Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Neurophysiol ; 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36695533

ABSTRACT

Presynaptic inputs determine the pattern of activation of postsynaptic neurons in a neural circuit. Molecular and genetic pathways that regulate the selective formation of subsets of presynaptic inputs are largely unknown, despite significant understanding of the general process of synaptogenesis. In this study, we have begun to identify such factors using the spinal monosynaptic stretch reflex circuit as a model system. In this neuronal circuit, Ia proprioceptive afferents establish monosynaptic connections with spinal motor neurons that project to the same muscle (termed homonymous connections) or muscles with related or synergistic function. However, monosynaptic connections are not formed with motor neurons innervating muscles with antagonistic functions. The ETS transcription factor ER81 (also known as ETV1) is expressed by all proprioceptive afferents, but only a small set of motor neuron pools in the lumbar spinal cord of the mouse. Here we use conditional mouse genetic techniques to eliminate Er81 expression selectively from motor neurons. We find that ablation of Er81 in motor neurons reduces synaptic inputs from proprioceptive afferents conveying information from homonymous and synergistic muscles, with no change observed in the connectivity pattern from antagonistic proprioceptive afferents. In summary, these findings suggest a role for ER81 in defined motor neuron pools to control the assembly of specific presynaptic inputs and thereby influence the profile of activation of these motor neurons.

2.
J Neurosci ; 42(9): 1820-1844, 2022 03 02.
Article in English | MEDLINE | ID: mdl-34992132

ABSTRACT

Neonatal hydrocephalus presents with various degrees of neuroinflammation and long-term neurologic deficits in surgically treated patients, provoking a need for additional medical treatment. We previously reported elevated neuroinflammation and severe periventricular white matter damage in the progressive hydrocephalus (prh) mutant which contains a point mutation in the Ccdc39 gene, causing loss of cilia-mediated unidirectional CSF flow. In this study, we identified cortical neuropil maturation defects such as impaired excitatory synapse maturation and loss of homeostatic microglia, and swimming locomotor defects in early postnatal prh mutant mice. Strikingly, systemic application of the anti-inflammatory small molecule bindarit significantly supports healthy postnatal cerebral cortical development in the prh mutant. While bindarit only mildly reduced the ventricular volume, it significantly improved the edematous appearance and myelination of the corpus callosum. Moreover, the treatment attenuated thinning in cortical Layers II-IV, excitatory synapse formation, and interneuron morphogenesis, by supporting the ramified-shaped homeostatic microglia from excessive cell death. Also, the therapeutic effect led to the alleviation of a spastic locomotor phenotype of the mutant. We found that microglia, but not peripheral monocytes, contribute to amoeboid-shaped activated myeloid cells in prh mutants' corpus callosum and the proinflammatory cytokines expression. Bindarit blocks nuclear factor (NF)-kB activation and its downstream proinflammatory cytokines, including monocyte chemoattractant protein-1, in the prh mutant. Collectively, we revealed that amelioration of neuroinflammation is crucial for white matter and neuronal maturation in neonatal hydrocephalus. Future studies of bindarit treatment combined with CSF diversion surgery may provide long-term benefits supporting neuronal development in neonatal hydrocephalus.SIGNIFICANCE STATEMENT In neonatal hydrocephalus, little is known about the signaling cascades of neuroinflammation or the impact of such inflammatory insults on neural cell development within the perinatal cerebral cortex. Here, we report that proinflammatory activation of myeloid cells, the majority of which are derived from microglia, impairs periventricular myelination and cortical neuronal maturation using the mouse prh genetic model of neonatal hydrocephalus. Administration of bindarit, an anti-inflammatory small molecule that blocks nuclear factor (NF)-kB activation, restored the cortical thinning and synaptic maturation defects in the prh mutant brain through suppression of microglial activation. These data indicate the potential therapeutic use of anti-inflammatory reagents targeting neuroinflammation in the treatment of neonatal hydrocephalus.


Subject(s)
Hydrocephalus , Microglia , Animals , Animals, Newborn , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Female , Humans , Hydrocephalus/drug therapy , Indazoles , Mice , Pregnancy , Propionates
3.
J Neurophysiol ; 126(6): 1948-1958, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34758279

ABSTRACT

Reflex abnormalities mediated by proprioceptive sensory neurons after peripheral nerve injury (PNI) can limit functional improvement, leaving patients with disability that affects their quality of life. We examined postinjury calcium transients in a subpopulation of dorsal root ganglion (DRG) neurons consisting primarily of proprioceptors to determine whether alterations in calcium homeostasis are present in proprioceptors, as has been documented in other DRG neurons after PNI. Using transgenic mice, we restricted expression of the calcium indicator GCaMP6s to DRG neurons containing parvalbumin (PV). Mice of both sexes were randomly assigned to sham, sciatic nerve crush, or sciatic nerve transection and resuture conditions. Calcium transients were recorded from ex vivo preparations of animals at one of three postsurgery time points: 1-3 days, 7-11 days, and after 60 days of recovery. Results demonstrated that the post-PNI calcium transients of PV DRG neurons are significantly different than sham. Abnormalities were not present during the acute response to injury (1-3 days), but transients were significantly different than sham at the recovery stage where axon regeneration is thought to be underway (7-11 days). During late-stage recovery (60 days postinjury), disturbances in the decay time course of calcium transients in transection animals persisted, whereas parameters of transients from crush animals returned to normal. These findings identify a deficit in calcium homeostasis in proprioceptive neurons, which may contribute to the failure to fully recover proprioceptive reflexes after PNI. Significant differences in the calcium transients of crush versus transection animals after reinnervation illustrate calcium homeostasis alterations are distinctive to injury type.NEW & NOTEWORTHY This study examines calcium homeostasis after peripheral nerve injury in dorsal root ganglion (DRG) neurons expressing parvalbumin, a group of large-diameter afferents primarily consisting of proprioceptors, using two-photon calcium imaging in the intact DRG. Our findings identify aberrant calcium homeostasis as an additional source of sensory neuron dysfunction following peripheral nerve injury, uncover differences between two injury models, and track how these changes develop and resolve over the course of recovery.


Subject(s)
Calcium/metabolism , Ganglia, Spinal/metabolism , Neurons/metabolism , Parvalbumins/metabolism , Peripheral Nerve Injuries/metabolism , Proprioception/physiology , Sciatic Neuropathy/metabolism , Sensory Receptor Cells/physiology , Animals , Disease Models, Animal , Female , Homeostasis/physiology , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Nerve Crush
4.
Ann Neurol ; 88(2): 297-308, 2020 08.
Article in English | MEDLINE | ID: mdl-32418267

ABSTRACT

OBJECTIVE: Myotonia is caused by involuntary firing of skeletal muscle action potentials and causes debilitating stiffness. Current treatments are insufficiently efficacious and associated with side effects. Myotonia can be triggered by voluntary movement (electrically induced myotonia) or percussion (mechanically induced myotonia). Whether distinct molecular mechanisms underlie these triggers is unknown. Our goal was to identify ion channels involved in mechanically induced myotonia and to evaluate block of the channels involved as a novel approach to therapy. METHODS: We developed a novel system to enable study of mechanically induced myotonia using both genetic and pharmacologic mouse models of myotonia congenita. We extended ex vivo studies of excitability to in vivo studies of muscle stiffness. RESULTS: As previous work suggests activation of transient receptor potential vanilloid 4 (TRPV4) channels by mechanical stimuli in muscle, we examined the role of this cation channel. Mechanically induced myotonia was markedly suppressed in TRPV4-null muscles and in muscles treated with TRPV4 small molecule antagonists. The suppression of mechanically induced myotonia occurred without altering intrinsic muscle excitability, such that myotonia triggered by firing of action potentials (electrically induced myotonia) was unaffected. When injected intraperitoneally, TRPV4 antagonists lessened the severity of myotonia in vivo by approximately 80%. INTERPRETATION: These data demonstrate that there are distinct molecular mechanisms triggering electrically induced and mechanically induced myotonia. Our data indicates that activation of TRPV4 during muscle contraction plays an important role in triggering myotonia in vivo. Elimination of mechanically induced myotonia by TRPV4 inhibition offers a new approach to treating myotonia. ANN NEUROL 2020;88:297-308.


Subject(s)
Isometric Contraction/physiology , Morpholines/pharmacology , Myotonia Congenita/genetics , Myotonia Congenita/metabolism , Pyrroles/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , Animals , Anthracenes/pharmacology , Isometric Contraction/drug effects , Mice , Mice, Knockout , Morpholines/therapeutic use , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Myotonia Congenita/prevention & control , Pyrroles/therapeutic use
5.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31311802

ABSTRACT

We investigated the calcium dynamics of dorsal root ganglion (DRG) neurons using transgenic mice to target expression of the genetically encoded calcium indicator (GECI), GCaMP6s, to a subset of neurons containing parvalbumin (PV), a calcium-binding protein present in proprioceptors and low-threshold mechanoreceptors. This study provides the first analysis of GECI calcium transient parameters from large-diameter DRG neurons. Our approach generated calcium transients of consistent shape and time-course, with quantifiable characteristics. Four parameters of calcium transients were determined to vary independently from each other and thus are likely influenced by different calcium-regulating mechanisms: peak amplitude, rise time (RT), decay time, and recovery time. Pooled analysis of 188 neurons demonstrated unimodal distributions, providing evidence that PV+ DRG neurons regulate calcium similarly as a population despite their differences in size, electrical properties, and functional sensitivities. Calcium transients increased in size with elevated extracellular calcium, longer trains of action potentials, and higher stimulation frequencies. RT and decay time increased with the addition of the selective sarco/endoplasmic reticulum calcium ATPases (SERCA) blocker, thapsigargin (TG), while peak amplitude and recovery time remained the same. When elevating bath pH to 8.8 to block plasma-membrane calcium ATPases (PMCA), all measured parameters significantly increased. These results illustrate that GECI calcium transients provide sufficient resolution to detect changes in electrical activity and intracellular calcium concentration, as well as discern information about the activity of specific subclasses of calcium regulatory mechanisms.


Subject(s)
Calcium Signaling/physiology , Ganglia, Spinal/physiology , Neurons/physiology , Parvalbumins/physiology , Animals , Calcium/analysis , Female , Male , Mice, Transgenic , Optical Imaging/methods
6.
Neurosci Lett ; 674: 36-41, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29526514

ABSTRACT

Reciprocal inhibition of motor neurons via Ia inhibitory interneurons recruited by stimulation of proprioceptive afferents supplying antagonist muscles has been well described. Changes in the efficacy of inhibition, and sometimes even a switch from inhibition to facilitation, have been reported in the literature after disruption of descending pathways. We sought to test whether such facilitation could be expressed in normal animals by evaluating the presence of facilitation in acute preparations from uninjured animals. Using an isolated spinal cord preparation from neonatal mice, changes in the monosynaptic stretch reflex response in knee flexor motor neurons (posterior biceps semitendinosus; PBST) were monitored following conditioning stimulation of proprioceptive sensory afferents in other muscle nerves. As expected for reciprocal inhibition, conditioning by stimulation of quadriceps (knee extensors and PBST antagonists) sensory afferents resulted in inhibition of the stretch reflex response. Facilitation, however, of the stretch reflex response by quadriceps conditioning stimulation was observed when the glycinergic reciprocal inhibitory pathway was blocked by application of strychnine. Facilitation was elicited by low-threshold proprioceptive afferents and occurred at latencies consistent with a disynaptic circuit. The magnitude of facilitation was larger at birth than at one week postnatal. Our results also suggest reciprocal facilitation is restricted to antagonist muscle pairs, as facilitation of PBST responses was not observed when conditioned with the obturator nerve supplying the adductor muscles. Overall, these data suggest the efficacy of facilitation is modulated during the first postnatal week, while the specificity of facilitation is already established by birth.


Subject(s)
Motor Neurons/physiology , Neurons, Afferent/physiology , Proprioception/physiology , Spinal Cord/physiology , Afferent Pathways/growth & development , Afferent Pathways/physiology , Animals , Animals, Newborn , Mice, Inbred C57BL , Muscle, Skeletal/growth & development , Muscle, Skeletal/innervation , Muscle, Skeletal/physiology , Neural Inhibition , Reflex, Stretch , Spinal Cord/growth & development
7.
Dev Dyn ; 247(1): 185-193, 2018 01.
Article in English | MEDLINE | ID: mdl-29090497

ABSTRACT

BACKGROUND: Expression of the calcium binding protein, calbindin (CB), is well established as a hallmark of Renshaw cells, a class of interneurons found in spatially restricted areas in the ventral spinal cord that directly modulate motor neuron activity. CB expression, however, is not restricted only to Renshaw cells in the ventral horn, and within this population other interneuron subtypes may be identifiable on the basis of cell position and the potential for coexpression of other calcium binding proteins. RESULTS: Here we have quantified the changing CB expression pattern in the ventral spinal cord across postnatal development in the mouse. Fewer neurons express CB as postnatal development progresses, and those neurons frequently coexpress other calcium binding proteins (calretinin and parvalbumin) in subpopulations with distinct spatial distributions. We also found a significant portion of CB-expressing interneurons receive putative synaptic contacts from primary sensory afferents. CONCLUSIONS: These findings suggest CB labels a heterogeneous group of interneurons in the ventral horn, some of which may process sensory information. Based on cellular position, CB expression may be a shared feature of subsets of interneurons arising from multiple ventral progenitor domains. Developmental Dynamics 247:185-193, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Anterior Horn Cells/metabolism , Calbindin 1/metabolism , Interneurons/metabolism , Spinal Cord/metabolism , Animals , Calbindin 1/genetics , Immunohistochemistry , Mice , Parvalbumins/metabolism
8.
PLoS One ; 12(1): e0170751, 2017.
Article in English | MEDLINE | ID: mdl-28122055

ABSTRACT

Muscle proprioceptive afferents provide feedback critical for successful execution of motor tasks via specialized mechanoreceptors housed within skeletal muscles: muscle spindles, supplied by group Ia and group II afferents, and Golgi tendon organs, supplied by group Ib afferents. The morphology of these proprioceptors and their associated afferents has been studied extensively in the cat soleus, and to a lesser degree, in the rat; however, quantitative analyses of proprioceptive innervation in the mouse soleus are comparatively limited. The present study employed genetically-encoded fluorescent reporting systems to label and analyze muscle spindles, Golgi tendon organs, and the proprioceptive sensory neuron subpopulations supplying them within the intact mouse soleus muscle using high magnification confocal microscopy. Total proprioceptive receptors numbered 11.3 ± 0.4 and 5.2 ± 0.2 for muscle spindles and Golgi tendon organs, respectively, and these receptor counts varied independently (n = 27 muscles). Analogous to findings in the rat, muscle spindles analyzed were most frequently supplied by two proprioceptive afferents, and in the majority of instances, both were classified as primary endings using established morphological criteria. Secondary endings were most frequently observed when spindle associated afferents totaled three or more. The mean diameter of primary and secondary afferent axons differed significantly, but the distributions overlap more than previously observed in cat and rat studies.


Subject(s)
Muscle Contraction/physiology , Muscle Spindles/physiology , Muscle, Skeletal/innervation , Proprioception/physiology , Sensory Receptor Cells/physiology , Animals , Mechanoreceptors/physiology , Mice , Mice, Transgenic
9.
J Neurophysiol ; 117(3): 1057-1069, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28003412

ABSTRACT

Neuronal cell identity is established during development and must be maintained throughout an animal's life (Fishell G, Heintz N. Neuron 80: 602-612, 2013). Transcription factors critical for establishing neuronal identity can be required for maintaining it (Deneris ES, Hobert O. Nat Neurosci 17: 899-907, 2014). Posttranscriptional regulation also plays an important role in neuronal differentiation (Bian S, Sun T. Mol Neurobiol 44: 359-373, 2011), but its role in maintaining cell identity is less established. To better understand how posttranscriptional regulation might contribute to cell identity, we examined the proprioceptive neurons in the dorsal root ganglion (DRG), a highly specialized sensory neuron class, with well-established properties that distinguish them from other neurons in the ganglion. By conditionally ablating Dicer in mice, using parvalbumin (Pvalb)-driven Cre recombinase, we impaired posttranscriptional regulation in the proprioceptive sensory neuron population. Knockout (KO) animals display a progressive form of ataxia at the beginning of the fourth postnatal week that is accompanied by a cell death within the DRG. Before cell loss, expression profiling shows a reduction of proprioceptor specific genes and an increased expression of nonproprioceptive genes normally enriched in other ganglion neurons. Furthermore, although central connections of these neurons are intact, the peripheral connections to the muscle are functionally impaired. Posttranscriptional regulation is therefore necessary to retain the transcriptional identity and support functional specialization of the proprioceptive sensory neurons.NEW & NOTEWORTHY We have demonstrated that selectively impairing Dicer in parvalbumin-positive neurons, which include the proprioceptors, triggers behavioral changes, a lack of muscle connectivity, and a loss of transcriptional identity as observed through RNA sequencing. These results suggest that Dicer and, most likely by extension, microRNAs are crucially important for maintaining proprioception. Additionally, this study hints at the larger question of how neurons maintain their functional and molecular specificity.


Subject(s)
Ataxia/physiopathology , DEAD-box RNA Helicases/physiology , Ganglia, Spinal/physiology , Proprioception , Protein Processing, Post-Translational , Ribonuclease III/physiology , Sensory Receptor Cells/physiology , Animals , Ataxia/genetics , Cell Death , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Ganglia, Spinal/metabolism , Mice , Mice, Knockout , Muscle Spindles/physiology , Muscle, Skeletal/cytology , Parvalbumins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Sensory Receptor Cells/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
10.
J Neurosci ; 36(21): 5724-35, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27225763

ABSTRACT

UNLABELLED: Spinal reflex circuit development requires the precise regulation of axon trajectories, synaptic specificity, and synapse formation. Of these three crucial steps, the molecular mechanisms underlying synapse formation between group Ia proprioceptive sensory neurons and motor neurons is the least understood. Here, we show that the Rho GTPase Cdc42 controls synapse formation in monosynaptic sensory-motor connections in presynaptic, but not postsynaptic, neurons. In mice lacking Cdc42 in presynaptic sensory neurons, proprioceptive sensory axons appropriately reach the ventral spinal cord, but significantly fewer synapses are formed with motor neurons compared with wild-type mice. Concordantly, electrophysiological analyses show diminished EPSP amplitudes in monosynaptic sensory-motor circuits in these mutants. Temporally targeted deletion of Cdc42 in sensory neurons after sensory-motor circuit establishment reveals that Cdc42 does not affect synaptic transmission. Furthermore, addition of the synaptic organizers, neuroligins, induces presynaptic differentiation of wild-type, but not Cdc42-deficient, proprioceptive sensory neurons in vitro Together, our findings demonstrate that Cdc42 in presynaptic neurons is required for synapse formation in monosynaptic sensory-motor circuits. SIGNIFICANCE STATEMENT: Group Ia proprioceptive sensory neurons form direct synapses with motor neurons, but the molecular mechanisms underlying synapse formation in these monosynaptic sensory-motor connections are unknown. We show that deleting Cdc42 in sensory neurons does not affect proprioceptive sensory axon targeting because axons reach the ventral spinal cord appropriately, but these neurons form significantly fewer presynaptic terminals on motor neurons. Electrophysiological analysis further shows that EPSPs are decreased in these mice. Finally, we demonstrate that Cdc42 is involved in neuroligin-dependent presynaptic differentiation of proprioceptive sensory neurons in vitro These data suggest that Cdc42 in presynaptic sensory neurons is essential for proper synapse formation in the development of monosynaptic sensory-motor circuits.


Subject(s)
Axon Guidance/physiology , Motor Neurons/physiology , Neurogenesis/physiology , Presynaptic Terminals/physiology , Sensory Receptor Cells/physiology , cdc42 GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Male , Mice , Mice, Knockout , Motor Neurons/cytology , Presynaptic Terminals/ultrastructure , Sensory Receptor Cells/cytology , Spinal Cord/cytology , Spinal Cord/physiology
11.
Cell Rep ; 5(3): 748-58, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24210822

ABSTRACT

In mammalian spinal cord, group Ia proprioceptive afferents form selective monosynaptic connections with a select group of motor pool targets. The extent to which sensory recognition of motor neurons contributes to the selectivity of sensory-motor connections remains unclear. We show here that proprioceptive sensory afferents that express PlexinD1 avoid forming monosynaptic connections with neurons in Sema3E(+) motor pools yet are able to form direct connections with neurons in Sema3E(off) motor pools. Anatomical and electrophysiological analysis of mice in which Sema3E-PlexinD1 signaling has been deregulated or inactivated genetically reveals that repellent signaling underlies aspects of the specificity of monosynaptic sensory-motor connectivity in these reflex arcs. A semaphorin-based system of motor neuron recognition and repulsion therefore contributes to the formation of specific sensory-motor connections in mammalian spinal cord.


Subject(s)
Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Neural Pathways/metabolism , Sensory Receptor Cells/metabolism , Animals , Cytoskeletal Proteins , Glycoproteins/biosynthesis , Glycoproteins/genetics , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Motor Neurons/cytology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neural Pathways/cytology , Semaphorins , Sensory Receptor Cells/cytology , Signal Transduction , Substrate Specificity
12.
Physiol Behav ; 120: 97-105, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911806

ABSTRACT

Proprioceptive feedback derived from specialized receptors in skeletal muscle is critical in forming an accurate map of limb position in space, and is used by the central nervous system to plan future movements and to determine accuracy of executed movements. Knockout mouse strains for genes expressed by proprioceptive sensory neurons have been generated that result in generalized motor deficits, but these deficits have not been quantitatively characterized. Here we characterize a conditional knockout mouse model wherein proprioceptive sensory neuron synaptic transmission has been blocked by selective ablation of munc18-1, a synaptic vesicle associated protein required for fusion of synaptic vesicles with the plasma membrane. Proprioceptive munc18-1 conditional mutants are impaired in surface righting--a dynamic postural adjustment task--and display several specific deficits in pivoting, an early locomotor behavior. Before the emergence of forward locomotion during postnatal development, animals explore their surroundings through pivoting, or rotating the upper torso around the relatively immobile base of the hind limbs. 3-D kinematic analysis was used to quantitatively describe this pivoting behavior at postnatal days 5 and 8 in control and munc18-1 conditional mutants. Mutant animals also pivot, but demonstrate alterations in movement strategy and in postural placement of the forelimbs during pivoting when compared to controls. In addition, brief forelimb stepping movements associated with pivoting are altered in mutant animals. Step duration and step height are increased in mutant animals. These results underscore the importance of proprioceptive feedback even at early stages in postnatal development.


Subject(s)
Animals, Newborn/physiology , Feedback, Physiological/physiology , Locomotion/physiology , Proprioception/genetics , Proprioception/physiology , Animals , Behavior, Animal/physiology , Biomechanical Phenomena , Forelimb/physiology , Mechanoreceptors/physiology , Mice , Mice, Neurologic Mutants , Munc18 Proteins/genetics , Munc18 Proteins/physiology , Muscle, Skeletal/physiology , Postural Balance/physiology , Posture/physiology , Software , Video Recording
13.
J Neurophysiol ; 109(8): 2118-28, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23343895

ABSTRACT

Sensory feedback is critical for normal locomotion and adaptation to external perturbations during movement. Feedback provided by group Ia afferents influences motor output both directly through monosynaptic connections and indirectly through spinal interneuronal circuits. For example, the circuit responsible for reciprocal inhibition, which acts to prevent co-contraction of antagonist flexor and extensor muscles, is driven by Ia afferent feedback. Additionally, circuits mediating presynaptic inhibition can limit Ia afferent synaptic transmission onto central neuronal targets in a task-specific manner. These circuits can also be activated by stimulation of proprioceptive afferents. Rodent locomotion rapidly matures during postnatal development; therefore, we assayed the functional status of reciprocal and presynaptic inhibitory circuits of mice at birth and compared responses with observations made after 1 wk of postnatal development. Using extracellular physiological techniques from isolated and hemisected spinal cord preparations, we demonstrate that Ia afferent-evoked reciprocal inhibition is as effective at blocking antagonist motor neuron activation at birth as at 1 wk postnatally. In contrast, at birth conditioning stimulation of muscle nerve afferents failed to evoke presynaptic inhibition sufficient to block functional transmission at synapses between Ia afferents and motor neurons, even though dorsal root potentials could be evoked by stimulating the neighboring dorsal root. Presynaptic inhibition at this synapse was readily observed, however, at the end of the first postnatal week. These results indicate Ia afferent feedback from the periphery to central spinal circuits is only weakly gated at birth, which may provide enhanced sensitivity to peripheral feedback during early postnatal experiences.


Subject(s)
GABAergic Neurons/physiology , Neural Inhibition , Proprioception/physiology , Sensory Receptor Cells/physiology , Spinal Cord/physiology , Synaptic Transmission , Action Potentials , Animals , Feedback, Sensory , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Nerve Net/growth & development , Nerve Net/physiology , Spinal Cord/growth & development , Spinal Nerve Roots/growth & development , Spinal Nerve Roots/physiology
14.
J Neurosci Methods ; 200(2): 129-43, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21736898

ABSTRACT

The spatial distribution of synaptic inputs on the dendritic tree of a neuron can have significant influence on neuronal function. Consequently, accurate anatomical reconstructions of neuron morphology and synaptic localization are critical when modeling and predicting physiological responses of individual neurons. Historically, generation of three-dimensional (3D) neuronal reconstructions together with comprehensive mapping of synaptic inputs has been an extensive task requiring manual identification of putative synaptic contacts directly from tissue samples or digital images. Recent developments in neuronal tracing software applications have improved the speed and accuracy of 3D reconstructions, but localization of synaptic sites through the use of pre- and/or post-synaptic markers has remained largely a manual process. To address this problem, we have developed an algorithm, based on 3D distance measurements between putative pre-synaptic terminals and the post-synaptic dendrite, to automate synaptic contact detection on dendrites of individually labeled neurons from 3D immunofluorescence image sets. In this study, the algorithm is implemented with custom routines in Matlab, and its effectiveness is evaluated through analysis of primary sensory afferent terminals on motor neurons. Optimization of algorithm parameters enabled automated identification of synaptic contacts that matched those identified by manual inspection with low incidence of error. Substantial time savings and the elimination of variability in contact detection introduced by different users are significant advantages of this method.


Subject(s)
Algorithms , Dendrites/ultrastructure , Imaging, Three-Dimensional/methods , Motor Neurons/cytology , Presynaptic Terminals/metabolism , Animals , Animals, Newborn , Biotin/analogs & derivatives , Biotin/metabolism , Electronic Data Processing , Mice , Mice, Inbred C57BL , Models, Neurological , Spinal Cord/cytology , Vesicular Glutamate Transport Protein 1/metabolism
15.
Proc Natl Acad Sci U S A ; 106(32): 13588-93, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19651609

ABSTRACT

Spinal motor neurons are specified to innervate different muscle targets through combinatorial programs of transcription factor expression. Whether transcriptional programs also establish finer aspects of motor neuron subtype identity, notably the prominent functional distinction between alpha and gamma motor neurons, remains unclear. In this study, we identify DNA binding proteins with complementary expression profiles in alpha and gamma motor neurons, providing evidence for molecular distinctions in these two motor neuron subtypes. The transcription factor Err3 is expressed at high levels in gamma but not alpha motor neurons, whereas the neuronal DNA binding protein NeuN marks alpha but not gamma motor neurons. Signals from muscle spindles are needed to support the differentiation of Err3(on)/NeuN(off) presumptive gamma motor neurons, whereas direct proprioceptive sensory input to a motor neuron pool is apparently dispensable. Together, these findings provide evidence that transcriptional programs define functionally distinct motor neuron subpopulations, even within anatomically defined motor pools.


Subject(s)
Motor Neurons/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Mice , Motor Neurons/enzymology , Motor Neurons/pathology , Muscle Spindles/metabolism , Muscle Spindles/pathology , Neuraminidase/metabolism , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Phenotype , Proprioception
16.
Neuron ; 56(2): 270-83, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17964245

ABSTRACT

Motor circuits in the spinal cord integrate information from various sensory and descending pathways to control appropriate motor behavior. Recent work has revealed that target-derived retrograde signaling mechanisms act to influence sequential assembly of motor circuits through combinatorial action of genetic and experience-driven programs. These parallel activities imprint somatotopic information at the level of the spinal cord in precisely interconnected circuits and equip animals with motor circuits capable of reacting to changing demands throughout life.


Subject(s)
Efferent Pathways/physiology , Motor Neurons/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Gene Expression , Models, Biological , Receptors, Nerve Growth Factor/genetics
17.
Neuron ; 55(5): 726-40, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17785180

ABSTRACT

Accumulation of specific proteins at synaptic structures is essential for synapse assembly and function, but mechanisms regulating local protein enrichment remain poorly understood. At the neuromuscular junction (NMJ), subsynaptic nuclei underlie motor axon terminals within extrafusal muscle fibers and are transcriptionally distinct from neighboring nuclei. In this study, we show that expression of the ETS transcription factor Erm is highly concentrated at subsynaptic nuclei, and its mutation in mice leads to severe downregulation of many genes with normally enriched subsynaptic expression. Erm mutant mice display an expansion of the muscle central domain in which acetylcholine receptor (AChR) clusters accumulate, show gradual fragmentation of AChR clusters, and exhibit symptoms of muscle weakness mimicking congenital myasthenic syndrome (CMS). Together, our findings define Erm as an upstream regulator of a transcriptional program selective to subsynaptic nuclei at the NMJ and underscore the importance of transcriptional control of local synaptic protein accumulation.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Muscle, Skeletal/growth & development , Neuromuscular Junction/growth & development , Receptors, Nicotinic/genetics , Synaptic Transmission/genetics , Transcription Factors/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Mice , Mice, Knockout , Muscle Weakness/genetics , Muscle Weakness/metabolism , Muscle Weakness/physiopathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Mutation/genetics , Myasthenia Gravis/genetics , Myasthenia Gravis/metabolism , Myasthenia Gravis/physiopathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Receptor Aggregation/genetics , Receptors, Nicotinic/metabolism , Synaptic Membranes/genetics , Synaptic Membranes/metabolism , Transcription Factors/metabolism , Transcriptional Activation/genetics
18.
PLoS Biol ; 3(5): e159, 2005 May.
Article in English | MEDLINE | ID: mdl-15836427

ABSTRACT

Two ETS transcription factors of the Pea3 subfamily are induced in subpopulations of dorsal root ganglion (DRG) sensory and spinal motor neurons by target-derived factors. Their expression controls late aspects of neuronal differentiation such as target invasion and branching. Here, we show that the late onset of ETS gene expression is an essential requirement for normal sensory neuron differentiation. We provide genetic evidence in the mouse that precocious ETS expression in DRG sensory neurons perturbs axonal projections, the acquisition of terminal differentiation markers, and their dependence on neurotrophic support. Together, our findings indicate that DRG sensory neurons exhibit a temporal developmental switch that can be revealed by distinct responses to ETS transcription factor signaling at sequential steps of neuronal maturation.


Subject(s)
Ganglia, Spinal/physiology , Neurons/physiology , Proto-Oncogene Proteins c-ets/physiology , Signal Transduction/physiology , Animals , Base Sequence , Cell Differentiation , DNA Primers , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exons , Ganglia, Spinal/growth & development , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Transgenic , Motor Neurons/physiology , Muscle, Skeletal/innervation , Neurons/cytology , Neurons, Afferent/physiology , Recombinant Proteins/metabolism , Spinal Cord/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Physiol Behav ; 77(4-5): 571-6, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12527001

ABSTRACT

The ETS family of transcription factors includes two members, ER81 and PEA3, which are expressed in groups of sensory and motor neurons supplying individual muscles. To investigate a possible role of these genes in determining sensory and/or motor neuron phenotype, we studied mice in which each of these genes was deleted. In contrast to the deletion of ER81, which blocks the formation of projections from muscle sensory neurons to motor neurons in the spinal cord, deletion of PEA3 causes no obvious effects on sensory neurons or on their synaptic connections with motor neurons. PEA3 does play a major role in the formation of some brachial motoneurons however. Motoneurons innervating the cutaneous maximus muscle, which are normally PEA3(+), fail to develop normally so that postnatally the muscle is innervated by few motoneurons and is severely atrophic. Other studies suggest that these motoneurons initially appear during development but fail to contact their normal muscle targets.


Subject(s)
Central Nervous System/growth & development , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Motor Neurons/physiology , Neurons, Afferent/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Cell Count , Central Nervous System/cytology , Central Nervous System/physiology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Mutation/genetics , Neural Pathways/physiology , Spinal Cord/cytology , Spinal Cord/growth & development , Spinal Cord/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...