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










Publication year range
1.
bioRxiv ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38853966

ABSTRACT

Astrocytes use Ca 2+ signals to regulate multiple aspects of normal and pathological brain function. Astrocytes display context-specific diversity in their functions, and in their response to noxious stimuli between brain regions. Indeed, astrocytic mitochondria have emerged as key players in governing astrocytic functional heterogeneity, given their ability to dynamically adapt their morphology to regional demands on their ATP generation and Ca 2+ buffering functions. Although there is reciprocal regulation between mitochondrial dynamics and mitochondrial Ca 2+ signaling in astrocytes, the extent of this regulation into the rich diversity of astrocytes in different brain regions remains largely unexplored. Brain-wide, experimentally induced mitochondrial DNA (mtDNA) loss in astrocytes showed that mtDNA integrity is critical for proper astrocyte function, however, few insights into possible diverse responses to this noxious stimulus from astrocytes in different brain areas were reported in these experiments. To selectively damage mtDNA in astrocytes in a brain-region-specific manner, we developed a novel adeno-associated virus (AAV)-based tool, Mito-PstI, which expresses the restriction enzyme PstI, specifically in astrocytic mitochondria. Here, we applied Mito-PstI to two distinct brain regions, the dorsolateral striatum, and the hippocampal dentate gyrus, and we show that Mito-PstI can induce astrocytic mtDNA loss in vivo , but with remarkable brain-region-dependent differences on mitochondrial dynamics, spontaneous Ca 2+ fluxes and astrocytic as well as microglial reactivity. Thus, AAV-Mito-PstI is a novel tool to explore the relationship between astrocytic mitochondrial network dynamics and astrocytic mitochondrial Ca 2+ signaling in a brain-region-selective manner.

2.
Neurosci Lett ; 771: 136468, 2022 02 06.
Article in English | MEDLINE | ID: mdl-35065247

ABSTRACT

Recent RNA-seq studies have generated a new crop of putative gene markers for terminal Schwann cells (tSCs), non-myelinating glia that cap axon terminals at the vertebrate neuromuscular junction (NMJ). While compelling, these studies did not validate the expression of the novel markers using in situ hybridization techniques. Here, we use RNAscope technology to study the expression of top candidates from recent tSC and non-myelinating Schwann cell marker RNA-seq studies. Our results validate the expression of these markers at tSCs but also demonstrate that they are present at other sites in the muscle tissue, specifically, at muscle spindles and along intramuscular nerves.


Subject(s)
Nerve Tissue Proteins/genetics , RNA-Seq/methods , Schwann Cells/metabolism , Animals , Female , In Situ Hybridization, Fluorescence/methods , In Situ Hybridization, Fluorescence/standards , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , RNA-Seq/standards , Reference Standards
3.
Front Mol Neurosci ; 13: 91, 2020.
Article in English | MEDLINE | ID: mdl-32595450
4.
Neurosci Lett ; 715: 134671, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31805372

ABSTRACT

The neuromuscular junction is the synapse between a motor neuron of the spinal cord and a skeletal muscle fiber in the periphery. Reciprocal interactions between these excitable cells, and between them and others cell types present within the muscle tissue, shape the development, homeostasis and plasticity of skeletal muscle. An important aim in the field is to understand the molecular mechanisms underlying these cellular interactions, which include identifying the nature of the signals and receptors involved but also of the downstream intracellular signaling cascades elicited by them. This review focuses on work that shows that skeletal muscle fiber-derived extracellular signal-regulated kinases 1 and 2 (ERK1/2), ubiquitous and prototypical intracellular mitogen-activated protein kinases, have modulatory roles in the maintenance of the neuromuscular synapse and in the acquisition and preservation of fiber type identity in skeletal muscle.


Subject(s)
Cell Communication/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Muscle Fibers, Skeletal/physiology , Neuromuscular Junction/physiology , Animals , Phenotype
5.
Sci Rep ; 9(1): 7799, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31127156

ABSTRACT

Spinal muscular atrophy (SMA) is caused by loss-of-function mutations in the survival of motoneuron gene 1 (SMN1). SMA is characterized by motoneuron death, skeletal muscle denervation and atrophy. Disease severity inversely correlates with copy number of a second gene (SMN2), which harbors a splicing defect that causes the production of inadequate levels of functional SMN protein. Small molecules that modify SMN2 splicing towards increased production of functional SMN significantly ameliorate SMA phenotypes in mouse models of severe SMA. At suboptimal doses, splicing modifiers, such as SMN-C1, have served to generate mice that model milder SMA, referred to as pharmacological SMA mice, which survive into early adulthood. Nerve sprouting at endplates, known as terminal sprouting, is key to normal muscle fiber reinnervation following nerve injury and its promotion might mitigate neuromuscular symptoms in mild SMA. Sprouting has been difficult to study in severe SMA mice due to their short lifespan. Here, we show that pharmacological SMA mice are capable of terminal sprouting following reinnervation that is largely SMN-C1 dose-independent, but that they display a reinnervation delay that is critically SMN-C1 dose-dependent. Data also suggest that SMN-C1 can induce by itself a limited terminal sprouting response in SMA and wild-type normally-innervated endplates.


Subject(s)
Muscle, Skeletal/innervation , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/physiopathology , Animals , Disease Models, Animal , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy, Spinal/chemically induced , Muscular Atrophy, Spinal/pathology , Nerve Regeneration , Neuromuscular Junction/pathology , Schwann Cells/pathology
7.
Sci Rep ; 6: 38745, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934942

ABSTRACT

To test the role of extracellular-signal regulated kinases 1 and 2 (ERK1/2) in slow-twitch, type 1 skeletal muscle fibers, we studied the soleus muscle in mice genetically deficient for myofiber ERK1/2. Young adult mutant soleus was drastically wasted, with highly atrophied type 1 fibers, denervation at most synaptic sites, induction of "fetal" acetylcholine receptor gamma subunit (AChRγ), reduction of "adult" AChRε, and impaired mitochondrial biogenesis and function. In weanlings, fiber morphology and mitochondrial markers were mostly normal, yet AChRγ upregulation and AChRε downregulation were observed. Synaptic sites with fetal AChRs in weanling muscle were ~3% in control and ~40% in mutants, with most of the latter on type 1 fibers. These results suggest that: (1) ERK1/2 are critical for slow-twitch fiber growth; (2) a defective γ/ε-AChR subunit switch, preferentially at synapses on slow fibers, precedes wasting of mutant soleus; (3) denervation is likely to drive this wasting, and (4) the neuromuscular synapse is a primary subcellular target for muscle ERK1/2 function in vivo.


Subject(s)
MAP Kinase Signaling System , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/pathology , Muscular Atrophy , Receptors, Nicotinic/physiology , Animals , Female , Male , Mice , Mice, Knockout , Muscle Fibers, Slow-Twitch/enzymology , Muscle, Skeletal/enzymology , Myosin Heavy Chains/genetics , RNA, Messenger/genetics , Receptors, Nicotinic/genetics
8.
Mol Cell Biol ; 35(7): 1238-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605336

ABSTRACT

The Ras-extracellular signal-regulated kinase 1 and 2 (ERK1/2) pathway appears to be important for the development, maintenance, aging, and pathology of mammalian skeletal muscle. Yet no gene targeting of Erk1/2 in muscle fibers in vivo has been reported to date. We combined a germ line Erk1 mutation with Cre-loxP Erk2 inactivation in skeletal muscle to produce, for the first time, mice lacking ERK1/2 selectively in skeletal myofibers. Animals lacking muscle ERK1/2 displayed stunted postnatal growth, muscle weakness, and a shorter life span. Their muscles examined in this study, sternomastoid and tibialis anterior, displayed fragmented neuromuscular synapses and a mixture of modest fiber atrophy and loss but failed to show major changes in fiber type composition or absence of cell surface dystrophin. Whereas the lack of only ERK1 had no effects on the phenotypes studied, the lack of myofiber ERK2 explained synaptic fragmentation in the sternomastoid but not the tibialis anterior and a decrease in the expression of the acetylcholine receptor (AChR) epsilon subunit gene mRNA in both muscles. A reduction in AChR protein was documented in line with the above mRNA results. Evidence of partial denervation was found in the sternomastoid but not the tibialis anterior. Thus, myofiber ERK1/2 are differentially required for the maintenance of myofibers and neuromuscular synapses in adult mice.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Skeletal/enzymology , Neuromuscular Junction/metabolism , Animals , Female , Gene Deletion , Male , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/physiology , Muscle, Skeletal/ultrastructure , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Receptors, Cholinergic/metabolism
9.
PLoS One ; 8(9): e75866, 2013.
Article in English | MEDLINE | ID: mdl-24086650

ABSTRACT

In the inherited childhood neuromuscular disease spinal muscular atrophy (SMA), lower motor neuron death and severe muscle weakness result from the reduction of the ubiquitously expressed protein survival of motor neuron (SMN). Although SMA mice recapitulate many features of the human disease, it has remained unclear if their short lifespan and motor weakness are primarily due to cell-autonomous defects in motor neurons. Using Hb9(Cre) as a driver, we selectively raised SMN expression in motor neurons in conditional SMAΔ7 mice. Unlike a previous study that used choline acetyltransferase (ChAT(Cre+) ) as a driver on the same mice, and another report that used Hb9(Cre) as a driver on a different line of conditional SMA mice, we found no improvement in survival, weight, motor behavior and presynaptic neurofilament accumulation. However, like in ChAT(Cre+) mice, we detected rescue of endplate size and mitigation of neuromuscular junction (NMJ) denervation status. The rescue of endplate size occurred in the absence of an increase in myofiber size, suggesting endplate size is determined by the motor neuron in these animals. Real time-PCR showed that the expression of spinal cord SMN transcript was sharply reduced in Hb9(Cre+) SMA mice relative to ChAT(Cre+) SMA mice. This suggests that our lack of overall phenotypic improvement is most likely due to an unexpectedly poor recombination efficiency driven by Hb9(Cre) . Nonetheless, the low levels of SMN were sufficient to rescue two NMJ structural parameters indicating that these motor neuron cell autonomous phenotypes are very sensitive to changes in motoneuronal SMN levels. Our results directly suggest that even those therapeutic interventions with very modest effects in raising SMN in motor neurons may provide mitigation of neuromuscular phenotypes in SMA patients.


Subject(s)
Motor Neurons/physiology , Muscular Atrophy, Spinal/physiopathology , Muscular Atrophy, Spinal/therapy , Phenotype , SMN Complex Proteins/metabolism , Synapses/physiology , Animals , DNA Primers/genetics , Genotype , Mice , Motor Endplate/metabolism , Motor Endplate/physiology , Real-Time Polymerase Chain Reaction , Survival Analysis
10.
Dev Biol ; 356(2): 432-44, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21658376

ABSTRACT

A mouse model of the devastating human disease "spinal muscular atrophy" (SMA) was used to investigate the severe muscle weakness and spasticity that precede the death of these animals near the end of the 2nd postnatal week. Counts of motor units to the soleus muscle as well as of axons in the soleus muscle nerve showed no loss of motor neurons. Similarly, neither immunostaining of neuromuscular junctions nor the measurement of the tension generated by nerve stimulation gave evidence of any significant impairment in neuromuscular transmission, even when animals were maintained up to 5days longer via a supplementary diet. However, the muscles were clearly weaker, generating less than half their normal tension. Weakness in 3 muscles examined in the study appears due to a severe but uniform reduction in muscle fiber size. The size reduction results from a failure of muscle fibers to grow during early postnatal development and, in soleus, to a reduction in number of fibers generated. Neuromuscular development is severely delayed in these mutant animals: expression of myosin heavy chain isoforms, the elimination of polyneuronal innervation, the maturation in the shape of the AChR plaque, the arrival of SCs at the junctions and their coverage of the nerve terminal, the development of junctional folds. Thus, if SMA in this particular mouse is a disease of motor neurons, it can act in a manner that does not result in their death or disconnection from their targets but nonetheless alters many aspects of neuromuscular development.


Subject(s)
Motor Neurons/pathology , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/physiology , Synaptic Transmission , Animals , Apoptosis , Mice , Microscopy, Electron , Models, Animal , Motor Neurons/cytology , Motor Neurons/physiology , Muscle Fibers, Skeletal/physiology , Muscular Atrophy, Spinal/pathology , Myosin Heavy Chains/analysis , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Synapses/pathology
11.
Commun Integr Biol ; 4(2): 143-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21655426

ABSTRACT

Information between neurons and the target cells they innervate passes through sites of functional contact called synapses. How synapses form and are altered by sensory or cognitive experience is central to understand nervous system function. Studies of synapse formation and plasticity have concentrated on a few "model" synapses. The vertebrate neuromuscular junction (NMJ), the synapse between a motoneuron in the spinal cord and a skeletal muscle fiber, is one such model synapse. The extracellular matrix proteoglycan agrin plays an essential organizing role at the NMJ. Agrin is also present at some synapses in the brain and in other organs in the periphery, but its function outside the NMJ is unclear. The core signaling pathway for agrin at the NMJ, which is still incompletely defined, includes molecules specifically involved in this cascade and molecules used in other signaling pathways in many cells. Mitogen-activated protein kinases (MAPKs) are evolutionarily conserved components of intracellular signaling modules that control a myriad of cellular processes. This article reviews emerging evidence that suggests that MAPKs are involved in agrin signaling at the NMJ and in the putative functions of agrin in the formation of a subset of synapses in the brain.

12.
J Biol Chem ; 285(42): 32370-7, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20696763

ABSTRACT

Agrin released by motoneurons induces and/or maintains acetylcholine receptor (AChR) clustering and other aspects of postsynaptic differentiation at the vertebrate neuromuscular junction. Agrin acts by binding and activating a receptor complex containing LDL receptor protein 4 (Lrp4) and muscle-specific kinase (MuSK). Two critical downstream components of this signaling cascade, Dox-7 and rapsyn, have been identified. However, additional intracellular essential elements remain unknown. Prior observations by others and us suggested antagonistic interactions between agrin and neuregulin-1 (Nrg-1) signaling in cultured myotubes and developing muscle fibers in vivo. A hallmark of Nrg-1 signaling in skeletal muscle cells is the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). ERK1/2 are also activated in most cells by phorbol 12-myristate 13-acetate, a classical inhibitor of agrin-induced AChR clustering in myotubes. Here, it was investigated whether agrin activates ERK1/2 directly and whether such activation modulates agrin-induced AChR clustering. Agrin induced a rapid but transient activation of ERK1/2 in myotubes that was Lrp4/MuSK-dependent. However, blocking this ERK1/2 activation did not prevent but potentiated AChR clustering induced by agrin. ERK1/2 activation was dispensable for Nrg-1-mediated inhibition of the AChR clustering activity of agrin, but was indispensable for such activity by phorbol 12-myristate 13-acetate. Together, these results suggest agrin-induced activation of ERK1/2 is a negative modulator of agrin signaling in skeletal muscle cells.


Subject(s)
Agrin/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Fibers, Skeletal/metabolism , Receptors, Cholinergic/metabolism , Animals , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/metabolism , Humans , LDL-Receptor Related Proteins , Mice , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Neuregulin-1/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, LDL/metabolism , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/metabolism
13.
Hum Mol Genet ; 19(18): 3614-22, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20603324

ABSTRACT

The neuromuscular disease myotonic dystrophy type I (DM1) affects multiple organ systems with the major symptoms being severe muscle weakness, progressive muscle wasting and myotonia. The causative mutation in DM1 is a CTG repeat expansion in the 3'-untranslated region of the DM protein kinase (DMPK) gene. RNA transcribed from the expanded allele contains the expanded CUG repeats and leads to the nuclear depletion of Muscleblind-like 1 (MBNL1) and to the increased steady-state levels of CUG-binding protein 1 (CUGBP1). The pathogenic effects of MBNL1 depletion have previously been tested by the generation of MBNL1 knockout mice, but the consequence of CUGBP1 overexpression in adult muscle is not known. In a DM1 mouse model expressing RNA containing 960 CUG repeats in skeletal muscle, CUGBP1 up-regulation is temporally correlated with severe muscle wasting. In this study, we generated transgenic mice with doxycycline-inducible and skeletal muscle-specific expression of CUGBP1. Adult mouse skeletal muscle overexpressing CUGBP1 reproduces molecular and physiological defects of DM1 tissue. The results from this study strongly suggest that CUGBP1 has a major role in DM1 skeletal muscle pathogenesis.


Subject(s)
Disease Models, Animal , Gene Expression , Muscle, Skeletal/metabolism , Myotonic Dystrophy/genetics , RNA-Binding Proteins/genetics , Alternative Splicing , Animals , CELF1 Protein , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Muscle, Skeletal/pathology , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA-Binding Proteins/metabolism , Up-Regulation
14.
J Neurosci ; 28(12): 3123-30, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18354015

ABSTRACT

Recent evidence challenges the prevalent view that neural factors induce the formation of a de novo postsynaptic apparatus during development of the vertebrate neuromuscular junction. The latest experiments suggest an alternative model in which the muscle fiber induces a nascent postsynaptic apparatus and sets the location of the future synapse. On axonal contact, these sites, laid out in a prepattern in the central area of developing muscle fibers, mature into synapses by the combined action of neural factors such as agrin and ACh. We sought to test in mammals these two models of neuromuscular synaptogenesis. Previously, we showed that continuous prenatal muscle expression of constitutively active ErbB2 (CAErbB2) led to synaptic loss, exuberant axonal sprouting, and lethality at birth. Here, we transiently induced CAErbB2 during midgestation and examined synapse restoration after inducer withdrawal. Centrally enriched ACh receptor (AChR) transcription and clustering were abolished after transient CAErbB2 induction. After inducer withdrawal, synapses were restored but were distributed widely over the entire diaphragm muscle. Under the nerve-dependent model, this distribution is explained by the wide pattern of axonal sprouting triggered by CAErbB2. Yet, in the absence of the nerve, introduced in our animals by mating to Hb9(+/-) mice, a very similar, wide distribution of aneural AChR clusters resulted. Thus, transient expression of CAErbB2 in skeletal muscles leads to reprogramming of the endogenous muscle AChR prepattern. This, and not the nerve, seems primarily responsible for the widely distributed pattern of synapses in our experimental animals.


Subject(s)
Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Neuromuscular Junction/physiology , Animals , Animals, Newborn , Bungarotoxins/pharmacokinetics , Diaphragm/cytology , Doxycycline/pharmacology , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins , Luciferases/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/genetics , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Transcription Factors/deficiency
15.
J Neurosci ; 27(22): 5948-57, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17537965

ABSTRACT

The intermediate filament nestin is localized postsynaptically at rodent neuromuscular junctions. The protein forms a filamentous network beneath and between the synaptic gutters, surrounds myofiber nuclei, and is associated with Z-discs adjacent to the junction. In situ hybridization shows that nestin mRNA is synthesized selectively by synaptic myonuclei. Although weak immunoreactivity is present in myelinating Schwann cells that wrap the preterminal axon, nestin is not detected in the terminal Schwann cells (tSCs) that cover the nerve terminal branches. However, after denervation of muscle, nestin is upregulated in tSCs and in SCs within the nerve distal to the lesion site. In contrast, immunoreactivity is strongly downregulated in the muscle fiber. Transgenic mice in which the nestin neural enhancer drives expression of a green fluorescent protein (GFP) reporter show that the regulation in SCs is transcriptional. However, the postsynaptic expression occurs through enhancer elements distinct from those responsible for regulation in SCs. Application of botulinum toxin shows that the upregulation in tSCs and the loss of immunoreactivity in muscle fibers occurs with blockade of transmitter release. Extrinsic stimulation of denervated muscle maintains the postsynaptic expression of nestin but does not affect the upregulation in SCs. Thus, a nestin-containing cytoskeleton is promoted in the postsynaptic muscle fiber by nerve-evoked muscle activity but suppressed in tSCs by transmitter release. Nestin antibodies and GFP driven by nestin promoter elements serve as excellent markers for the reactive state of SCs. Vital imaging of GFP shows that SCs grow a dynamic set of processes after denervation.


Subject(s)
Intermediate Filament Proteins/physiology , Muscle, Skeletal/innervation , Muscle, Skeletal/physiology , Nerve Tissue Proteins/physiology , Neuromuscular Junction/physiology , Animals , Enhancer Elements, Genetic/physiology , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Nestin , Neuromuscular Junction/cytology , Rats , Schwann Cells/cytology , Schwann Cells/physiology , Sciatic Neuropathy/pathology
16.
J Neurosci Res ; 85(9): 1827-33, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17335087

ABSTRACT

At the developing vertebrate neuromuscular junction, neuregulins are growth/differentiation factors essential for terminal Schwann cell survival. Neuregulins have also been thought as the critical signals responsible for the increased transcription of acetylcholine receptor subunit genes at the neuromuscular synapse. This latter role is now highly controversial. This article reviews the evidence that has shaped the views of the neuregulins and how these views have been challenged. The most recent experiments indicate that neuregulin signaling to postsynaptic muscle fibers may modulate, rather than determine, acetylcholine receptor expression at the neuromuscular junction. Based on findings from my lab and those of others, I propose that this modulation might involve novel posttranscriptional molecular mechanisms. Finally, I also suggest that neuregulin signaling may have an important role to play in mediating the response of adult terminal Schwann cells to denervation.


Subject(s)
Neuregulins/physiology , Neuromuscular Junction/physiology , Synapses/physiology , Animals , Humans , Motor Endplate/physiology , Neuregulin-1/physiology , Signal Transduction/physiology
17.
Glia ; 54(6): 630-7, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16944454

ABSTRACT

Here we show that neuregulin-2 (Nrg-2) alpha- and beta-isoforms can activate acetylcholine receptor (AChR) transcription as surface-attached ligands. More importantly, we demonstrate that Schwann cells that express Nrg-2alpha on their cell surface, the same Nrg-2 isoform expressed by terminal Schwann cells at the neuromuscular junction, can induce AChR expression if brought into cell-to-cell contact with myotubes specifically expressing ErbB4. These Schwann cells, the D6P2T cell line, induce AChR expression apparently as well as 293T cells transfected with Nrg-2beta, the isoform with the highest AChR-inducing activity when presented in a soluble form. These results provide a potential role for the previously reported, paradoxical perisynaptic accumulation of Nrg-2alpha, the isoform with the least AChR-inducing activity when presented in a soluble form. They also raise the possibility that Schwann cell-derived Nrg-2 could activate ErbB receptors on the synaptic sarcolemma and that this could account, at least in part, for the Nrg-mediated regulation of AChR expression.


Subject(s)
Muscle, Skeletal/innervation , Nerve Growth Factors/metabolism , Neuromuscular Junction/metabolism , Receptors, Nicotinic/metabolism , Schwann Cells/metabolism , Synaptic Transmission/physiology , Animals , Animals, Newborn , Cell Communication/physiology , Cell Differentiation/physiology , Cell Line , Cells, Cultured , ErbB Receptors/metabolism , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/drug effects , Protein Isoforms/metabolism , Rats , Receptor, ErbB-4 , Receptors, Nicotinic/drug effects , Schwann Cells/drug effects , Synaptic Membranes/metabolism
18.
J Neurosci ; 26(25): 6873-84, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16793894

ABSTRACT

Neuregulins play crucial roles in early development of Schwann cells (SCs), but their roles in the activities of SCs during denervation and reinnervation of muscle are less clear. In the present study, the Tet-On system has been used in transgenic mice to enable inducible expression of a mutant, constitutively active neuregulin receptor (ErbB2) in SCs. This induction simulates neuregulin signaling to these cells. Reporter transgenes were used to show a tightly regulated, SC-selective expression in muscle. Induction leads to a number of changes in SCs at neuromuscular junctions that mimic the response to muscle denervation/reinnervation. These include process extension, soma migration, and proliferation. SCs also come to express nestin, a protein characteristic of their reaction to muscle denervation. This activation of SCs results in the sprouting of nerve terminals, and these sprouts follow the extensions of the SCs. However, these sprouts and their associated SCs disappear after the removal of the inducer. Last, induction of the active receptor is sufficient to rescue SCs in neonatal muscle from denervation-induced apoptosis. These findings show that the responses of SCs in muscle to denervation can be explained by induction of an autocrine/paracrine neuregulin signaling cascade suggested by previous molecular studies.


Subject(s)
Muscle Denervation/methods , Neuregulins/metabolism , Schwann Cells/metabolism , Signal Transduction/physiology , Animals , Bromodeoxyuridine , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cloning, Molecular/methods , Doxycycline/pharmacology , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , In Vitro Techniques , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Neuromuscular Junction/radiation effects , RNA, Messenger/metabolism , Receptor, ErbB-2/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , Schwann Cells/drug effects , Signal Transduction/radiation effects , Time Factors
19.
Mol Cell Neurosci ; 31(2): 334-45, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16278083

ABSTRACT

We overexpressed a constitutively active form of the neuregulin receptor ErbB2 (CAErbB2) in skeletal muscle fibers in vivo and in vitro by tetracycline-inducible expression. Surprisingly, CAErbB2 expression during embryonic development was lethal and impaired synaptogenesis yielding a phenotype with loss of synaptic contacts, extensive axonal sprouting, and diffuse distribution of acetylcholine receptor (AChR) transcripts, reminiscent of agrin-deficient mice. CAErbB2 expression in cultured myotubes inhibited the formation and maintenance of agrin-induced AChR clusters, suggesting a muscle- and not a nerve-origin for the defect in CAErbB2-expressing mice. Levels of tyrosine phosphorylated MuSK, the signaling component of the agrin receptor, were similar, while tyrosine phosphorylation of AChRbeta subunits was dramatically reduced in CAErbB2-expressing embryos relative to controls. Thus, a gain-of-function manipulation of ErbB2 signaling pathways renders an agrin-deficient-like phenotype that uncouples MuSK and AChR tyrosine phosphorylation.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Neuromuscular Junction/embryology , Receptor, ErbB-2/metabolism , Synapses/physiology , Agrin/genetics , Agrin/metabolism , Animals , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/genetics , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Signal Transduction/physiology , Tyrosine/metabolism
20.
Neuroreport ; 16(3): 271-5, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15706234

ABSTRACT

Genetic and behavioral studies in humans and mouse mutants have implicated the gene encoding neuregulin-1 (Nrg-1) as a candidate susceptibility gene for schizophrenia. We examined the behavior of mice heterozygous for a mutation in neuregulin-1's immunoglobulin (Ig)-like domain (Ig-nrg-1 mice). We found that these animals displayed behaviors related to a schizophrenia-like phenotype, such as clozapine suppression of open-field and running wheel activity and impaired latent inhibition. Contrary to findings with other nrg-1 mutants, Ig-nrg-1 mice did not exhibit significantly elevated locomotion relative to littermate controls. These results suggest that Ig-Nrg-1's contribute to some - but not all - aspects of the schizophrenia-like phenotype of nrg-1 mutants, and further support nrg-1 as a candidate gene for schizophrenia.


Subject(s)
Antipsychotic Agents/therapeutic use , Clozapine/therapeutic use , Neural Inhibition/drug effects , Neuregulin-1/chemistry , Neuregulin-1/genetics , Schizophrenia/drug therapy , Animals , Behavior, Animal/physiology , Disease Models, Animal , Exploratory Behavior/drug effects , Immunoglobulins , Mice , Mice, Mutant Strains , Motor Activity/drug effects , Phenotype , Protein Structure, Tertiary/physiology , Running
SELECTION OF CITATIONS
SEARCH DETAIL
...