correlated with emergence of Tregs following human NPC transplantation in a murine viral model of immune-mediated demyelination. In the current study we utilized the prototypic murine autoimmune model of demyelination experimental autoimmune encephalomyelitis (EAE) to test the efficacy of hNSC transplantation. Eight-week-old, male EAE mice receiving an intraspinal transplant of hNSCs during the chronic phase of disease displayed remyelination, dampened neuroinflammation, and an increase in CNS CD4+CD25+FoxP3+ regulatory T cells (Tregs). Importantly, ablation of Tregs abrogated histopathological improvement. Tregs are essential for maintenance of T cell homeostasis and prevention of autoimmunity, and an emerging role for Tregs in maintenance of tissue homeostasis through interactions with stem and progenitor cells has recently been suggested. The data presented here provide direct evidence for collaboration between CNS Tregs and hNSCs promoting remyelination.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple/terapia , Células-Madre Neurales/trasplante , Remielinización , Linfocitos T Reguladores , Animales , Humanos , Masculino , Ratones , Vaina de Mielina , Trasplante de Células Madre
6.
Muscle Nerve ; 61(3): 390-395, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31820462

RESUMEN

INTRODUCTION: After traumatic nerve injury, neuromuscular junction remodeling plays a key role in determining functional outcomes. Immunohistochemical analyses of denervated muscle biopsies may provide valuable prognostic data regarding clinical outcomes to supplement electrodiagnostic studies. METHODS: We performed biopsies on nonfunctioning deltoid muscles in two patients after gunshot wounds and visualized the neuromuscular junctions using two-photon microscopy with immunohistochemistry. RESULTS: Although the nerves in both patients showed evidence of acute Wallerian degeneration, some of the motor endplates were intact but exhibited significantly decreased surface area and volume. Both patients exhibited substantial recovery of motor function over several weeks postinjury. DISCUSSION: Two-photon microscopic assessment of neuromuscular junction integrity and motor endplate morphometry in muscle biopsies provided evidence of partial sparing of muscle innervation. This finding supported the clinical judgment that eventual recovery would occur. With further study, this technique may help to guide operative decisionmaking after traumatic nerve injuries.


Asunto(s)
Neuropatías del Plexo Braquial/diagnóstico , Neuropatías del Plexo Braquial/patología , Placa Motora/patología , Adulto , Neuropatías del Plexo Braquial/fisiopatología , Músculo Deltoides/inervación , Músculo Deltoides/patología , Electromiografía , Humanos , Masculino , Microscopía , Placa Motora/fisiología , Conducción Nerviosa , Imagen Óptica , Adulto Joven
7.
J Neurosci ; 42(50): 9296-9297, 2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36517249
8.
Nature ; 490(7419): 187-91, 2012 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23060188

RESUMEN

The US National Institute of Neurological Disorders and Stroke convened major stakeholders in June 2012 to discuss how to improve the methodological reporting of animal studies in grant applications and publications. The main workshop recommendation is that at a minimum studies should report on sample-size estimation, whether and how animals were randomized, whether investigators were blind to the treatment, and the handling of data. We recognize that achieving a meaningful improvement in the quality of reporting will require a concerted effort by investigators, reviewers, funding agencies and journal editors. Requiring better reporting of animal studies will raise awareness of the importance of rigorous study design to accelerate scientific progress.


Asunto(s)
Edición/normas , Proyectos de Investigación/normas , Animales , Edición/tendencias , Distribución Aleatoria , Tamaño de la Muestra , Estadística como Asunto
9.
Learn Mem ; 24(8): 341-357, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28716954

RESUMEN

High-frequency stimulation of the medial perforant path triggers robust phosphorylation of ribosomal protein S6 (rpS6) in activated dendritic domains and granule cell bodies. Here we dissect the signaling pathways responsible for synaptically driven rpS6 phosphorylation in the dentate gyrus using pharmacological agents to inhibit PI3-kinase/mTOR and MAPK/ERK-dependent kinases. Using phospho-specific antibodies for rpS6 at different sites (ser235/236 versus ser240/244), we show that delivery of the PI3-kinase inhibitor, wortmannin, decreased rpS6 phosphorylation throughout the somatodendritic compartment (granule cell layer, inner molecular layer, outer molecular layer), especially in granule cell bodies while sparing phosphorylation at activated synapses (middle molecular layer). In contrast, delivery of U0126, an MEK inhibitor, attenuated rpS6 phosphorylation specifically in the dendritic laminae leaving phosphorylation in the granule cell bodies intact. Delivery of the mTOR inhibitor, rapamycin, abolished activation of rpS6 phosphorylation in granule cell bodies and dendrites, whereas delivery of a selective S6K1 inhibitor, PF4708671, or RSK inhibitor, SL0101-1, attenuated rpS6 phosphorylation throughout the postsynaptic cell. These results reveal that MAPK/ERK-dependent signaling is predominately responsible for the selective induction of rpS6 phosphorylation at active synapses. In contrast, PI3-kinase/mTOR-dependent signaling induces rpS6 phosphorylation throughout the somatodendritic compartment but plays a minimal role at active synapses. Collectively, these results suggest a potential mechanism by which PI3-kinase/mTOR and MAPK/ERK pathways regulate translation at specific subcellular compartments in response to synaptic activity.


Asunto(s)
Cuerpo Celular/metabolismo , Dendritas/metabolismo , Sistema de Señalización de MAP Quinasas , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína S6 Ribosómica/metabolismo , Sinapsis/metabolismo , Androstadienos/farmacología , Animales , Benzopiranos/farmacología , Butadienos/farmacología , Cuerpo Celular/efectos de los fármacos , Cromonas/farmacología , Dendritas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Monosacáridos/farmacología , Morfolinas/farmacología , Nitrilos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas/metabolismo , Sirolimus/farmacología , Sinapsis/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Wortmanina
10.
J Biol Chem ; 291(25): 13335-48, 2016 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-27129212

RESUMEN

Peripheral nerve injury induces increased expression of thrombospondin-4 (TSP4) in spinal cord and dorsal root ganglia that contributes to neuropathic pain states through unknown mechanisms. Here, we test the hypothesis that TSP4 activates its receptor, the voltage-gated calcium channel Cavα2δ1 subunit (Cavα2δ1), on sensory afferent terminals in dorsal spinal cord to promote excitatory synaptogenesis and central sensitization that contribute to neuropathic pain states. We show that there is a direct molecular interaction between TSP4 and Cavα2δ1 in the spinal cord in vivo and that TSP4/Cavα2δ1-dependent processes lead to increased behavioral sensitivities to stimuli. In dorsal spinal cord, TSP4/Cavα2δ1-dependent processes lead to increased frequency of miniature and amplitude of evoked excitatory post-synaptic currents in second-order neurons as well as increased VGlut2- and PSD95-positive puncta, indicative of increased excitatory synapses. Blockade of TSP4/Cavα2δ1-dependent processes with Cavα2δ1 ligand gabapentin or genetic Cavα2δ1 knockdown blocks TSP4 induced nociception and its pathological correlates. Conversely, TSP4 antibodies or genetic ablation blocks nociception and changes in synaptic transmission in mice overexpressing Cavα2δ1 Importantly, TSP4/Cavα2δ1-dependent processes also lead to similar behavioral and pathological changes in a neuropathic pain model of peripheral nerve injury. Thus, a TSP4/Cavα2δ1-dependent pathway activated by TSP4 or peripheral nerve injury promotes exaggerated presynaptic excitatory input and evoked sensory neuron hyperexcitability and excitatory synaptogenesis, which together lead to central sensitization and pain state development.


Asunto(s)
Canales de Calcio/metabolismo , Neuralgia/metabolismo , Trombospondinas/fisiología , Animales , Células HEK293 , Humanos , Masculino , Ratones Transgénicos , Células del Asta Posterior/fisiología , Sinapsis/fisiología , Potenciales Sinápticos
11.
Learn Mem ; 23(6): 255-69, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27194793

RESUMEN

Previous studies have shown that induction of long-term potentiation (LTP) induces phosphorylation of ribosomal protein S6 (rpS6) in postsynaptic neurons, but the functional significance of rpS6 phosphorylation is poorly understood. Here, we show that synaptic stimulation that induces perforant path LTP triggers phosphorylation of rpS6 (p-rpS6) locally near active synapses. Using antibodies specific for phosphorylation at different sites (ser235/236 versus ser240/244), we show that strong synaptic activation led to dramatic increases in immunostaining throughout postsynaptic neurons with selectively higher staining for p-ser235/236 in the activated dendritic lamina. Following LTP induction, phosphorylation at ser235/236 was detectable by 5 min, peaked at 30 min, and was maintained for hours. Phosphorylation at both sites was completely blocked by local infusion of the NMDA receptor antagonist, APV. Despite robust induction of p-rpS6 following high frequency stimulation, assessment of protein synthesis by autoradiography revealed no detectable increases. Exploration of a novel environment led to increases in the number of p-rpS6-positive neurons throughout the forebrain in a pattern reminiscent of immediate early gene induction and many individual neurons that were p-rpS6-positive coexpressed Arc protein. Our results constrain hypotheses about the possible role of rpS6 phosphorylation in regulating postsynaptic protein synthesis during induction of synaptic plasticity.


Asunto(s)
Dendritas/metabolismo , Aprendizaje/fisiología , Potenciación a Largo Plazo , Proteína S6 Ribosómica/metabolismo , Sinapsis/metabolismo , Animales , Astrocitos/metabolismo , Conducta Exploratoria , Femenino , Neuronas/metabolismo , Fosforilación , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo
13.
Am J Pathol ; 185(10): 2819-32, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26435414

RESUMEN

The oral drug FTY720 affects sphingosine-1-phosphate (S1P) signaling on targeted cells that bear the S1P receptors S1P1, S1P3, S1P4, and S1P5. We examined the effect of FTY720 treatment on the biology of mouse neural progenitor cells (NPCs) after transplantation in a viral model of demyelination. Intracerebral infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in an acute encephalomyelitis, followed by demyelination similar in pathology to the human demyelinating disease, multiple sclerosis. We have previously reported that intraspinal transplantation of mouse NPCs into JHMV-infected animals resulted in selective colonization of demyelinated lesions, preferential differentiation into oligodendroglia accompanied by axonal preservation, and increased remyelination. Cultured NPCs expressed transcripts for S1P receptors S1P1, S1P2, S1P3, S1P4, and S1P5. FTY720 treatment of cultured NPCs resulted in increased mitogen-activated protein kinase phosphorylation and migration after exposure to the chemokine CXCL12. Administration of FTY720 to JHMV-infected mice resulted in enhanced migration and increased proliferation of transplanted NPCs after spinal cord engraftment. FTY720 treatment did not improve clinical disease, diminish neuroinflammation or the severity of demyelination, nor increase remyelination. These findings argue that FTY720 treatment selectively increases NPC proliferation and migration but does not either improve clinical outcome or enhance remyelination after transplantation into animals in which immune-mediated demyelination is initiated by the viral infection of the central nervous system.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Enfermedades Desmielinizantes/patología , Virus de la Hepatitis Murina/aislamiento & purificación , Células-Madre Neurales/citología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Animales , Axones/patología , Células Cultivadas , Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/virología , Clorhidrato de Fingolimod/farmacología , Inmunosupresores/farmacología , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Esclerosis Múltiple/patología , Esclerosis Múltiple/virología , Células-Madre Neurales/efectos de los fármacos , Oligodendroglía/citología
14.
J Neurosci ; 34(30): 9951-62, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25057197

RESUMEN

Conditional genetic deletion of phosphatase and tensin homolog (PTEN) in the sensorimotor cortex of neonatal mice enables regeneration of corticospinal tract (CST) axons after spinal cord injury (SCI). The present study addresses three questions: (1) whether PTEN knockdown in adult rats by nongenetic techniques enables CST regeneration, (2) whether interventions to enable CST regeneration enhance recovery of voluntary motor function, and (3) whether delivery of salmon fibrin into the injury site further enhances CST regeneration and motor recovery. Adult rats were trained in a staircase-reaching task and then received either intracortical injections of AAVshPTEN to delete PTEN or a control vector expressing shRNA for luciferase (AAVshLuc). Rats then received cervical dorsal hemisection injuries and salmon fibrin was injected into the injury site in half the rats, yielding four groups (AAVshPTEN, AAVshLuc, AAVshPTEN + fibrin, and AAVshLuc + fibrin). Forepaw function was assessed for 10 weeks after injury and CST axons were traced by injecting biotin-conjugated dextran amine into the sensorimotor cortex. Rats that received AAVshPTEN alone did not exhibit improved motor function, whereas rats that received AAVshPTEN and salmon fibrin had significantly higher forelimb-reaching scores. Tract tracing revealed that CST axons extended farther caudally in the group that received AAVshPTEN and salmon fibrin versus other groups. There were no significant differences in lesion size between the groups. Together, these data suggest that the combination of PTEN deletion and salmon fibrin injection into the lesion can significantly improve voluntary motor function after SCI by enabling regenerative growth of CST axons.


Asunto(s)
Fibrina/administración & dosificación , Regeneración Nerviosa/efectos de los fármacos , Fosfohidrolasa PTEN/antagonistas & inhibidores , Tractos Piramidales/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , Traumatismos de la Médula Espinal/tratamiento farmacológico , Factores de Edad , Animales , Axones/efectos de los fármacos , Axones/fisiología , Vértebras Cervicales , Dependovirus/genética , Femenino , Inyecciones Intraventriculares , Regeneración Nerviosa/genética , Fosfohidrolasa PTEN/deficiencia , Tractos Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética , Salmón , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo
15.
J Neurosci ; 34(13): 4481-93, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24671994

RESUMEN

Arc is an immediate early gene that is unique among neuronal mRNAs because its transcripts are transported into dendrites and accumulate near activated synapses, presumably to be translated locally. These qualities pose Arc as playing an important, yet not fully understood, role in the activity-dependent modifications of synapses that are thought to underlie memory storage. Here we show in vivo in rats that newly synthesized Arc mRNA accumulates at activated synapses and that synaptic activity simultaneously triggers mRNA decay that eliminates Arc mRNA from inactive dendritic domains. Arc mRNA degradation occurs throughout the dendrite and requires both NMDA receptor activation and active translation. Synaptic activation did not lead to decreases in another dendritic mRNA (αCaMKII), indicating that there is not a general activation of mRNA degradation in dendrites. These data reveal a novel mechanism for controlling mRNA distribution within dendrites and highlight activity-dependent mRNA degradation as a regulatory process involved in synaptic plasticity.


Asunto(s)
Proteínas del Citoesqueleto/genética , Dendritas/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , Transcripción Genética/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Antagonistas de Aminoácidos Excitadores , Femenino , Lateralidad Funcional , Hipocampo/citología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Transducción de Señal , Sinapsis/metabolismo , Factores de Tiempo
16.
J Neurosci ; 34(42): 14013-21, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25319698

RESUMEN

We reported previously the formation of ectopic colonies in widespread areas of the nervous system after transplantation of fetal neural stem cells (NSCs) into spinal cord transection sites. Here, we characterize the incidence, distribution, and cellular composition of the colonies. NSCs harvested from E14 spinal cords from rats that express GFP were treated with a growth factor cocktail and grafted into the site of a complete spinal cord transection. Two months after transplant, spinal cord and brain tissue were analyzed histologically. Ectopic colonies were found at long distances from the transplant in the central canal of the spinal cord, the surface of the brainstem and spinal cord, and in the fourth ventricle. Colonies were present in 50% of the rats, and most rats had multiple colonies. Axons extended from the colonies into the host CNS. Colonies were strongly positive for nestin, a marker for neural precursors, and contained NeuN-positive cells with processes resembling dendrites, GFAP-positive astrocytes, APC/CC1-positive oligodendrocytes, and Ki-67-positive cells, indicating ongoing proliferation. Stereological analyses revealed an estimated 21,818 cells in a colony in the fourth ventricle, of which 1005 (5%) were Ki-67 positive. Immunostaining for synaptic markers (synaptophysin and VGluT-1) revealed large numbers of synaptophysin-positive puncta within the colonies but fewer VGluT-1 puncta. Continuing expansion of NSC-derived cell masses in confined spaces in the spinal cord and brain could produce symptoms attributable to compression of nearby tissue. It remains to be determined whether other cell types with self-renewing potential can also form colonies.


Asunto(s)
Coristoma , Sistema Nervioso , Células-Madre Neurales/trasplante , Índice de Severidad de la Enfermedad , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Animales , Femenino , Sistema Nervioso/patología , Embarazo , Ratas , Ratas Endogámicas F344 , Traumatismos de la Médula Espinal/patología
17.
J Biol Chem ; 289(30): 20615-29, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24917666

RESUMEN

The mammalian target of rapamycin (mTOR) pathway has multiple important physiological functions, including regulation of protein synthesis, cell growth, autophagy, and synaptic plasticity. Activation of mTOR is necessary for the many beneficial effects of brain-derived neurotrophic factor (BDNF), including dendritic translation and memory formation in the hippocampus. At present, however, the role of mTOR in BDNF's support of survival is not clear. We report that mTOR activation is necessary for BDNF-dependent survival of primary rat hippocampal neurons, as either mTOR inhibition by rapamycin or genetic manipulation of the downstream molecule p70S6K specifically blocked BDNF rescue. Surprisingly, however, BDNF did not promote neuron survival by up-regulating mTOR-dependent protein synthesis or through mTOR-dependent suppression of caspase-3 activation. Instead, activated mTOR was responsible for BDNF's suppression of autophagic flux. shRNA against the autophagic machinery Atg7 or Atg5 prolonged the survival of neurons co-treated with BDNF and rapamycin, suggesting that suppression of mTOR in BDNF-treated cells resulted in excessive autophagy. Finally, acting as a physiological analog of rapamycin, IL-1ß impaired BDNF signaling by way of inhibiting mTOR activation as follows: the cytokine induced caspase-independent neuronal death and accelerated autophagic flux in BDNF-treated cells. These findings reveal a novel mechanism of BDNF neuroprotection; BDNF not only prevents apoptosis through inhibiting caspase activation but also promotes neuron survival through modulation of autophagy. This protection mechanism is vulnerable under chronic inflammation, which deregulates autophagy through impairing mTOR signaling. These results may be relevant to age-related changes observed in neurodegenerative diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Inmunosupresores/farmacología , Interleucina-1beta/metabolismo , Neuronas/metabolismo , Sirolimus/farmacología , Animales , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Masculino , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo
18.
J Biol Chem ; 289(10): 7025-7037, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24459143

RESUMEN

To investigate a potential mechanism underlying trigeminal nerve injury-induced orofacial hypersensitivity, we used a rat model of chronic constriction injury to the infraorbital nerve (CCI-ION) to study whether CCI-ION caused calcium channel α2δ1 (Cavα2δ1) protein dysregulation in trigeminal ganglia and associated spinal subnucleus caudalis and C1/C2 cervical dorsal spinal cord (Vc/C2). Furthermore, we studied whether this neuroplasticity contributed to spinal neuron sensitization and neuropathic pain states. CCI-ION caused orofacial hypersensitivity that correlated with Cavα2δ1 up-regulation in trigeminal ganglion neurons and Vc/C2. Blocking Cavα2δ1 with gabapentin, a ligand for the Cavα2δ1 proteins, or Cavα2δ1 antisense oligodeoxynucleotides led to a reversal of orofacial hypersensitivity, supporting an important role of Cavα2δ1 in orofacial pain processing. Importantly, increased Cavα2δ1 in Vc/C2 superficial dorsal horn was associated with increased excitatory synaptogenesis and increased frequency, but not the amplitude, of miniature excitatory postsynaptic currents in dorsal horn neurons that could be blocked by gabapentin. Thus, CCI-ION-induced Cavα2δ1 up-regulation may contribute to orofacial neuropathic pain states through abnormal excitatory synapse formation and enhanced presynaptic excitatory neurotransmitter release in Vc/C2.


Asunto(s)
Canales de Calcio/metabolismo , Dolor Facial/metabolismo , Neuralgia/metabolismo , Ganglio del Trigémino/metabolismo , Traumatismos del Nervio Trigémino/complicaciones , Animales , Canales de Calcio/genética , Canales de Calcio Tipo L , Modelos Animales de Enfermedad , Dolor Facial/etiología , Dolor Facial/genética , Masculino , Neuralgia/etiología , Neuralgia/genética , Ratas , Ratas Sprague-Dawley , Núcleo Caudal del Trigémino/metabolismo
19.
Ann Neurol ; 73(2): 210-23, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23281061

RESUMEN

OBJECTIVE: Traumatic peripheral nerve injuries often produce permanent functional deficits despite optimal surgical and medical management. One reason for the impaired target organ reinnervation is degradation of motor endplates during prolonged denervation. Here we investigate the effect of preserving agrin on the stability of denervated endplates. Because matrix metalloproteinase 3 (MMP3) is known to degrade agrin, we examined the changes in endplate structure following traumatic nerve injury in MMP3 knockout mice. METHODS: After creation of a critical size nerve defect to preclude reinnervation, we characterized receptor area, receptor density, and endplate morphology in denervated plantaris muscles in wild-type and MMP3 null mice. The level of agrin and muscle-specific kinase (MuSK) was assessed at denervated endplates. In addition, denervated muscles were subjected to ex vivo stimulation with acetylcholine. Finally, reinnervation potential was compared after long-term denervation. RESULTS: In wild-type mice, the endplates demonstrated time-dependent decreases in area and receptor density and conversion to an immature receptor phenotype. In striking contrast, all denervation-induced changes were attenuated in MMP3 null mice, with endplates retaining their differentiated form. Agrin and MuSK were preserved in endplates from denervated MMP3 null animals. Furthermore, denervated muscles from MMP3 null mice demonstrated greater endplate efficacy and reinnervation. INTERPRETATION: These results demonstrate a critical role for MMP3 in motor endplate remodeling, and reveal a potential target for therapeutic intervention to prevent motor endplate degradation following nerve injury.


Asunto(s)
Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Placa Motora/enzimología , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/fisiopatología , Acetilcolina/farmacología , Agrina/metabolismo , Animales , Línea Celular , Agonistas Colinérgicos/farmacología , Modelos Animales de Enfermedad , Eliminación de Gen , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Actividad Motora/fisiología , Placa Motora/efectos de los fármacos , Desnervación Muscular/métodos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inervación , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Degeneración Walleriana/metabolismo , Degeneración Walleriana/fisiopatología
20.
J Immunol ; 188(11): 5257-66, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22523388

RESUMEN

Spinal cord injury (SCI) results in immune depression. To better understand how injury inhibits humoral immunity, the effects of chronic thoracic SCI on B cell development and immune responses to thymus-independent type 2 and thymus-dependent Ags were determined. Mice received complete crush injury or control laminectomy at either thoracic level 3, which disrupts descending autonomic control of the spleen, or at thoracic level 9, which conserves most splenic sympathetic activity. Although mature B cell numbers were only mildly reduced, bone marrow B cell production was transiently but profoundly depressed immediately after injury. Despite the return of normal B cell production 4 wk after SCI, mice receiving thoracic level 3 injury showed a significant reduction in their ability to mount primary thymus-independent type 2 or thymus-dependent immune responses. The latter were marked by decreases in germinal center B cells as well as class-switched high-affinity Ab-secreting cells. Importantly, injury did not affect affinity maturation per se, pre-existing B cell memory, or secondary humoral immune responses. Taken together, these findings show that chronic high thoracic SCI impairs the ability to mount optimal Ab responses to new antigenic challenges, but spares previously established humoral immunity.


Asunto(s)
Formación de Anticuerpos/inmunología , Traumatismos de la Médula Espinal/inmunología , Enfermedad Aguda , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Enfermedad Crónica , Femenino , Recuento de Linfocitos , Linfopoyesis/inmunología , Ratones , Ratones Endogámicos C57BL , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Bazo/citología , Bazo/inmunología , Bazo/patología , Timo/citología , Timo/inmunología , Timo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA