Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(5): 101546, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38703766

ABSTRACT

Mutations in SOD1 cause amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by motor neuron (MN) loss. We previously discovered that macrophage migration inhibitory factor (MIF), whose levels are extremely low in spinal MNs, inhibits mutant SOD1 misfolding and toxicity. In this study, we show that a single peripheral injection of adeno-associated virus (AAV) delivering MIF into adult SOD1G37R mice significantly improves their motor function, delays disease progression, and extends survival. Moreover, MIF treatment reduces neuroinflammation and misfolded SOD1 accumulation, rescues MNs, and corrects dysregulated pathways as observed by proteomics and transcriptomics. Furthermore, we reveal low MIF levels in human induced pluripotent stem cell-derived MNs from familial ALS patients with different genetic mutations, as well as in post mortem tissues of sporadic ALS patients. Our findings indicate that peripheral MIF administration may provide a potential therapeutic mechanism for modulating misfolded SOD1 in vivo and disease outcome in ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Macrophage Migration-Inhibitory Factors , Motor Neurons , Superoxide Dismutase-1 , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Humans , Motor Neurons/metabolism , Motor Neurons/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Mice , Induced Pluripotent Stem Cells/metabolism , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/genetics , Mice, Transgenic , Dependovirus/genetics , Disease Models, Animal , Male , Mutation/genetics , Female , Protein Folding
2.
Neurobiol Dis ; 145: 105052, 2020 11.
Article in English | MEDLINE | ID: mdl-32827689

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset disease characterized by the progressive death of motoneurons and denervation of muscle fibers. To restore motor function, surviving motoneurons in partially denervated muscles typically sprout axons to reinnervate denervated endplates. However, studies on the SOD1G93A rodent models of ALS indicate that sprouting is significantly limited in fast, but not slow, twitch muscles after disease onset. This limitation hastens the rate of muscle weakness and loss of motor function. The causes of this limitation are currently unknown. Sprouting could be limited because the SOD1G93A mutation weakens motoneurons making them incapable of expanding their field of innervation. Alternatively, motoneurons may be capable of sprouting, but unable to do so due to the loss of a permissive sprouting environment. To distinguish between the two possibilities, we compared the sprouting capacity of motoneuron subtypes by partially denervating the fast twitch plantaris (composed of type IIa/IIb muscle fibers) and slow twitch soleus muscles (type I/IIa fibers) prior to disease onset and weakening in SOD1G93A and WT mice. We found that only motoneurons innervating the SOD1G93A plantaris had a limited sprouting capacity. This was correlated with the selective loss of terminal Schwann cells (TSCs) at IIb fibers and an increase in macrophage infiltration. Treating SOD1G93A mice with the tyrosine kinase inhibitor, masitinib, significantly reduced infiltration, prevented TSC loss, and increased the sprouting capacity to near normal. These results suggest that TSCs at denervated type IIb muscle fibers are aberrantly targeted by infiltrating macrophages in SOD1G93A mice, and their loss accounts, at least in part, for the compromised sprouting capacity of the largest motoneurons during early stages of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/physiology , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Nerve Regeneration/physiology , Schwann Cells/pathology , Animals , Mice , Mice, Transgenic , Muscle Denervation , Muscle, Skeletal/innervation , Superoxide Dismutase-1/genetics
3.
Exp Neurol ; 327: 113201, 2020 05.
Article in English | MEDLINE | ID: mdl-31953040

ABSTRACT

This study of medial gastrocnemius (MG) muscle and motor units (MUs) after spinal cord hemisection and deafferentation (HSDA) in adult cats, asked 1) whether the absence of muscle atrophy and unaltered contractile speed demonstrated previously in HSDA-paralyzed peroneus longus (PerL) muscles, was apparent in the unloaded HSDA-paralyzed MG muscle, and 2) how ankle unloading impacts MG muscle and MUs after dorsal root sparing (HSDA-SP) with foot placement during standing and locomotion. Chronic isometric contractile forces and speeds were maintained for up to 12 months in all conditions, but fatigability increased exponentially. MU recordings at 8-11½ months corroborated the unchanged muscle force and speed with significantly increased fatigability; normal weights of MG muscle confirmed the lack of disuse atrophy. Fast MUs transitioned from fatigue resistant and intermediate to fatigable accompanied by corresponding fiber type conversion to fast oxidative (FOG) and fast glycolytic (FG) accompanied by increased GAPDH enzyme activity in absolute terms and relative to oxidative citrate synthase enzyme activity. Myosin heavy chain composition, however, was unaffected. MG muscle behaved like the PerL muscle after HSDA with maintained muscle and MU contractile force and speed but with a dramatic increase in fatigability, irrespective of whether all the dorsal roots were transected. We conclude that reduced neuromuscular activity accounts for increased fatigability but is not, in of itself, sufficient to promote atrophy and slow to fast conversion. Position and relative movements of hindlimb muscles are likely contributors to sustained MG muscle and MU contractile force and speed after HSDA and HSDA-SP surgeries.


Subject(s)
Muscle Fatigue/physiology , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Paralysis/physiopathology , Spinal Cord Injuries/physiopathology , Animals , Cats , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Paralysis/pathology , Spinal Cord Injuries/pathology
4.
J Neurophysiol ; 117(4): 1683-1689, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28148646

ABSTRACT

Motoneurons derived from embryonic stem cells can be transplanted in the tibial nerve, where they extend axons to functionally innervate target muscle. Here, we studied spontaneous muscle contractions in these grafts 3 mo following transplantation. One-half of the transplanted grafts generated rhythmic muscle contractions of variable patterns, either spontaneously or in response to brief electrical stimulation. Activity generated by transplanted embryonic stem cell-derived neurons was driven by glutamate and was modulated by muscarinic and GABAergic/glycinergic transmission. Furthermore, rhythmicity was promoted by the same transmitter combination that evokes rhythmic locomotor activity in spinal cord circuits. These results demonstrate that there is a degree of self-assembly of microcircuits in these peripheral grafts involving embryonic stem cell-derived motoneurons and interneurons. Such spontaneous activity is reminiscent of embryonic circuit development in which spontaneous activity is essential for proper connectivity and function and may be necessary for the grafts to form functional connections with muscle.NEW & NOTEWORTHY This manuscript demonstrates that, following peripheral transplantation of neurons derived from embryonic stem cells, the grafts are spontaneously active. The activity is produced and modulated by a number of transmitter systems, indicating that there is a degree of self-assembly of circuits in the grafts.


Subject(s)
Motor Neurons/physiology , Mouse Embryonic Stem Cells/physiology , Nerve Net/physiology , Nerve Regeneration/physiology , Peripheral Nerve Injuries/surgery , Action Potentials/drug effects , Animals , Electric Stimulation/methods , Embryo, Mammalian , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/transplantation , Muscle, Skeletal/innervation , Neurotransmitter Agents/pharmacology
5.
Ann Clin Transl Neurol ; 3(8): 637-49, 2016 08.
Article in English | MEDLINE | ID: mdl-27606345

ABSTRACT

OBJECTIVE: Nerve injuries resulting in prolonged periods of denervation result in poor recovery of motor function. We have previously shown that embryonic stem cell-derived motoneurons transplanted at the time of transection into a peripheral nerve can functionally reinnervate muscle. For clinical relevance, we now focused on delaying transplantation to assess reinnervation after prolonged denervation. METHODS: Embryonic stem cell-derived motoneurons were transplanted into the distal segments of transected tibial nerves in adult mice after prolonged denervation of 1-8 weeks. Twitch and tetanic forces were measured ex vivo 3 months posttransplantation. Tissue was harvested from the transplants for culture and immunohistochemical analysis. RESULTS: In this delayed reinnervation model, teratocarcinomas developed in about one half of transplants. A residual multipotent cell population (~ 6% of cells) was found despite neural differentiation. Exposure to the alkylating drug mitomycin C eliminated this multipotent population in vitro while preserving motoneurons. Treating neural differentiated stem cells prior to delayed transplantation prevented tumor formation and resulted in twitch and tetanic forces similar to those in animals transplanted acutely after denervation. INTERPRETATION: Despite a neural differentiation protocol, embryonic stem cell-derived motoneurons still carry a risk of tumorigenicity. Pretreating with an antimitotic agent leads to survival and functional muscle reinnervation if performed within 4 weeks of denervation in the mouse.

6.
Nat Commun ; 6: 8506, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26460719

ABSTRACT

Neural prostheses can restore meaningful function to paralysed muscles by electrically stimulating innervating motor axons, but fail when muscles are completely denervated, as seen in amyotrophic lateral sclerosis, or after a peripheral nerve or spinal cord injury. Here we show that channelrhodopsin-2 is expressed within the sarcolemma and T-tubules of skeletal muscle fibres in transgenic mice. This expression pattern allows for optical control of muscle contraction with comparable forces to nerve stimulation. Force can be controlled by varying light pulse intensity, duration or frequency. Light-stimulated muscle fibres depolarize proportionally to light intensity and duration. Denervated triceps surae muscles transcutaneously stimulated optically on a daily basis for 10 days show a significant attenuation in atrophy resulting in significantly greater contractile forces compared with chronically denervated muscles. Together, this study shows that channelrhodopsin-2/H134R can be used to restore function to permanently denervated muscles and reduce pathophysiological changes associated with denervation pathologies.


Subject(s)
Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/therapy , Phototherapy , Animals , Channelrhodopsins , Female , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Skeletal/radiation effects , Random Allocation
7.
J Neurosci ; 35(3): 1291-306, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25609642

ABSTRACT

Induced pluripotent cell-derived motoneurons (iPSCMNs) are sought for use in cell replacement therapies and treatment strategies for motoneuron diseases such as amyotrophic lateral sclerosis (ALS). However, much remains unknown about the physiological properties of iPSCMNs and how they compare with endogenous spinal motoneurons or embryonic stem cell-derived motoneurons (ESCMNs). In the present study, we first used a proteomic approach and compared protein expression profiles between iPSCMNs and ESCMNs to show that <4% of the proteins identified were differentially regulated. Like ESCs, we found that mouse iPSCs treated with retinoic acid and a smoothened agonist differentiated into motoneurons expressing the LIM homeodomain protein Lhx3. When transplanted into the neural tube of developing chick embryos, iPSCMNs selectively targeted muscles normally innervated by Lhx3 motoneurons. In vitro studies showed that iPSCMNs form anatomically mature and functional neuromuscular junctions (NMJs) when cocultured with chick myofibers for several weeks. Electrophysiologically, iPSCMNs developed passive membrane and firing characteristic typical of postnatal motoneurons after several weeks in culture. Finally, iPSCMNs grafted into transected mouse tibial nerve projected axons to denervated gastrocnemius muscle fibers, where they formed functional NMJs, restored contractile force. and attenuated denervation atrophy. Together, iPSCMNs possess many of the same cellular and physiological characteristics as ESCMNs and endogenous spinal motoneurons. These results further justify using iPSCMNs as a source of motoneurons for cell replacement therapies and to study motoneuron diseases such as ALS.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Motor Neurons/cytology , Muscle, Skeletal/cytology , Neurogenesis/physiology , Neuromuscular Junction/cytology , Animals , Axons/physiology , Chick Embryo , LIM-Homeodomain Proteins/metabolism , Mice , Muscle, Skeletal/physiology , Neuromuscular Junction/physiology , Phenotype , Proteomics , Transcription Factors/metabolism
8.
J Neurosci ; 34(32): 10497-510, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25100585

ABSTRACT

The function of neural cell adhesion molecule (NCAM) expression in motor neurons during axonal sprouting and compensatory reinnervation was explored by partially denervating soleus muscles in mice lacking presynaptic NCAM (Hb9(cre)NCAM(flx)). In agreement with previous studies, the contractile force of muscles in wild-type (NCAM(+/+)) mice recovered completely 2 weeks after 75% of the motor innervation was removed because motor unit size increased by 2.5 times. In contrast, similarly denervated muscles in Hb9(cre)NCAM(flx) mice failed to recover the force lost due to the partial denervation because motor unit size did not change. Anatomical analysis indicated that 50% of soleus end plates were completely denervated 1-4 weeks post-partial denervation in Hb9(cre)NCAM(flx) mice, while another 25% were partially reinnervated. Synaptic vesicles (SVs) remained at extrasynaptic regions in Hb9(cre)NCAM(flx) mice rather than being distributed, as occurs normally, to newly reinnervated neuromuscular junctions (NMJs). Electrophysiological analysis revealed two populations of NMJs in partially denervated Hb9(cre)NCAM(flx) soleus muscles, one with high (mature) quantal content, and another with low (immature) quantal content. Extrasynaptic SVs in Hb9(cre)NCAM(flx) sprouts were associated with L-type voltage-dependent calcium channel (L-VDCC) immunoreactivity and maintained an immature, L-VDCC-dependent recycling phenotype. Moreover, acute nifedipine treatment potentiated neurotransmission at newly sprouted NMJs, while chronic intraperitoneal treatment with nifedipine during a period of synaptic consolidation enhanced functional motor unit expansion in the absence of presynaptic NCAM. We propose that presynaptic NCAM bridges a critical link between the SV cycle and the functional expansion of synaptic territory through the regulation of L-VDCCs.


Subject(s)
Motor Neurons/cytology , Muscle Denervation , Nerve Regeneration/physiology , Neural Cell Adhesion Molecules/metabolism , Neuromuscular Junction/physiology , Presynaptic Terminals/physiology , Animals , CD24 Antigen/genetics , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Cholera Toxin/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Motor Neurons/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Nerve Regeneration/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neuromuscular Junction/genetics , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Transcription Factors/genetics , omega-Agatoxin IVA/pharmacology
9.
PLoS One ; 9(3): e91643, 2014.
Article in English | MEDLINE | ID: mdl-24626225

ABSTRACT

Pluripotent stem cells can be directed to differentiate into motor neurons and assessed for functionality in vitro. An emerging application of this technique is to model genetically inherited diseases in differentiated motor neurons and to screen for new therapeutic targets. The neuromuscular junction (NMJ) is essential to the functionality of motor neurons and its dysfunction is a primary hallmark of motor neuron disease. However, mature NMJs that possess the functional and morphological characteristics of those formed in vivo have so far not been obtained in vitro. Here we describe the generation and analysis of mature NMJs formed between embryonic stem cell-derived motor neurons (ESCMNs) and primary myotubes. We compared the formation and maturation of NMJs generated by wild-type (NCAM+/+) ESCMNs to those generated by neural cell adhesion molecule null (NCAM-/-) ESCMNs in order to definitively test the sensitivity of this assay to identify synaptic pathology. We find that co-cultures using NCAM-/- ESCMNs replicate key in vivo NCAM-/- phenotypes and reveal that NCAM influences neuromuscular synaptogenesis by controlling the mode of synaptic vesicle endocytosis. Further, we could improve synapse formation and function in NCAM-/- co-cultures by chronic treatment with nifedipine, which blocks an immature synaptic vesicle recycling pathway. Together, our results demonstrate that this ESCMN/myofiber co-culture system is a highly sensitive bioassay for examining molecules postulated to regulate synaptic function and for screening therapeutics that will improve the function of compromised NMJs.


Subject(s)
Biological Assay/methods , Neuromuscular Junction/pathology , Stem Cells/cytology , Animals , CD56 Antigen/metabolism , Calcium Channels/metabolism , Cell Adhesion Molecules , Cell Differentiation , Chick Embryo , Coculture Techniques , Electrophysiological Phenomena , Embryonic Stem Cells/cytology , Endocytosis , Female , Mice , Mice, Transgenic , Motor Neurons/cytology , Muscle Fibers, Skeletal/metabolism , Nifedipine/chemistry , Phenotype , Pluripotent Stem Cells/cytology , Synapses/metabolism
10.
Prog Brain Res ; 201: 313-31, 2012.
Article in English | MEDLINE | ID: mdl-23186721

ABSTRACT

Alpha motor neurons (also known as lower or skeletal motor neurons) have been studied extensively for over 100 years. Motor neurons control the contraction of skeletal muscles and thus are the final common pathway in the nervous system responsible for motor behavior. Muscles become paralyzed when their innervating motor neurons die because of injury or disease. Motor neuron diseases (MNDs), such as Amyotrophic Lateral Sclerosis, progressively destroy motor neurons until those inflicted succumb to the illness due to respiratory failure. One strategy being explored to study and treat muscle paralysis due to motor neuron loss involves deriving surrogate motor neurons from pluripotent stem cells. Guided by decades of research on the development of the spinal cord, recent advances in neurobiology have shown that functional motor neurons can be derived from mouse and human embryonic stem (ES) cells. Furthermore, ES cell-derived motor neurons restore motor behavior when transplanted into animal models of motor dysfunction. The recent discovery that mouse and human motor neurons can be derived from induced pluripotent stem (iPS) cells (i.e., somatic cells converted to pluripotency) has set the stage for the development of patient-specific therapies designed to treat movement disorders. Indeed, there is now hope within the scientific community that motor neurons derived from pluripotent stem cells will be used to treat MNDs through cell transplantation and/or to screen molecules that will prevent motor neuron death. In this chapter, we review the journey that led to the generation of motor neurons from ES and iPS cells, how stem cell-derived motor neurons have been used to treat/study motor dysfunction, and where the technology will likely lead to in the future.


Subject(s)
Cell Differentiation/physiology , Motor Neurons/physiology , Pluripotent Stem Cells/physiology , Animals , Disease Models, Animal , Humans , Motor Neuron Disease/surgery , Pluripotent Stem Cells/transplantation
11.
Cell Stem Cell ; 9(3): 205-18, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21852222

ABSTRACT

The mammalian nervous system comprises many distinct neuronal subtypes, each with its own phenotype and differential sensitivity to degenerative disease. Although specific neuronal types can be isolated from rodent embryos or engineered from stem cells for translational studies, transcription factor-mediated reprogramming might provide a more direct route to their generation. Here we report that the forced expression of select transcription factors is sufficient to convert mouse and human fibroblasts into induced motor neurons (iMNs). iMNs displayed a morphology, gene expression signature, electrophysiology, synaptic functionality, in vivo engraftment capacity, and sensitivity to degenerative stimuli similar to those of embryo-derived motor neurons. We show that the converting fibroblasts do not transit through a proliferative neural progenitor state, and thus form bona fide motor neurons via a route distinct from embryonic development. Our findings demonstrate that fibroblasts can be converted directly into a specific differentiated and functional neural subtype, the spinal motor neuron.


Subject(s)
Electrical Synapses/metabolism , Fibroblasts/metabolism , Motor Neurons/metabolism , Spinal Cord/metabolism , Transcription Factors/metabolism , Animals , Cell Transdifferentiation , Cells, Cultured , Chick Embryo , Electrical Synapses/pathology , Electrophysiology , Embryonic Development , Fibroblasts/pathology , Fibroblasts/transplantation , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/pathology , Mice , Motor Neurons/pathology , Neuronal Plasticity , Spinal Cord/embryology , Spinal Cord/pathology , Stem Cell Transplantation , Transcription Factors/genetics , Transgenes/genetics
12.
J Neurosci ; 30(19): 6595-606, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463222

ABSTRACT

Identification of intracellular signaling pathways necessary for appropriate axon guidance is challenging because many CNS populations used to study these events contain multiple cell types. Here, we resolve this issue by using mouse embryonic stem (ES) cells that were directed to differentiate into a population of motoneurons that exclusively innervate epaxial muscles [medial median motor column (MMCm) motoneurons]. These ES cell-derived MMCm motoneurons, like their endogenous counterparts, express fibroblast growth factor receptor 1 (FGFR1) and selectively extend axons toward the epaxial trophin FGF8. Unlike wild-type MMCm motoneurons, FGFR1(-/-) MMCm motoneurons show guidance defects when transplanted into the neural tube of chick embryos. Furthermore, activation of FGFR1 selectively signals through mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) for appropriate guidance in vitro, whereas overexpression of constitutively active MAPK/ERK in transplanted, or endogenous chick, MMCm cells causes guidance defects in vivo. These results indicate that MAPK/ERK activation downstream of FGFR1 is necessary for MMCm motor axon guidance and that ES cell-derived neurons provide an important tool for dissecting intracellular pathways required for axon guidance.


Subject(s)
Axons/physiology , Cell Movement/physiology , MAP Kinase Signaling System , Motor Neurons/physiology , Posture/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cells, Cultured , Chick Embryo , Embryonic Stem Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Extremities/growth & development , Extremities/innervation , Extremities/physiology , Fibroblast Growth Factor 8/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Neural Tube/growth & development , Neural Tube/physiology , Receptor, Fibroblast Growth Factor, Type 1/deficiency , Receptor, Fibroblast Growth Factor, Type 1/genetics , Stem Cell Transplantation
13.
Eur J Neurosci ; 31(2): 238-49, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20074227

ABSTRACT

Studies examining the etiology of motoneuron diseases usually focus on motoneuron death as the defining pathophysiology of the disease. However, impaired neuromuscular transmission and synapse withdrawal often precede cell death, raising the possibility that abnormalities in synaptic function contribute to disease onset. Although little is known about the mechanisms maintaining the synaptic integrity of neuromuscular junctions (NMJs), Drosophila studies suggest that Fasciclin II plays an important role. Inspired by these studies we used a reinnervation model of synaptogenesis to analyze neuromuscular function in mice lacking neural cell adhesion molecule (NCAM), the Fasciclin II vertebrate homolog. Our results showed that the recovery of contractile force was the same in wild-type and NCAM-/- mice at 1 month after nerve injury, indicating that endplates were appropriately reformed. This normality was only transient because the contractile force and myofiber number decreased at 3 months after injury in NCAM-/- mice. Both declined further 3 months later. Myofibers degenerated, not because motoneurons died but because synapses were withdrawn. Although neurotransmission was initially normal at reinnervated NCAM-/- NMJs, it was significantly compromised 3 months later. Interestingly, the selective ablation of NCAM from motoneurons, or muscle fibers, did not mimic the deficits observed in reinnervated NCAM-/- mice. Taken together, these results indicate that NCAM is required to maintain normal synaptic function at reinnervated NMJs, although its loss pre-synaptically or post-synaptically is not sufficient to induce synaptic destabilization. Consideration is given to the role of NCAM in terminal Schwann cells for maintaining synaptic integrity and how NCAM dysfunction may contribute to motoneuron disorders.


Subject(s)
Nerve Regeneration/physiology , Neural Cell Adhesion Molecules/metabolism , Neuromuscular Junction/physiology , Animals , Electromyography , Electrophysiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/physiology , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Neural Cell Adhesion Molecules/genetics , Signal Transduction/physiology , Synaptic Transmission/physiology
14.
Eur J Neurosci ; 30(11): 2190-204, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-20128854

ABSTRACT

Recent evidence suggests that synaptic plasticity occurs during homeostatic processes, including sleep-wakefulness regulation, although the underlying mechanisms are not well understood. Polysialylated neural cell adhesion molecule (PSA NCAM) is a transmembrane protein that has been implicated in various forms of plasticity. To investigate whether PSA NCAM is involved in the neuronal plasticity associated with spontaneous sleep-wakefulness regulation and sleep homeostasis, four studies were conducted using rats. First, we showed that PSA NCAM immunoreactivity is present in close proximity to key neurons in several nuclei of the sleep-wakefulness system, including the tuberomammillary hypothalamic nucleus, dorsal raphe nucleus, and locus coeruleus. Second, using western blot analysis and densitometric image analysis of immunoreactivity, we found that 6 h of sleep deprivation changed neither the levels nor the general location of PSA NCAM in the sleep-wakefulness system. Finally, we injected endoneuraminidase (Endo N) intracerebroventricularly to examine the effects of polysialic acid removal on sleep-wakefulness states and electroencephalogram (EEG) slow waves at both baseline and during recovery from 6 h of sleep deprivation. Endo N-treated rats showed a small but significant decrease in baseline rapid eye movement (REM) sleep selectively in the late light phase, and a facilitated REM sleep rebound after sleep deprivation, as compared with saline-injected controls. Non-REM sleep and wakefulness were unaffected by Endo N. These results suggest that PSA NCAM is not particularly involved in the regulation of wakefulness or non-REM sleep, but plays a role in the diurnal pattern of REM sleep as well as in some aspects of REM sleep homeostasis.


Subject(s)
Brain/metabolism , Gene Expression Regulation/physiology , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Sleep, REM/physiology , Animals , Electroencephalography/methods , Electromyography/methods , Gene Expression Regulation/drug effects , Glycoside Hydrolases/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular/methods , Male , Rats , Rats, Wistar , Sleep Deprivation/metabolism , Sleep Deprivation/pathology , Sleep, REM/drug effects , Statistics, Nonparametric , Time Factors , Wakefulness/physiology
15.
J Neurosci ; 28(47): 12409-18, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-19020033

ABSTRACT

Prolonged muscle denervation resulting from motor neuron (MN) damage leads to atrophy and degeneration of neuromuscular junctions (NMJs), which can impart irreversible damage. In this study, we ask whether transplanted embryonic stem (ES) cells differentiated into MNs can form functional synapses with host muscle, and if so what effects do they have on the muscle. After transplantation into transected tibial nerves of adult mice, ES-cell-derived MNs formed functional synapses with denervated host muscle, which resulted in the ability to produce average tetanic forces of 44% of nonlesioned controls. ES-cell-derived motor units (MUs) had mean force values and ranges similar to control muscles. The number of type I fibers and fatigue resistance of the MUs were increased, and denervation-associated muscle atrophy was significantly reduced. These results demonstrate the capacity for ES-cell-derived MNs not only to incorporate into the adult host tissue, but also to exert changes in the target tissue. By providing the signals normally active during embryonic development and placing the cells in an environment with their target tissue, ES cells differentiate into MNs that give rise to functional MU output which resembles the MU output of endogenous MNs. This suggests that these signals combined with those present in the graft environment, lead to the activation of a program intended to produce a normal range of MU forces.


Subject(s)
Embryonic Stem Cells/physiology , Motor Neurons/physiology , Muscular Atrophy/therapy , Nerve Regeneration/physiology , Stem Cell Transplantation/methods , Animals , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Electric Stimulation/methods , Electromyography/methods , Embryo, Mammalian , Embryonic Stem Cells/drug effects , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Muscle Contraction/physiology , Muscle Contraction/radiation effects , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/radiation effects , Muscular Atrophy/pathology , Myosins/metabolism , Nerve Regeneration/radiation effects , Neuromuscular Junction/physiopathology
16.
Brain ; 131(Pt 6): 1492-505, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18334536

ABSTRACT

Despite advances in microsurgical techniques, recovery of motor function after peripheral nerve injury is often poor because many regenerating axons reinnervate inappropriate targets. Consequently, surgical repair must include treatment strategies that improve motor axon targeting. Development of such treatments will require a better understanding of the molecular mechanisms governing selective motor axon targeting. This study used a well-established model of nerve transection and repair to examine (1) whether intrinsic differences exist between different pools of motoneurons after peripheral nerve injury, (2) if such differences regulate selective axon targeting, (3) if regenerating motor axons must express polysialic acid (PSA) in order to preferentially reinnervate muscle and (4) whether brief electrical stimulation improves regeneration accuracy because it increases PSA expression on regenerating axons. We found that different motor pools differentially express PSA after injury and that the capacity to re-express PSA appears to be an intrinsic neuronal property established during development. Second, motoneuron pools not up-regulating PSA did not preferentially reinnervate muscle after injury. Third, brief electrical stimulation of the proximal nerve stump immediately after injury only improved selective motor axon targeting if the motoneurons were capable of up-regulating PSA. Finally, the benefits of stimulation were completely abolished if PSA was removed from the regenerating axons. These results indicate that (1) intrinsic neuronal differences between motor pools must be considered in the development of treatments designed to improve axon targeting and (2) therapeutics aimed at increasing PSA levels on regenerating motor axons may lead to superior functional outcomes.


Subject(s)
Axons/physiology , Femoral Nerve/injuries , Motor Neurons/physiology , Nerve Regeneration/physiology , Animals , Axotomy , Electric Stimulation , Femoral Nerve/pathology , Femoral Nerve/surgery , Male , Mice , Mice, Inbred C57BL , Models, Animal , Motor Neurons/metabolism , Neural Cell Adhesion Molecules/metabolism , Sialic Acids/genetics , Sialic Acids/metabolism , Up-Regulation
17.
Stem Cells ; 25(7): 1697-706, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17395777

ABSTRACT

Directing embryonic stem (ES) cells to differentiate into functional motoneurons has proven to be a strong technique for studying neuronal development as well as being a potential source of tissue for cell replacement therapies involving spinal cord disorders. Unfortunately, one of the mitogenic factors (i.e., sonic hedgehog agonist) used for directed differentiation is not readily available, and thus this technique has not been widely accessible. Here, we present a novel and simple method to derive motoneurons from ES cells using readily attainable reagents. ES cells were derived from a mouse in which enhanced green fluorescent protein (eGFP) was linked to a motoneuron specific promoter. The cells were plated onto a monolayer of 293 EcR-Shh cells that carry an integrated construct for the expression of sonic hedgehog (Shh) under ecdysone-inducible control. To initiate motoneuron differentiation, 293 EcR-Shh:ES cell cocultures were treated with ponasterone A (PA) and retinoic acid for 5 days. PA induces ecdysone, and thus drives Shh expression. To assess differentiation, putative ES cell-derived motoneurons were studied immunocytochemically and cultured on chick myotubes for functional analysis. We found that ES cells differentiated into eGFP+ cells that expressed transcription factors typical of motoneurons. Furthermore, ES cell-derived motoneurons were capable of forming functional connections with muscle fibers in vitro. Finally, when transplanted into the developing chick spinal cord, ES cell-derived motoneurons migrated to the ventral horn and projected axons to appropriate muscle targets. In summary, this simple treatment paradigm produces functional motoneurons that can be used for both developmental and preclinical studies. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Hedgehog Proteins/metabolism , Motor Neurons/cytology , Animals , Axons/drug effects , Axons/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Separation , Cells, Cultured , Chickens , Coculture Techniques , Embryonic Stem Cells/drug effects , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , LIM-Homeodomain Proteins , Mice , Motor Neurons/drug effects , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Receptors, Cholinergic/metabolism , Receptors, Steroid/metabolism , Stem Cell Transplantation , Transcription Factors , Tretinoin/pharmacology
18.
J Neurosci ; 26(12): 3256-68, 2006 Mar 22.
Article in English | MEDLINE | ID: mdl-16554476

ABSTRACT

Embryonic stem (ES) cells differentiate into functional motoneurons when treated with a sonic hedgehog (Shh) agonist and retinoic acid (RA). Whether ES cells can be directed to differentiate into specific subtypes of motoneurons is unknown. We treated embryoid bodies generated from HBG3 ES cells with a Shh agonist and RA for 5 d in culture to induce motoneuron differentiation. Enhanced green fluorescent protein (eGFP) expression was used to identify putative motoneurons, because eGFP is expressed under the control of the Hb9 promoter in HBG3 cells. We found that 96 +/- 0.7% of the differentiated eGFP+ motoneurons expressed Lhx3, a homeobox gene expressed by postmitotic motoneurons in the medial motor column (MMCm), when the treated cells were plated on a neurite-promoting substrate for 5 d. When the treated embryoid bodies were transplanted into stage 17 chick neural tubes, the eGFP+ motoneurons migrated to the MMCm, expressed Lhx3, projected axons to the appropriate target for MMCm motoneurons (i.e., epaxial muscles), and contained synaptic vesicles within intramuscular axonal branches. In ovo and in vitro studies indicated that chemotropic factors emanating from the epaxial muscle and/or surrounding mesenchyme likely guide Lhx3+ motoneurons to their correct target. Finally, whole-cell patch-clamp recordings of transplanted ES cell-derived motoneurons demonstrated that they received synaptic input, elicited repetitive trains of action potentials, and developed passive membrane properties that were similar to host MMCm motoneurons. These results indicate that ES cells can be directed to form subtypes of neurons with specific phenotypic properties.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Motor Neurons/cytology , Pluripotent Stem Cells/cytology , Spinal Cord/cytology , Spinal Cord/embryology , Transcription Factors/metabolism , Action Potentials/genetics , Animals , Body Patterning/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Chemotactic Factors/metabolism , Chick Embryo , Graft Survival/physiology , Green Fluorescent Proteins , Growth Cones/metabolism , Growth Cones/ultrastructure , Hedgehog Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins , Motor Neurons/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/innervation , Neuromuscular Junction/cytology , Neuromuscular Junction/embryology , Neuromuscular Junction/metabolism , Patch-Clamp Techniques , Phenotype , Pluripotent Stem Cells/metabolism , Spinal Cord/metabolism , Stem Cell Transplantation/methods , Trans-Activators/agonists , Trans-Activators/metabolism , Transcription Factors/genetics , Tretinoin/metabolism , Tretinoin/pharmacology
19.
J Neurosci ; 25(8): 2081-91, 2005 Feb 23.
Article in English | MEDLINE | ID: mdl-15728848

ABSTRACT

It is well established that peripheral nerves regenerate after injury. Therefore, incomplete functional recovery usually results from misguided axons rather than a lack of regeneration per se. Despite this knowledge very little is known about the molecular mechanisms regulating axon guidance during regeneration. In the developing neuromuscular system the neural cell adhesion molecule (NCAM) and its polysialic acid (PSA) moiety are essential for proper motor axon guidance. In this study we used a well established model of nerve transection and repair to examine whether NCAM and/or PSA promotes selective regeneration of femoral motor nerves in wild-type and NCAM (-/-) mice. We found that regenerating axons innervating the muscle pathway and, to a lesser extent, cutaneous axons in the sensory pathway reexpress high levels of PSA during the time when the cut axons are crossing the lesion site. Second, we found that motor neurons in wild-type mice preferentially reinnervated muscle pathways, whereas motor neurons in NCAM (-/-) mice reinnervated muscle and cutaneous pathways with equal preference. Preferential regeneration was not observed in wild-type mice when PSA was removed enzymatically from the regenerating nerve, indicating that this form of selective motor axon targeting requires PSA. Finally, transgenic mice were used to show that the number of collateral sprouts, their field of arborization, and the withdrawal of misprojected axons were all attenuated significantly in mice lacking PSA. These results indicate that regenerating motor axons must express polysialylated NCAM, which reduces axon-axon adhesion and enables motor neurons to reinnervate their appropriate muscle targets selectively.


Subject(s)
Axons/physiology , Femoral Nerve/injuries , Motor Neurons/physiology , Nerve Regeneration/physiology , Neural Cell Adhesion Molecules/physiology , Sialic Acids/physiology , Animals , Axotomy , Femoral Nerve/physiology , Glycoside Hydrolases/pharmacology , Glycosylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/innervation , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/deficiency , Neural Cell Adhesion Molecules/genetics , Protein Processing, Post-Translational , Rats , Species Specificity
20.
J Neurosci ; 24(36): 7848-58, 2004 Sep 08.
Article in English | MEDLINE | ID: mdl-15356197

ABSTRACT

The capacity of embryonic stem (ES) cells to form functional motoneurons (MNs) and appropriate connections with muscle was investigated in vitro. ES cells were obtained from a transgenic mouse line in which the gene for enhanced green fluorescent protein (eGFP) is expressed under the control of the promotor of the MN specific homeobox gene Hb9. ES cells were exposed to retinoic acid (RA) and sonic hedgehog agonist (Hh-Ag1.3) to stimulate differentiation into MNs marked by expression of eGFP and the cholinergic transmitter synthetic enzyme choline acetyltransferase. Whole-cell patch-clamp recordings were made from eGFP-labeled cells to investigate the development of functional characteristics of MNs. In voltage-clamp mode, currents, including EPSCs, were recorded in response to exogenous applications of GABA, glycine, and glutamate. EGFP-labeled neurons also express voltage-activated ion channels including fast-inactivating Na(+) channels, delayed rectifier and I(A)-type K(+) channels, and Ca(2+) channels. Current-clamp recordings demonstrated that eGFP-positive neurons generate repetitive trains of action potentials and that l-type Ca(2+) channels mediate sustained depolarizations. When cocultured with a muscle cell line, clustering of acetylcholine receptors on muscle fibers adjacent to developing axons was seen. Intracellular recordings of muscle fibers adjacent to eGFP-positive axons revealed endplate potentials that increased in amplitude and frequency after glutamate application and were sensitive to TTX and curare. In summary, our findings demonstrate that MNs derived from ES cells develop appropriate transmitter receptors, intrinsic properties necessary for appropriate patterns of action potential firing and functional synapses with muscle fibers.


Subject(s)
Motor Neurons/physiology , Pluripotent Stem Cells/cytology , Acetylcholine/pharmacology , Action Potentials/drug effects , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured/physiology , Chick Embryo , Embryo, Mammalian/cytology , Gene Expression Regulation , Genes, Reporter , Glutamic Acid/pharmacology , Glycine/pharmacology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Membrane Potentials , Mice , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/drug effects , Neuromuscular Junction/physiology , Neuromuscular Junction/ultrastructure , Organ Specificity , Organoids/drug effects , Organoids/metabolism , Patch-Clamp Techniques , Phrenic Nerve/embryology , Phrenic Nerve/physiology , Promoter Regions, Genetic , Rats , Tetrodotoxin/pharmacology , Transcription Factors/genetics , Tretinoin/pharmacology , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...