Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Rep ; 16(10): 2723-2735, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568565

ABSTRACT

Developing tissues dictate the amount and type of innervation they require by secreting neurotrophins, which promote neuronal survival by activating distinct tyrosine kinase receptors. Here, we show that nerve growth factor (NGF) signaling through neurotrophic tyrosine kinase receptor type 1 (TrkA) directs innervation of the developing mouse femur to promote vascularization and osteoprogenitor lineage progression. At the start of primary ossification, TrkA-positive axons were observed at perichondrial bone surfaces, coincident with NGF expression in cells adjacent to centers of incipient ossification. Inactivation of TrkA signaling during embryogenesis in TrkA(F592A) mice impaired innervation, delayed vascular invasion of the primary and secondary ossification centers, decreased numbers of Osx-expressing osteoprogenitors, and decreased femoral length and volume. These same phenotypic abnormalities were observed in mice following tamoxifen-induced disruption of NGF in Col2-expressing perichondrial osteochondral progenitors. We conclude that NGF serves as a skeletal neurotrophin to promote sensory innervation of developing long bones, a process critical for normal primary and secondary ossification.


Subject(s)
Femur/blood supply , Femur/innervation , Neovascularization, Physiologic , Nerve Growth Factor/metabolism , Osteogenesis , Receptor, trkA/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction , Animals , Animals, Newborn , Embryo, Mammalian/innervation , Femur/growth & development , Hindlimb/innervation , Mice
2.
J Reconstr Microsurg ; 32(6): 491-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26975563

ABSTRACT

Background Electrical stimulation immediately following nerve lesion helps regenerating axons cross the subsequently grafted nerve repair site. However, the results and the mechanisms remain open to debate. Some findings show that stimulation after crush injury increases axonal crossing of the repair site without affecting regeneration speed. Others show that stimulation after transection and fibrin glue repair doubles regeneration distance. Methods Using a sciatic-nerve-transection-graft in vivo model, we investigated the morphological behavior of regenerating axons around the repair site after unilateral nerve stimulation (20 Hz, 1 hour). With mice expressing axonal fluorescent proteins (thy1-YFP), we were able to calculate the following at 5 and 7 days: percentage of regenerating axons and arborizing axons, branches per axon, and regeneration distance and speed. Results Brief stimulation significantly increases the percentage of regenerating axons (5 days: 35.5 vs. 27.3% nonstimulated, p < 0.05; 7 days: 43.3 vs. 33.9% nonstimulated, p < 0.05), mainly by increasing arborizing axons (5 days: 49.3 [4.4] vs. 33.9 [4.1]% [p < 0.001]; 7 days: 42.2 [5.6] vs. 33.2 [3.1]% [p < 0.001]). Neither branches per arborizing axon nor regeneration speed were affected. Conclusion Our morphological data analysis revealed that electrical stimulation in this model increases axonal crossing of the repair site and promotes homogeneous perilesional branching, but does not affect regeneration speed.


Subject(s)
Axons/physiology , Electric Stimulation , Models, Animal , Nerve Regeneration/physiology , Sciatic Nerve/injuries , Sciatic Nerve/transplantation , Synaptic Transmission/physiology , Animals , Electrophysiology , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Sciatic Nerve/physiology
3.
AJR Am J Roentgenol ; 203(5): 1075-84, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25341148

ABSTRACT

OBJECTIVE: Sports-related peripheral nerve injuries are common among athletes and are often underrecognized because of symptom overlap with more usual sports-related bone, soft-tissue, and joint injuries. CONCLUSION: MRI plays an increasingly important role in the workup of peripheral nerve injuries and may reveal severe nerve abnormalities before they are diagnosed by electrodiagnostic testing or a clinical examination. Sport-specific peripheral nerve injuries and their MRI appearance will be discussed in this article.


Subject(s)
Athletic Injuries/pathology , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Patient Positioning/methods , Peripheral Nerve Injuries/pathology , Peripheral Nerves/pathology , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged
4.
J Neurosci Methods ; 232: 84-92, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-24837281

ABSTRACT

BACKGROUND: Schwann cells in the distal stump of transected nerve upregulate growth factors that support regeneration on a modality-specific basis. It is unclear, however, which of these preferentially support motor axon regeneration. Identification of these factors will require a model that can isolate growth factor effects to growing axons while reproducing the complex three-dimensional structure of peripheral nerve. NEW METHOD: A two-compartment PDMS base is topped by a collagen-coated membrane that supports a spinal cord cross-section above one compartment. Fluorescent motoneurons in this section reinnervate a segment of peripheral nerve that directs axons through a water-tight barrier to the second compartment, where nerve repair is performed. RESULTS: Motoneurons remain healthy for several weeks. The axons they project through the water-tight barrier survive transection and cross a nerve repair in substantial numbers to reinnervate an additional nerve segment. Fluidic isolation of the two compartments was confirmed with a dye leakage test, and the physiologic integrity of the system was tested by retrograde labeling of only those motor neurons to which tracer was exposed and by limitation of toxin effects to a single compartment. COMPARISON WITH EXISTING METHODS: Nerve repair cannot be modeled in monolayer cell culture. Our previous organotypic model accurately modeled nerve repair, but did not allow individual control of motoneuron and growth cone environments. CONCLUSIONS: This model isolates treatment effects to growing axons while reproducing the complex three-dimensional structure of peripheral nerve. Additionally, it facilitates surgical manipulation of tissues and high-resolution imaging.


Subject(s)
Motor Neurons/physiology , Nerve Regeneration/physiology , Organ Culture Techniques , Spinal Cord/cytology , Animals , Animals, Newborn , Axotomy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Collagen , Dextrans , Diffusion Chambers, Culture , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Motor Neurons/drug effects , Nerve Regeneration/drug effects , Nocodazole/pharmacology , Organ Culture Techniques/instrumentation , Organ Culture Techniques/methods , Rhodamines , Tubulin Modulators/pharmacology
5.
J Neurosci ; 34(5): 1689-700, 2014 Jan 29.
Article in English | MEDLINE | ID: mdl-24478351

ABSTRACT

Previous studies demonstrated that Schwann cells (SCs) express distinct motor and sensory phenotypes, which impact the ability of these pathways to selectively support regenerating neurons. In the present study, unbiased microarray analysis was used to examine differential gene expression in denervated motor and sensory pathways in rats. Several genes that were significantly upregulated in either denervated sensory or motor pathways were identified and two secreted factors were selected for further analysis: osteopontin (OPN) and clusterin (CLU) which were upregulated in denervated motor and sensory pathways, respectively. Sciatic nerve transection induced upregulation of OPN and CLU and expression of both returned to baseline levels with ensuing regeneration. In vitro analysis using exogenously applied OPN induced outgrowth of motor but not sensory neurons. CLU, however, induced outgrowth of sensory neurons, but not motor neurons. To assess the functional importance of OPN and CLU, peripheral nerve regeneration was examined in OPN and CLU(-/-) mice. When compared with OPN(+/+) mice, motor neuron regeneration was reduced in OPN(-/-) mice. Impaired regeneration through OPN(-/-) peripheral nerves grafted into OPN(+/+) mice indicated that loss of OPN in SCs was responsible for reduced motor regeneration. Sensory neuron regeneration was impaired in CLU(-/-) mice following sciatic nerve crush and impaired regeneration nerve fibers through CLU(-/-) nerve grafts transplanted into CLU(+/+) mice indicated that reduced sensory regeneration is likely due to SC-derived CLU. Together, these studies suggest unique roles for SC-derived OPN and CLU in regeneration of peripheral motor and sensory axons.


Subject(s)
Clusterin/metabolism , Motor Neurons/physiology , Nerve Regeneration/genetics , Osteopontin/metabolism , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Choline O-Acetyltransferase/genetics , Clusterin/genetics , Denervation , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Nerve Fibers, Myelinated/metabolism , Neural Conduction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Organ Culture Techniques , Osteopontin/genetics , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/surgery , Sensation/genetics , Spinal Cord/cytology , Temperature
6.
Exp Neurol ; 223(1): 112-8, 2010 May.
Article in English | MEDLINE | ID: mdl-19464291

ABSTRACT

The role of pathway-derived growth factors in the support of peripheral axon regeneration remains elusive. Few appropriate knock-out mice are available, and gene silencing techniques are rarely 100% effective. To overcome these difficulties, we have developed an in vitro organotypic co-culture system that accurately models peripheral nerve repair in the adult mammal. Spinal cord sections from P4 mice that express YFP in their neurons are used to innervate segments of P4 peripheral nerve. This reconstructed ventral root is then transected and joined to a nerve graft. Growth of axons across the nerve repair and into the graft can be imaged repeatedly with fluorescence microscopy to define regeneration speed, and parent neurons can be labeled in retrograde fashion to identify contributing neurons. Nerve graft harvested from adult mice remains viable in culture by both morphologic and functional criteria. Motoneurons are supported with GDNF for the first week in culture, after which they survive axotomy, and are thus functionally adult. This platform can be modified by using motoneurons from any genetically modified mouse that can be bred to express XFP, by harvesting nerve graft from any source, or by treating the culture systemically with antibodies, growth factors, or pathway inhibitors. The regeneration environment is controlled to a degree not possible in vivo, and the use of experimental animals is reduced substantially. The flexibility and control offered by this technique should thus make it a useful tool for the study of regeneration biology.


Subject(s)
Models, Animal , Motor Neurons/physiology , Nerve Regeneration/physiology , Animals , Animals, Newborn , Coculture Techniques/methods , Dextrans , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Insulin-Like Growth Factor I/pharmacology , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Motor Neurons/drug effects , Nerve Growth Factor/pharmacology , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Organ Culture Techniques/methods , Peripheral Nerves/physiology , Peripheral Nerves/ultrastructure , Rhodamines , Spinal Cord/cytology
7.
Neurosurgery ; 65(4 Suppl): A132-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19927058

ABSTRACT

OBJECTIVE: Injured peripheral nerves regenerate at very slow rates. Therefore, proximal injury sites such as the brachial plexus still present major challenges, and the outcomes of conventional treatments remain poor. This is in part attributable to a progressive decline in the Schwann cells' ability to provide a supportive milieu for the growth cone to extend and to find the appropriate target. These challenges are compounded by the often considerable delay of regeneration across the site of nerve laceration. Recently, low-frequency electrical stimulation (as brief as an hour) has shown promise, as it significantly accelerated regeneration in animal models through speeding of axon growth across the injury site. METHODS: To test whether this might be a useful clinical tool, we carried out a randomized controlled trial in patients who had experienced substantial axonal loss in the median nerve owing to severe compression in the carpal tunnel. To further elucidate the potential mechanisms, we applied rolipram, a cyclic adenosine monophosphate agonist, to rats after axotomy of the femoral nerve. RESULTS: We demonstrated that effects similar to those observed in animal studies could also be attained in humans. The mechanisms of action of electrical stimulation likely operate through up-regulation of neurotrophic factors and cyclic adenosine monophosphate. Indeed, the application of rolipram significantly accelerated nerve regeneration. CONCLUSION: With new mechanistic insights into the influencing factors of peripheral nerve regeneration, the novel treatments described above could form part of an armament of synergistic therapies that could make a meaningful difference to patients with peripheral nerve injuries.


Subject(s)
Cyclic AMP/agonists , Electric Stimulation Therapy/methods , Growth Cones/drug effects , Nerve Regeneration/drug effects , Peripheral Nerves/drug effects , Peripheral Nervous System Diseases/therapy , Animals , Cyclic AMP/metabolism , Disease Models, Animal , Growth Cones/metabolism , Humans , Nerve Regeneration/physiology , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Rats , Recovery of Function/drug effects , Rolipram/pharmacology , Rolipram/therapeutic use , Treatment Outcome
8.
Exp Neurol ; 205(2): 347-59, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17428474

ABSTRACT

Brief electrical stimulation enhances the regenerative ability of axotomized motor [Nix, W.A., Hopf, H.C., 1983. Electrical stimulation of regenerating nerve and its effect on motor recovery. Brain Res. 272, 21-25; Al-Majed, A.A., Neumann, C.M., Brushart, T.M., Gordon, T., 2000. Brief electrical stimulation promotes the speed and accuracy of motor axonal regeneration. J. Neurosci. 20, 2602-2608] and sensory [Brushart, T.M., Jari, R., Verge, V., Rohde, C., Gordon, T., 2005. Electrical stimulation restores the specificity of sensory axon regeneration. Exp. Neurol. 194, 221-229] neurons. Here we examined the parameter of duration of stimulation on regenerative capacity, including the intrinsic growth programs, of sensory neurons. The effect of 20 Hz continuous electrical stimulation on the number of DRG sensory neurons that regenerate their axons was evaluated following transection and surgical repair of the femoral nerve trunk. Stimulation was applied proximal to the repair site for 1 h, 3 h, 1 day, 7 days or 14 days at the time of nerve repair. Following a 21-day regeneration period, DRG neurons that regenerated axons into the muscle and cutaneous sensory nerve branches were retrogradely identified. Stimulation of 1 h led to a significant increase in DRG neurons regenerating into cutaneous and muscle branches when compared to 0 h (sham) stimulation or longer periods of stimulation. Stimulation for 1 h also significantly increased the numbers of neurons that regenerated axons beyond the repair site 4 days after lesion and was correlated with a significant increase in expression of growth-associated protein 43 (GAP-43) mRNA in the regenerating neurons at 2 days post-repair. An additional indicator of heightened plasticity following 1 h stimulation was elevated expression of brain-derived neurotrophic factor (BDNF). The effect of brief stimulation on enhancing sensory and motoneuron regeneration holds promise for inducing improved peripheral nerve repair in the clinical setting.


Subject(s)
Gene Expression/physiology , Nerve Regeneration/physiology , Neurons, Afferent/physiology , Action Potentials/drug effects , Activating Transcription Factor 3/metabolism , Anesthetics, Local/pharmacology , Animals , Axons/physiology , Brain-Derived Neurotrophic Factor/metabolism , Cell Count , Cell Proliferation , Electric Stimulation , Female , Femoral Nerve/cytology , Femoral Nerve/physiology , GAP-43 Protein/metabolism , Immunohistochemistry , In Situ Hybridization , Perfusion , Rats , Rats, Sprague-Dawley , Tetrodotoxin/pharmacology , Tissue Fixation
9.
Exp Neurol ; 198(2): 500-10, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16460731

ABSTRACT

Functional recovery after peripheral nerve injury is often poor despite high regenerative capacity of peripheral neurons. In search for novel treatments, brief electrical stimulation of the acutely lesioned nerve has recently been identified as a clinically feasible approach increasing precision of axonal regrowth. The effects of this stimulation appear to be mediated by BDNF and its receptor, TrkB, but the down-stream effectors are unknown. A potential candidate is the HNK-1 carbohydrate known to be selectively reexpressed in motor but not sensory nerve branches of the mouse femoral nerve and to enhance growth of motor but not sensory axons in vitro. Here, we show that short-term low-frequency electrical stimulation (1 h, 20 Hz) of the lesioned and surgically repaired femoral nerve in wild-type mice causes a motor nerve-specific enhancement of HNK-1 expression correlating with previously reported acceleration of muscle reinnervation. Such enhanced HNK-1 expression was not observed after electrical stimulation in heterozygous BDNF or TrkB-deficient mice. Accordingly, the degree of proper reinnervation of the quadriceps muscle, as indicated by retrograde labeling of motoneurons, was reduced in TrkB+/- mice compared to wild-type littermates. Also, recovery of quadriceps muscle function, evaluated by a novel single-frame motion analysis approach, and axonal regrowth into the distal nerve stump, assessed morphologically, were considerably delayed in TrkB+/- mice. These findings indicate that BDNF/TrkB signaling is important for functional recovery after nerve repair and suggest that up-regulation of the HNK-1 glycan is linked to this phenomenon.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CD57 Antigens/metabolism , Gene Expression Regulation , Nerve Regeneration/physiology , Receptor, trkB/metabolism , Recovery of Function/physiology , Signal Transduction/physiology , Animals , Axotomy/methods , Brain-Derived Neurotrophic Factor/deficiency , Disease Models, Animal , Electric Stimulation/methods , Femoral Nerve/cytology , Femoral Nerve/physiology , Gene Expression Regulation/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/pathology , Motor Neurons/physiology , Peripheral Nerves/pathology , Peripheral Nerves/physiopathology , Peripheral Nervous System Diseases/physiopathology , Receptor, trkB/deficiency , Time Factors
10.
J Neurosci ; 25(41): 9406-12, 2005 Oct 12.
Article in English | MEDLINE | ID: mdl-16221849

ABSTRACT

Motor axons regenerating after repair of mixed nerve reinnervate pathways leading to muscle more often than those leading to skin [preferential motor reinnervation (PMR)]. Motoneurons that initially project collaterals to both muscle and skin prune incorrect projections to generate specificity. The number of motor axon collaterals maintained entirely within cutaneous or muscle pathways, however, is unknown. To overcome this shortcoming, dorsal root ganglion excision has been used to allow only motor axons to regenerate after a peripheral lesion. Motor axon number in reinnervated cutaneous and muscle pathways can then be correlated with the number of parent motoneurons determined by retrograde labeling. The number of collaterals per neuron can be calculated for each environment and the relative roles of pathway and end organ assessed by blocking the distal pathways to prevent target reinnervation. Without sensory competition, PMR develops in two stages: a limited response to muscle nerve and then a robust response to muscle that may involve retrograde signaling to the proximal pathway. Motoneurons maintain more collaterals in cutaneous nerve than in muscle nerve, even without muscle contact. This difference could result either from increased collateral formation in cutaneous nerve or from increased collateral pruning in muscle nerve. In either instance, these findings confirm that muscle and cutaneous pathways have functionally significant identities that can be recognized by motor axons and can regulate their arborization. Decreased arborization in muscle pathways could promote regeneration by focusing neuronal resources on high-yield projections; increased arborization in cutaneous pathways, conversely, would enhance pathfinding abilities.


Subject(s)
Motor Neurons/physiology , Nerve Regeneration/physiology , Animals , Efferent Pathways/physiology , Female , Femoral Nerve/physiology , Peripheral Nerves/physiology , Rats , Rats, Sprague-Dawley
11.
Exp Neurol ; 194(1): 221-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15899259

ABSTRACT

Electrical stimulation at the time of nerve repair promotes motoneurons to reinnervate appropriate pathways leading to muscle and stimulates sensory neurons to regenerate. The present experiments examine the effects of electrical stimulation on the specificity of sensory axon regeneration. The unoperated rat femoral cutaneous branch is served by 2-3 times more DRG neurons than is the muscle branch. After transection and repair of the femoral trunk, equal numbers of DRG neurons project to both branches. However, 1 h of electrical stimulation restores the normal proportion of DRG neurons reinnervating skin and muscle. To ask if the redistribution of stimulated neurons results from enhanced specificity of target reinnervation, we developed a new technique of sequential double labeling. DRG neurons projecting to the femoral muscle branch were prelabeled with Fluoro Gold 2 weeks before the nerve was transected proximally and repaired with or without 1 h of 20-Hz electrical stimulation. Three weeks after repair, the muscle nerve was labeled a second time with Fluororuby. The percentage of regenerating neurons that both originally served muscle and returned to muscle after nerve repair increased from 40% without stimulation to 75% with stimulation. Electrical stimulation thus dramatically alters the distribution of regenerating sensory axons, replacing normally random behavior with selective reinnervation of tissue-specific targets. If the enhanced regeneration specificity resulting from electrical stimulation is found to improve function in a large animal model, this convenient and safe technique may be a useful adjunct to clinical nerve repair.


Subject(s)
Axons/physiology , Femoral Nerve/physiology , Ganglia, Spinal/physiology , Nerve Regeneration/physiology , Neurons, Afferent/physiology , Animals , Axonal Transport/physiology , Axotomy , Cell Communication/physiology , Dextrans , Disease Models, Animal , Electric Stimulation , Female , Growth Cones/physiology , Muscle, Skeletal/innervation , Rats , Rats, Sprague-Dawley , Rhodamines , Staining and Labeling/methods , Stilbamidines
12.
J Comp Neurol ; 485(3): 183-90, 2005 May 09.
Article in English | MEDLINE | ID: mdl-15791642

ABSTRACT

A century ago, Ramon y Cajal described the generalized response of regenerating peripheral axons to their environment. By using mice that express fluorescent proteins in their axons, we are now able to quantify the response of individual axons to nerve transection and repair. Sciatic nerves from nonexpressing mice were grafted into those expressing a yellow variant of green fluorescent protein, then examined at 5, 7, or 10 days after repair. Regeneration was found to be a staggered process, with only 25% of axons crossing the repair in the first week. In the setting of Wallerian degeneration, this stagger will expose growth cones to an evolving menu of molecular cues upon which to base pathway decisions. Many axons arborize, allowing them to interact simultaneously with several pathways. Arborization could serve as the anatomical substrate for specificity generation through collateral pruning. Axons often travel laterally across the face of the distal stump before choosing a pathway. As a result, the average unbranched axon has access to over 100 distal Schwann cell tubes. This extensive access, however, does not ensure correct matching of axon and end organ, suggesting that pathway choice is made on the basis of factors other than end organ identity. These observations explain the failure of refined surgical techniques to restore normal function after nerve injury. The apparent wandering of axons across the repair defies surgical control and mandates a biological approach to reuniting severed axons with appropriate distal pathways.


Subject(s)
Axons/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Neural Pathways/physiology , Sciatic Nerve/transplantation , Animals , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Neurites/physiology , Sciatic Nerve/cytology , Sciatic Nerve/metabolism
13.
J Peripher Nerv Syst ; 8(2): 91-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12795713

ABSTRACT

Predegeneration of nerve enhances its ability to support axon regeneration. Trophic factors are upregulated by reactive Schwann cells while potentially inhibitory molecules are removed. These experiments isolate the effects of one such inhibitory molecule, the myelin-associated glycoprotein (MAG), to determine its role in modifying regeneration after nerve repair. Suture of the mouse femoral nerve was followed by daily intraperitoneal injection of antibodies to MAG, antibodies to HNK-1, a specific muscle pathway marker, or no further treatment. Regeneration was assayed by double-labeling the femoral cutaneous and muscle branches with horseradish peroxidase and fluoro-gold after 4 weeks or 6 weeks of regeneration. Four weeks after nerve repair, selective reinnervation of the muscle branch by motoneurons, or preferential motor reinnervation (PMR), was not seen in either controls or L2-antibody-treated animals. In contrast, treatment with MAG antibodies resulted in dramatic PMR. By 6 weeks, the controls had achieved borderline specificity, substantial PMR developed in the L2 antibody group and the MAG group changed little. Blocking access to MAG in the distal nerve stump thus accelerated and enhanced PMR. Sensory regeneration was depressed by both antibody treatments at 4 weeks but recovered by 6 weeks. Antibody administration has a generalized effect on sensory regeneration that is unrelated to the behavior of motoneurons in the same nerve.


Subject(s)
Antibodies/administration & dosage , Femoral Nerve/drug effects , Motor Neurons/drug effects , Myelin-Associated Glycoprotein/immunology , Nerve Regeneration/drug effects , Stilbamidines , Animals , Antibodies/physiology , CD57 Antigens/immunology , CD57 Antigens/physiology , Fluorescent Dyes/pharmacokinetics , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Horseradish Peroxidase/pharmacokinetics , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Muscles/metabolism , Myelin-Associated Glycoprotein/physiology , Nerve Regeneration/physiology , Skin/innervation , Skin/metabolism , Time Factors
14.
J Neurosci ; 22(15): 6631-8, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12151542

ABSTRACT

Motoneurons reinnervate the distal stump at variable rates after peripheral nerve transection and suture. In the rat femoral nerve model, reinnervation is already substantial 3 weeks after repair, but is not completed for an additional 7 weeks. However, this "staggered regeneration" can be temporally compressed by application of 20 Hz electrical stimulation to the nerve for 1 hr. The present experiments explore two possible mechanisms for this stimulation effect: (1) synchronization of distal stump reinnervation and (2) enhancement of regeneration speed. The first possibility was investigated by labeling all motoneurons that have crossed the repair at intervals from 4 d to 4 weeks after rat femoral nerve transection and suture. Although many axons did not cross until 3-4 weeks after routine repair, stimulation significantly increased the number crossing at 4 and 7 d, with only a few crossing after 2 weeks. Regeneration speed was studied by radioisotope labeling of transported proteins and by anterograde labeling of regenerating axons, and was not altered by stimulation. Attempts to condition the neuron by stimulating the femoral nerve 1 week before injury were also without effect. Electrical stimulation thus promotes the onset of motor axon regeneration without increasing its speed. This finding suggests a combined approach to improving the outcome of nerve repair, beginning with stimulation to recruit all motoneurons across the repair, followed by other treatments to speed and prolong axonal elongation.


Subject(s)
Electric Stimulation Therapy/methods , Femoral Nerve/physiology , Motor Neurons/physiology , Nerve Regeneration/physiology , Sciatic Nerve/physiology , Animals , Axons/physiology , Axotomy , Female , Ligation , Models, Animal , Nerve Crush , Rats , Rats, Sprague-Dawley , Time
SELECTION OF CITATIONS
SEARCH DETAIL
...