Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Glia ; 71(8): 2045-2066, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37132422

RESUMEN

Remyelination and neurodegeneration prevention mitigate disability in Multiple Sclerosis (MS). We have shown acute intermittent hypoxia (AIH) is a novel, non-invasive and effective therapy for peripheral nerve repair, including remyelination. Thus, we posited AIH would improve repair following CNS demyelination and address the paucity of MS repair treatments. AIH's capacity to enhance intrinsic repair, functional recovery and alter disease course in the experimental autoimmune encephalomyelitis (EAE) model of MS was assessed. EAE was induced by MOG35-55 immunization in C57BL/6 female mice. EAE mice received either AIH (10 cycles-5 min 11% oxygen alternating with 5 min 21% oxygen) or Normoxia (control; 21% oxygen for same duration) once daily for 7d beginning at near peak EAE disease score of 2.5. Mice were followed post-treatment for an additional 7d before assessing histopathology or 14d to examine maintenance of AIH effects. Alterations in histopathological correlates of multiple repair indices were analyzed quantitatively in focally demyelinated ventral lumbar spinal cord areas to assess AIH impacts. AIH begun at near peak disease significantly improved daily clinical scores/functional recovery and associated histopathology relative to Normoxia controls and the former were maintained for at least 14d post-treatment. AIH enhanced correlates of myelination, axon protection and oligodendrocyte precursor cell recruitment to demyelinated areas. AIH also effected a dramatic reduction in inflammation, while polarizing remaining macrophages/microglia toward a pro-repair state. Collectively, this supports a role for AIH as a novel non-invasive therapy to enhance CNS repair and alter disease course following demyelination and holds promise as a neuroregenerative MS strategy.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Remielinización , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Animales , Ratones , Ratones Endogámicos C57BL , Anaerobiosis , Oxígeno , Femenino
2.
Clin Exp Pharmacol Physiol ; 48(8): 1090-1102, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33638234

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is an inflammatory demyelinating disease featured with neuroinflammation, demyelination, and the loss of oligodendrocytes. Cognitive impairment and depression are common neuropsychiatric symptoms in MS that are poorly managed with the present interventions. OBJECTIVE: This study aimed to investigate the effects of low field magnetic stimulation (LFMS), a novel non-invasive neuromodulation technology, on cognitive impairment and depressive symptoms associated with MS using a mouse model of demyelination. METHODS: C57BL female mice were fed with a 0.2% cuprizone diet for 12 weeks to induce a chronic demyelinating model followed by 4 weeks of cuprizone withdrawal with either sham or LFMS treatment. RESULTS: Improved cognition and depression-like behaviour and restored weight gain were observed in mice with LFMS treatment. Immunohistochemical and immunoblotting data showed enhanced myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein expressions (MOG) in the prefrontal cortex of mice with LFMS treatment, supporting that myelin repair was promoted. LFMS also increased the protein expression of mature oligodendrocyte biomarker glutathione-S-transferase (GST-π). In addition, expression of TGF-ß and associated receptors were elevated with LFMS treatment, implicating this pathway in the response. CONCLUSION: Results from the present study revealed LFMS to have neuroprotective effects, suggesting that LFMS has potential therapeutic value for treating cognitive impairment and depression related to demyelination disorders.


Asunto(s)
Cuprizona , Animales , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina , Enfermedades Neuroinflamatorias , Oligodendroglía
3.
Ann Neurol ; 83(4): 691-702, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29537631

RESUMEN

By altering the intrinsic metabolism of the cell, including the upregulation of regeneration-associated genes (RAGs) and the production of structural proteins for axonal outgrowth, the conditioning lesion sets up an environment highly conducive to regeneration. In this review, we assess 40 years of research to provide a comprehensive overview of the conditioning lesion literature, directed at (1) discussing the mechanisms of and barriers to nerve regeneration that can be mitigated by the conditioning lesion, (2) describing the cellular and molecular pathways implicated in the conditioning lesion effect, and (3) deliberating on how these insights might be applied clinically. The consequential impact on regeneration is profound, with a conditioned nerve demonstrating longer neurite extensions in vitro, enhanced expression of RAGs within the dorsal root ganglia, early assembly and transportation of cytoskeletal elements, accelerated axonal growth, and improved functional recovery in vivo. Although this promising technique is not yet feasible to be performed in humans, there are potential strategies, such as conditioning electrical stimulation that may be explored to allow nerve conditioning in a clinically safe and well-tolerated manner. Ann Neurol 2018;83:691-702.


Asunto(s)
Regeneración Nerviosa/fisiología , Neuritas/fisiología , Nervios Periféricos/fisiología , Nervios Periféricos/fisiopatología , Animales , Humanos
4.
Proc Natl Acad Sci U S A ; 111(45): 16142-7, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25349404

RESUMEN

Luman/cAMP response element binding protein 3 is an endoplasmic reticulum (ER) transmembrane basic leucine zipper transcription factor whose mRNA and protein localize to adult sensory axons, the latter with axonal ER components along the axon length. Here we show that axon-derived Luman plays an important role in relaying information about axonal injury to the neuronal cell body. Axotomy induces axonal Luman synthesis and also release from the axonal ER of Luman's transcriptionally active amino terminus, which is transported to the cell body in an importin-mediated manner. Visualization of the activation and retrograde translocation of Luman into the nucleus in real time both in vivo and in vitro was accomplished using a specially created N- and C-terminal-tagged Luman adenoviral vector. Small interfering RNA used to reduce Luman expression either neuronally or just axonally significantly impaired the ability of 24-h injury-conditioned sensory neurons to extend the regeneration-associated elongating form of axon growth but had no impact on axon outgrowth in naïve neurons. Collectively, these findings link injury-associated axonal ER responses proximal to the site of injury to the intrinsic regenerative growth capacity of adult sensory neurons.


Asunto(s)
Axones/metabolismo , Núcleo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Retículo Endoplásmico/metabolismo , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular , Animales , Axones/patología , Núcleo Celular/genética , Chlorocebus aethiops , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/patología , Regulación de la Expresión Génica , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/patología , Estructura Terciaria de Proteína , Ratas , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología , Células Vero
5.
J Neurosci ; 35(43): 14557-70, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26511246

RESUMEN

We recently revealed that the axon endoplasmic reticulum resident transcription factor Luman/CREB3 (herein called Luman) serves as a unique retrograde injury signal in regulation of the intrinsic elongating form of sensory axon regeneration. Here, evidence supports that Luman contributes to axonal regeneration through regulation of the unfolded protein response (UPR) and cholesterol biosynthesis in adult rat sensory neurons. One day sciatic nerve crush injury triggered a robust increase in UPR-associated mRNA and protein expression in both neuronal cell bodies and the injured axons. Knockdown of Luman expression in 1 d injury-conditioned neurons by siRNA attenuated axonal outgrowth to 48% of control injured neurons and was concomitant with reduced UPR- and cholesterol biosynthesis-associated gene expression. UPR PCR-array analysis coupled with qRT-PCR identified and confirmed that four transcripts involved in cholesterol regulation were downregulated >2-fold by the Luman siRNA treatment of the injury-conditioned neurons. Further, the Luman siRNA-attenuated outgrowth could be significantly rescued by either cholesterol supplementation or 2 ng/ml of the UPR inducer tunicamycin, an amount determined to elevate the depressed UPR gene expression to a level equivalent of that observed with crush injury. Using these approaches, outgrowth increased significantly to 74% or 69% that of injury-conditioned controls, respectively. The identification of Luman as a regulator of the injury-induced UPR and cholesterol at levels that benefit the intrinsic ability of axotomized adult rat sensory neurons to undergo axonal regeneration reveals new therapeutic targets to bolster nerve repair.


Asunto(s)
Axones/fisiología , Colesterol/biosíntesis , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Regeneración Nerviosa/genética , Desplegamiento Proteico , Células Receptoras Sensoriales/fisiología , Animales , Recuento de Células , Ganglios Espinales/citología , Técnicas de Silenciamiento del Gen , Masculino , Compresión Nerviosa , Neuritas/efectos de los fármacos , Neuritas/fisiología , Desplegamiento Proteico/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Wistar , Neuropatía Ciática/genética , Neuropatía Ciática/patología , Tunicamicina/farmacología
6.
Glia ; 64(9): 1546-61, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27353566

RESUMEN

Demyelinating peripheral nerves are infiltrated by cells of the monocyte lineage, including macrophages, which are highly plastic, existing on a continuum from pro-inflammatory M1 to pro-repair M2 phenotypic states. Whether one can therapeutically manipulate demyelinated peripheral nerves to promote a pro-repair M2 phenotype remains to be elucidated. We previously identified brief electrical nerve stimulation (ES) as therapeutically beneficial for remyelination, benefits which include accelerated clearance of macrophages, making us theorize that ES alters the local immune response. Thus, the impact of ES on the immune microenvironment in the zone of demyelination was examined. Adult male rat tibial nerves were focally demyelinated via 1% lysophosphatidyl choline (LPC) injection. Five days later, half underwent 1 hour 20 Hz sciatic nerve ES proximal to the LPC injection site. ES had a remarkable and significant impact, shifting the macrophage phenotype from predominantly pro-inflammatory/M1 toward a predominantly pro-repair/M2 one, as evidenced by an increased incidence of expression of M2-associated phenotypic markers in identified macrophages and a decrease in M1-associated marker expression. This was discernible at 3 days post-ES (8 days post-LPC) and continued at the 5 day post-ES (10 days post-LPC) time point examined. ES also affected chemokine (C-C motif) ligand 2 (CCL2; aka MCP-1) expression in a manner that correlated with increases and decreases in macrophage numbers observed in the demyelination zone. The data establish that briefly increasing neuronal activity favorably alters the immune microenvironment in demyelinated nerve, rapidly polarizing macrophages toward a pro-repair phenotype, a beneficial therapeutic concept that may extend to other pathologies. GLIA 2016;64:1546-1561.


Asunto(s)
Axones/patología , Enfermedades Desmielinizantes/patología , Estimulación Eléctrica , Macrófagos/citología , Vaina de Mielina/patología , Nervio Ciático/patología , Animales , Enfermedades Desmielinizantes/terapia , Masculino , Monocitos/patología , Regeneración Nerviosa/fisiología , Ratas Wistar , Células de Schwann/citología
7.
J Neurosci ; 33(19): 8202-15, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23658159

RESUMEN

Activation of the nerve growth factor (NGF) receptor trkA and tissue acidosis are critically linked to inflammation-associated nociceptor sensitization. This study explored how increased acidity is linked to sensory neuron sensitization to NGF. Adult Wistar rat primary sensory neurons grown at physiological pH 7.4, then either kept at pH 7.4 or challenged for 30 min in pH 6.5 medium, provided a model of acidosis. Nonpermeabilizing trkA immunofluorescence revealed a significant increase in trkA mobilization to the plasma membrane from intracellular stores in response to proton challenge. This was confirmed using a surface protein biotinylation assay and Brefeldin A disruption of the rough endoplasmic reticulum-Golgi-trans-Golgi network. Mobilization of trkA to the membrane at pH 6.5 was abolished in neurons treated with the acid-sensitive ion channel blocker, amiloride. While elevated levels of NGF-independent trkA phosphorylation occurred at pH 6.5 alone, the level of activation was significantly increased in response to NGF challenge. Exposure of sensory neurons to pH 6.5 medium also resulted in strong calcium (Ca(2+)) transients that were reversible upon reintroduction to physiological pH. The pH 6.5-induced mobilization of trkA to the membrane was Ca(2+) dependent, as BAPTA-AM Ca(2+) chelation abrogated the response. Interestingly, KCl-induced depolarization was sufficient to induce mobilization of trkA to the cell surface at pH 7.4, but did not augment the response to pH 6.5. In conclusion, increased mobilization of trkA to neuronal membranes in response to either acidosis or neuronal depolarization provides two novel mechanisms by which sensory neurons can rapidly sensitize to NGF and has important implications for inflammatory pain states.


Asunto(s)
Líquido Extracelular/metabolismo , Receptor trkA/metabolismo , Células Receptoras Sensoriales/metabolismo , Acidosis/fisiopatología , Animales , Anticuerpos/farmacología , Biotinilación , Brefeldino A/farmacología , Calcio/metabolismo , Células Cultivadas , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Líquido Extracelular/efectos de los fármacos , Ganglios Espinales/citología , Concentración de Iones de Hidrógeno , Isótopos de Yodo/farmacocinética , Masculino , Factor de Crecimiento Nervioso/inmunología , Factor de Crecimiento Nervioso/farmacocinética , Cloruro de Potasio/farmacología , Unión Proteica/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos
8.
Glia ; 62(5): 763-77, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24616056

RESUMEN

Satellite glial cells (SGCs) surrounding primary sensory neurons are similar to astrocytes of the central nervous system in that they buffer the extracellular environment via potassium and calcium channels and express the intermediate filament glial fibrillary acidic protein (GFAP). Peripheral nerve injury induces a reactive state in SGCs that includes SGC proliferation, increased SGC/SGC coupling via gap junctions, decreased inward rectifying potassium channel 4.1 (Kir 4.1) expression and increased expression of GFAP and the common neurotrophin receptor, p75NTR. In contrast, neuronal p75NTR expression, normally detected in ∼80% of adult rat sensory neurons, decreases in response to peripheral axotomy. Given the differential regulation of p75NTR expression in neurons versus SGCs with injury, we hypothesized that reduced signaling via neuronal p75NTR contributes to the induction of a reactive state in SGCs. We found that reducing neuronal p75NTR protein expression in uninjured sensory neurons by intrathecal subarachnoid infusion of p75NTR-selective anti-sense oligodeoxynucleotides for one week was sufficient to induce a "reactive-like" state in the perineuronal SGCs akin to that normally observed following peripheral nerve injury. This reactive state included significantly increased SGC p75NTR, GFAP and gap junction protein connexin-43 protein expression, increased numbers of SGCs surrounding individual sensory neurons and decreased SGC Kir 4.1 channel expression. Collectively, this supports the tenet that reductions in target-derived trophic support leading to, or as a consequence of, reduced neuronal p75NTR expression plays a critical role in switching the SGC to a reactive state.


Asunto(s)
Ganglios Espinales/lesiones , Ganglios Espinales/metabolismo , Neuroglía/metabolismo , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Células Satélites Perineuronales/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Ganglios Espinales/efectos de los fármacos , Regulación de la Expresión Génica , Inyecciones Espinales , Masculino , Proteínas del Tejido Nervioso , Neuroglía/efectos de los fármacos , Oligonucleótidos Antisentido/administración & dosificación , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento , Células Satélites Perineuronales/efectos de los fármacos , Células Receptoras Sensoriales/efectos de los fármacos
9.
Handb Clin Neurol ; 201: 1-17, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38697733

RESUMEN

Peripheral nerves are functional networks in the body. Disruption of these networks induces varied functional consequences depending on the types of nerves and organs affected. Despite the advances in microsurgical repair and understanding of nerve regeneration biology, restoring full functions after severe traumatic nerve injuries is still far from achieved. While a blunted growth response from axons and errors in axon guidance due to physical barriers may surface as the major hurdles in repairing nerves, critical additional cellular and molecular aspects challenge the orderly healing of injured nerves. Understanding the systematic reprogramming of injured nerves at the cellular and molecular levels, referred to here as "hallmarks of nerve injury regeneration," will offer better ideas. This chapter discusses the hallmarks of nerve injury and regeneration and critical points of failures in the natural healing process. Potential pharmacological and nonpharmacological intervention points for repairing nerves are also discussed.


Asunto(s)
Regeneración Nerviosa , Traumatismos de los Nervios Periféricos , Animales , Humanos , Axones/fisiología , Axones/patología , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/terapia , Traumatismos de los Nervios Periféricos/fisiopatología , Nervios Periféricos
10.
Neural Regen Res ; 17(5): 1042-1050, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34558531

RESUMEN

Our lab has shown that brief electrical nerve stimulation (ES) has a dramatic impact on remyelination of lysophosphatidyl choline (LPC)-induced focally demyelinated rat peripheral nerves, while also inducing an axon-protective phenotype and shifting macrophages from a predominantly pro-inflammatory toward a pro-repair phenotype. Whether this same potential exists in the central nervous system is not known. Thus, for proof of principle studies, the peripheral nerve demyelination and ES model was adapted to the central nervous system, whereby a unilateral focal LPC-induced demyelination of the dorsal column at the lumbar enlargement where the sciatic nerve afferents enter was created, so that subsequent ipsilateral sciatic nerve ES results in increased neural activity in the demyelinated axons. Data reveal a robust focal demyelination at 7 days post-LPC injection. Delivery of 1-hour ES at 7 days post-LPC polarizes macrophages/microglia toward a pro-repair phenotype when examined at 14 days post-LPC; results in smaller LPC-associated regions of inflammation compared to non-stimulated controls; results in significantly more cells of the oligodendroglial lineage in the demyelinated region; elevates myelin basic protein levels; and shifts the paranodal protein Caspr along demyelinated axons to a more restricted distribution, consistent with reformation of the paranodes of the nodes of Ranvier. ES also significantly enhanced levels of phosphorylated neurofilaments detected in the zones of demyelination, which has been shown to confer axon protection. Collectively these findings support that strategies that increase neural activity, such as brief electrical stimulation, can be beneficial for promoting intrinsic repair following focal demyelinating insults in demyelinating diseases such as multiple sclerosis. All animal procedures performed were approved by the University of Saskatchewan's Animal Research Ethics Board (protocol# 20090087; last approval date: November 5, 2020).

11.
Neural Regen Res ; 15(12): 2353-2361, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32594060

RESUMEN

Emerging evidence supports that the stress response to peripheral nerve injury extends beyond the injured neuron, with alterations in associated transcription factors detected both locally and remote to the lesion. Stress-induced nuclear translocation of the transcription factor forkhead class box O3a (FOXO3a) was initially linked to activation of apoptotic genes in many neuronal subtypes. However, a more complex role of FOXO3a has been suggested in the injury response of sensory neurons, with the injured neuron expressing less FOXO3a. To elucidate this response and test whether non-injured sensory neurons also alter FOXO3a expression, the temporal impact of chronic unilateral L4-6 spinal nerve transection on FOXO3a expression and nuclear localization in adult rat dorsal root ganglion neurons ipsilateral, contralateral or remote to injury relative to naïve controls was examined. In naïve neurons, high cytoplasmic and nuclear levels of FOXO3a colocalized with calcitonin gene related peptide, a marker of the nociceptive subpopulation. One hour post-injury, an acute increase in nuclear FOXO3a in small size injured neurons occurred followed by a significant decrease after 1, 2 and 4 days, with levels increasing toward pre-injury levels by 1 week post-injury. A more robust biphasic response to the injury was observed in uninjured neurons contralateral to and those remote to injury. Nuclear levels of FOXO3a peaked at 1 day, decreased by 4 days, then increased by 1 week post-injury, a response mirrored in C4 dorsal root ganglion neurons remote to injury. This altered expression contralateral and remote to injury supports that spinal nerve damage has broader systemic impacts, a response we recently reported for another stress transcription factor, Luman/CREB3. The early decreased expression and nuclear localization of FOXO3a in the injured neuron implicate these changes in the cell body response to injury that may be protective. Finally, the broader systemic changes support the existence of stress/injury-induced humeral factor(s) influencing transcriptional and potentially behavioral changes in uninjured dorsal root ganglion neurons. Approval to conduct this study was obtained from the University of Saskatchewan Animal Research Ethics Board (protocol #19920164).

12.
eNeuro ; 7(2)2020.
Artículo en Inglés | MEDLINE | ID: mdl-32001550

RESUMEN

In contrast to neurons in the CNS, damaged neurons from the peripheral nervous system (PNS) regenerate, but this process can be slow and imperfect. Successful regeneration is orchestrated by cytoskeletal reorganization at the tip of the proximal axon segment and cytoskeletal disassembly of the distal segment. Collapsin response mediator protein 4 (CRMP4) is a cytosolic phospho-protein that regulates the actin and microtubule cytoskeleton. During development, CRMP4 promotes growth cone formation and dendrite development. Paradoxically, in the adult CNS, CRMP4 impedes axon regeneration. Here, we investigated the involvement of CRMP4 in peripheral nerve injury in male and female Crmp4-/- mice following sciatic nerve injury. We find that sensory axon regeneration and Wallerian degeneration are impaired in Crmp4-/- mice following sciatic nerve injury. In vitro analysis of dissociated dorsal root ganglion (DRG) neurons from Crmp4-/- mice revealed that CRMP4 functions in the proximal axon segment to promote the regrowth of severed DRG neurons and in the distal axon segment where it facilitates Wallerian degeneration through calpain-dependent formation of harmful CRMP4 fragments. These findings reveal an interesting dual role for CRMP4 in proximal and distal axon segments of injured sensory neurons that coordinately facilitate PNS axon regeneration.


Asunto(s)
Traumatismos de los Nervios Periféricos , Degeneración Walleriana , Animales , Axones , Femenino , Ganglios Espinales , Masculino , Ratones , Proteínas Musculares , Regeneración Nerviosa , Nervio Ciático , Semaforina-3A
14.
J Neuropathol Exp Neurol ; 78(4): 348-364, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30863858

RESUMEN

Luman/CREB3 is an important early retrograde axotomy signal regulating acute axon outgrowth in sensory neurons through the adaptive unfolded protein response. As the injury response is transcriptionally multiphasic, a spatiotemporal analysis of Luman/CREB3 localization in rat dorsal root ganglion (DRG) with unilateral L4-L6 spinal nerve injury was conducted to determine if Luman/CREB3 expression was similarly regulated. Biphasic alterations in Luman/CREB3 immunofluorescence and nuclear localization occurred in neurons ipsilateral to 1-hour, 1-day, 2-day, 4-day, and 1-week injury, with a largely parallel, but less avid response contralaterally. This biphasic response was not observed at the transcript level. To assess whether changes in neuronal Luman expression corresponded with an altered intrinsic capacity to grow an axon/neurite in vitro, injury-conditioned and contralateral uninjured DRG neurons underwent a 24-hour axon growth assay. Two-day injury-conditioned neurons exhibited maximal outgrowth capacity relative to naïve, declining at later injury-conditioned timepoints. Only neurons contralateral to 1-week injury exhibited significantly higher axon growth capacity than naïve. In conclusion, alterations in neuronal injury-associated Luman/CREB3 expression support that a multiphasic cell body response occurs and reveal a novel contralateral plasticity in axon growth capacity at 1-week post-injury. These adaptive responses have the potential to inform when repair or therapeutic intervention may be most effective.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Lateralidad Funcional/fisiología , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Axones/metabolismo , Axotomía , Ganglios Espinales/metabolismo , Masculino , Neuritas/metabolismo , Ratas , Ratas Wistar
15.
Exp Neurol ; 315: 60-71, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30731076

RESUMEN

Peripheral nerve regeneration following injury is often incomplete, resulting in significant personal and socioeconomic costs. Although a conditioning crush lesion prior to surgical nerve transection and repair greatly promotes nerve regeneration and functional recovery, feasibility and ethical considerations have hindered its clinical applicability. In a recent proof of principle study, we demonstrated that conditioning electrical stimulation (CES) had effects on early nerve regeneration, similar to that seen in conditioning crush lesions (CCL). To convincingly determine its clinical utility, establishing the effects of CES on target reinnervation and functional outcomes is of utmost importance. In this study, we found that CES improved nerve regeneration and reinnervation well beyond that of CCL. Specifically, compared to CCL, CES resulted in greater intraepidermal skin and NMJ reinnervation, and greater physiological and functional recovery including mechanosensation, compound muscle action potential on nerve conduction studies, normalization of gait pattern, and motor performance on the horizontal ladder test. These findings have direct clinical relevance as CES could be delivered at the bedside before scheduled nerve surgery.


Asunto(s)
Terapia por Estimulación Eléctrica , Regeneración Nerviosa , Potenciales de Acción , Animales , Marcha , Masculino , Compresión Nerviosa , Conducción Nerviosa , Unión Neuromuscular/patología , Traumatismos de los Nervios Periféricos/patología , Desempeño Psicomotor , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Sensación , Piel/inervación
16.
J Neuropathol Exp Neurol ; 67(3): 212-22, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18344912

RESUMEN

Misdirection of axons after nerve injury impairs successful regeneration of adult neurons. Investigations of axon guidance in development have provided an understanding of pathfinding, but their relevance to regenerating adult axons is unclear. We investigated adult mammalian axon guidance during regeneration after peripheral nerve injury and focused on the effects of the prototypic guidance molecule nerve growth factor (NGF). Adult rat sensory neurons from dorsal root ganglia that expressed the NGF receptor tropomyosin-related kinase A (trkA) were presented with a point source of NGF in vitro. Naive trkA neurons had no net turning response to NGF, but if they had been preconditioned by a peripheral nerve transection in vivo before culturing, their growth cones were attracted toward the NGF gradient. A laminin substrate was required for this behavior and an anti-trkA antibody interrupted turning. These data demonstrate that injured adult mammalian axons can be guided as they regenerate. Moreover, despite the downregulation of trkA mRNA and protein levels within the dorsal root ganglion after injury, sensory neurons retain and increase trkA protein at the injury site where the regenerating axons are found. This may enhance the axonal response to NGF and allow guidance along an NGF gradient created in vivo in the distal nerve stump.


Asunto(s)
Ganglios Espinales/metabolismo , Conos de Crecimiento/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Regeneración Nerviosa/fisiología , Neuronas Aferentes/metabolismo , Traumatismos de los Nervios Periféricos , Nervios Periféricos/metabolismo , Animales , Anticuerpos/farmacología , Axotomía , Bioensayo , Comunicación Celular/fisiología , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/ultraestructura , Laminina/metabolismo , Laminina/farmacología , Masculino , Factor de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Neuronas Aferentes/citología , Neuronas Aferentes/efectos de los fármacos , Nervios Periféricos/fisiopatología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkA/efectos de los fármacos , Receptor trkA/genética , Receptor trkA/metabolismo
17.
PLoS One ; 13(5): e0197486, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29775479

RESUMEN

One of the most promising approaches to improve recovery after spinal cord injury (SCI) is the augmentation of spontaneously occurring plasticity in uninjured neural pathways. Acute intermittent hypoxia (AIH, brief exposures to reduced O2 levels alternating with normal O2 levels) initiates plasticity in respiratory systems and has been shown to improve recovery in respiratory and non-respiratory spinal systems after SCI in experimental animals and humans. Although the mechanism by which AIH elicits its effects after SCI are not well understood, AIH is known to alter protein expression in spinal neurons in uninjured animals. Here, we examine hypoxia- and plasticity-related protein expression using immunofluorescence in spinal neurons in SCI rats that were treated with AIH combined with motor training, a protocol which has been demonstrated to improve recovery of forelimb function in this lesion model. Specifically, we assessed protein expression in spinal neurons from animals with incomplete cervical SCI which were exposed to AIH treatment + motor training either for 1 or 7 days. AIH treatment consisted of 10 episodes of AIH: (5 min 11% O2: 5 min 21% O2) for 7 days beginning at 4 weeks post-SCI. Both 1 or 7 days of AIH treatment + motor training resulted in significantly increased expression of the transcription factor hypoxia-inducible factor-1α (HIF-1α) relative to normoxia-treated controls, in neurons both proximal (cervical) and remote (lumbar) to the SCI. All other markers examined were significantly elevated in the 7 day AIH + motor training group only, at both cervical and lumbar levels. These markers included vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), and phosphorylated and nonphosphorylated forms of the BDNF receptor tropomyosin-related kinase B (TrkB). In summary, AIH induces plasticity at the cellular level after SCI by altering the expression of major plasticity- and hypoxia-related proteins at spinal regions proximal and remote to the SCI. These changes occur under the same AIH protocol which resulted in recovery of limb function in this animal model. Thus AIH, which induces plasticity in spinal circuitry, could also be an effective therapy to restore motor function after nervous system injury.


Asunto(s)
Vértebras Cervicales/fisiopatología , Hipoxia/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal , Neuronas/metabolismo , Neuronas/patología , Recuperación de la Función , Traumatismos de la Médula Espinal/fisiopatología , Enfermedad Aguda , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Vértebras Cervicales/patología , Colina O-Acetiltransferasa/metabolismo , Sustancia Gris/patología , Sustancia Gris/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Vértebras Lumbares/patología , Vértebras Lumbares/fisiopatología , Masculino , Actividad Motora , Ratas Endogámicas Lew , Receptor trkB/metabolismo , Traumatismos de la Médula Espinal/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Neurosci ; 25(3): 758-67, 2005 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-15659614

RESUMEN

Neurotrophin-3 (NT-3) negatively modulates nerve growth factor (NGF) receptor expression and associated nociceptive phenotype in intact neurons, suggesting a beneficial role in treating aspects of neuropathic pain mediated by NGF. We report that NT-3 is effective at suppressing thermal hyperalgesia associated with chronic constriction injury (CCI); however, NT-3 does not alter the mechanical hypersensitivity that also develops with CCI. Thermal hyperalgesia is critically linked to expression and activation of the capsaicin receptor, transient receptor potential vanilloid receptor-1 (TRPV1). Thus, its modulation by NT-3 after CCI was examined. CCI results in elevated TRPV1 expression at both the mRNA and protein levels in predominantly small-to-medium neurons, with the percentage of neurons expressing TRPV1 remaining unchanged at approximately 56%. Attenuation of thermal hyperalgesia mediated by NT-3 correlates with decreased TRPV1 expression such that only approximately 26% of neurons ipsilateral to CCI expressed detectable TRPV1 mRNA. NT-3 effected a decrease in expression of the activated component of the signaling pathway linked to regulation of TRPV1 expression, phospho-p38 MAPK (Ji et al., 2002), in neurons ipsilateral to CCI. Exogenous NT-3 could both prevent the onset of thermal hyperalgesia and reverse established thermal hyperalgesia and elevated TRPV1 expression 1 week after CCI. Continuous infusion is required for suppression of both thermal hyperalgesia and TRPV1 expression, because removal of NT-3 resulted in a prompt reestablishment of the hyperalgesic state and corresponding CCI-associated TRPV1 phenotype. In conclusion, although NGF drives inflammation-associated thermal hyperalgesia via its regulation of TRPV1 expression, NT-3 is now identified as a potent negative modulator of this state.


Asunto(s)
Calor , Hiperalgesia/fisiopatología , Canales Iónicos/biosíntesis , Neuronas Aferentes/efectos de los fármacos , Neurotrofina 3/farmacología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Constricción , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/metabolismo , Infusiones Parenterales , Masculino , Neuronas Aferentes/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Ratas , Ratas Wistar , Canales Catiónicos TRPV , Tacto , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Mol Neurosci ; 55(2): 347-54, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24894591

RESUMEN

Human Luman/CREB3 is a basic leucine zipper transcription factor involved in regulation of the unfolded protein response, dendritic cell maturation, and cell migration. But despite reported expression in primary sensory neurons, little is known about its role in the nervous system. To begin investigations into its role in the adult rat nervous system, the rat Luman/CREB3 coding sequence was isolated so its expression within the nervous system could be determined. The rat Luman/CREB3 clone contains a full-length open reading frame encoding 387 amino acids. The recombinant protein generated from this clone activated transcription in a manner equivalent to human Luman/CREB3 from a CAT reporter plasmid construct containing the unfolded protein response element. Quantitative RT-PCR revealed that rat Luman/CREB3 transcripts in a variety of rat tissues with the highest levels in nervous system tissue. In situ hybridization performed on tissue sections confirmed the findings and demonstrated that the Luman/CREB3 mRNA hybridization signal localizes to neurons and satellite glial cells in dorsal root ganglia, the cytoplasm of hepatocytes in liver, and the hippocampal pyramidal cell layers of CA1 and CA3 and the granular cell layer of the dentate gyrus. Collectively, these findings support a role for Luman/CREB3 in the regulation of nervous system function.


Asunto(s)
Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/química , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ganglios Espinales/metabolismo , Hígado/metabolismo , Datos de Secuencia Molecular , Especificidad de Órganos , Ratas , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA