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










Publication year range
1.
Elife ; 122023 Dec 19.
Article in English | MEDLINE | ID: mdl-38113081

ABSTRACT

Neurons coordinate their activity to produce an astonishing variety of motor behaviors. Our present understanding of motor control has grown rapidly thanks to new methods for recording and analyzing populations of many individual neurons over time. In contrast, current methods for recording the nervous system's actual motor output - the activation of muscle fibers by motor neurons - typically cannot detect the individual electrical events produced by muscle fibers during natural behaviors and scale poorly across species and muscle groups. Here we present a novel class of electrode devices ('Myomatrix arrays') that record muscle activity at unprecedented resolution across muscles and behaviors. High-density, flexible electrode arrays allow for stable recordings from the muscle fibers activated by a single motor neuron, called a 'motor unit,' during natural behaviors in many species, including mice, rats, primates, songbirds, frogs, and insects. This technology therefore allows the nervous system's motor output to be monitored in unprecedented detail during complex behaviors across species and muscle morphologies. We anticipate that this technology will allow rapid advances in understanding the neural control of behavior and identifying pathologies of the motor system.


Subject(s)
Motor Neurons , Primates , Rats , Mice , Animals , Motor Neurons/physiology , Electrodes , Muscle Fibers, Skeletal
2.
bioRxiv ; 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-36865176

ABSTRACT

Neurons coordinate their activity to produce an astonishing variety of motor behaviors. Our present understanding of motor control has grown rapidly thanks to new methods for recording and analyzing populations of many individual neurons over time. In contrast, current methods for recording the nervous system's actual motor output - the activation of muscle fibers by motor neurons - typically cannot detect the individual electrical events produced by muscle fibers during natural behaviors and scale poorly across species and muscle groups. Here we present a novel class of electrode devices ("Myomatrix arrays") that record muscle activity at unprecedented resolution across muscles and behaviors. High-density, flexible electrode arrays allow for stable recordings from the muscle fibers activated by a single motor neuron, called a "motor unit", during natural behaviors in many species, including mice, rats, primates, songbirds, frogs, and insects. This technology therefore allows the nervous system's motor output to be monitored in unprecedented detail during complex behaviors across species and muscle morphologies. We anticipate that this technology will allow rapid advances in understanding the neural control of behavior and in identifying pathologies of the motor system.

3.
bioRxiv ; 2023 Mar 21.
Article in English | MEDLINE | ID: mdl-36993220

ABSTRACT

Innate and goal-directed movements require a high-degree of trunk and appendicular muscle coordination to preserve body stability while ensuring the correct execution of the motor action. The spinal neural circuits underlying motor execution and postural stability are finely modulated by propriospinal, sensory and descending feedback, yet how distinct spinal neuron populations cooperate to control body stability and limb coordination remains unclear. Here, we identified a spinal microcircuit composed of V2 lineage-derived excitatory (V2a) and inhibitory (V2b) neurons that together coordinate ipsilateral body movements during locomotion. Inactivation of the entire V2 neuron lineage does not impair intralimb coordination but destabilizes body balance and ipsilateral limb coupling, causing mice to adopt a compensatory festinating gait and be unable to execute skilled locomotor tasks. Taken together our data suggest that during locomotion the excitatory V2a and inhibitory V2b neurons act antagonistically to control intralimb coordination, and synergistically to coordinate forelimb and hindlimb movements. Thus, we suggest a new circuit architecture, by which neurons with distinct neurotransmitter identities employ a dual-mode of operation, exerting either synergistic or opposing functions to control different facets of the same motor behavior.

4.
Neuron ; 109(1): 73-90.e7, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33181066

ABSTRACT

The spinal dorsal horn is a major site for the induction and maintenance of mechanical allodynia, but the circuitry that underlies this clinically important form of pain remains unclear. The studies presented here provide strong evidence that the neural circuits conveying mechanical allodynia in the dorsal horn differ by the nature of the injury. Calretinin (CR) neurons in lamina II inner convey mechanical allodynia induced by inflammatory injuries, while protein kinase C gamma (PKCγ) neurons at the lamina II/III border convey mechanical allodynia induced by neuropathic injuries. Cholecystokinin (CCK) neurons located deeper within the dorsal horn (laminae III-IV) are important for both types of injuries. Interestingly, the Maf+ subset of CCK neurons is composed of transient vesicular glutamate transporter 3 (tVGLUT3) neurons, which convey primarily dynamic allodynia. Identification of an etiology-based circuitry for mechanical allodynia in the dorsal horn has important implications for the mechanistic and clinical understanding of this condition.


Subject(s)
Hyperalgesia/metabolism , Nerve Net/metabolism , Pain Measurement/methods , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Injuries/metabolism , Amino Acid Transport Systems, Acidic/metabolism , Animals , Female , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/chemistry , Nerve Net/pathology , Spinal Cord Dorsal Horn/chemistry , Spinal Cord Dorsal Horn/pathology , Spinal Cord Injuries/pathology
5.
Neuron ; 109(1): 91-104.e5, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33181065

ABSTRACT

Cutaneous somatosensory modalities play pivotal roles in generating a wide range of sensorimotor behaviors, including protective and corrective reflexes that dynamically adapt ongoing movement and posture. How interneurons (INs) in the dorsal horn encode these modalities and transform them into stimulus-appropriate motor behaviors is not known. Here, we use an intersectional genetic approach to functionally assess the contribution that eight classes of dorsal excitatory INs make to sensorimotor reflex responses. We demonstrate that the dorsal horn is organized into spatially restricted excitatory modules composed of molecularly heterogeneous cell types. Laminae I/II INs drive chemical itch-induced scratching, laminae II/III INs generate paw withdrawal movements, and laminae III/IV INs modulate dynamic corrective reflexes. These data reveal a key principle in spinal somatosensory processing, namely, sensorimotor reflexes are driven by the differential spatial recruitment of excitatory neurons.


Subject(s)
Pain Measurement/methods , Psychomotor Performance/physiology , Reflex/physiology , Spinal Cord/metabolism , Spinal Cord/pathology , Animals , Female , Male , Mice , Mice, Transgenic , Physical Stimulation/adverse effects , Spinal Cord/chemistry
6.
Curr Opin Neurobiol ; 56: 167-174, 2019 06.
Article in English | MEDLINE | ID: mdl-30953870

ABSTRACT

A recent flurry of genetic studies in mice have provided key insights into how the somatosensory system is organized at a cellular level to encode itch, pain, temperature, and touch. These studies are largely predicated on the idea that functional cell types can be identified by their unique developmental provenance and gene expression profile. However, the extent to which gene expression profiles can be correlated with functional cell types and circuit organization remains an open question. In this review, we focus on recent progress in characterizing the sensory afferent and dorsal horn neuron cell types that process cutaneous somatosensory information and ongoing circuit studies that are beginning to bridge the divide between cell type and function.


Subject(s)
Neurons , Animals , Pain , Pruritus , Touch
7.
Curr Biol ; 28(6): R256-R259, 2018 03 19.
Article in English | MEDLINE | ID: mdl-29558639

ABSTRACT

Three new and closely complementary studies have defined the architecture of the circuits underlying the descending control of locomotion, identifying neurons that drive fast motor responses and those that seem to be specialised for exploratory behaviors.


Subject(s)
Brain Stem , Locomotion , Exploratory Behavior , Neurons
8.
Neuron ; 96(6): 1419-1431.e5, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29224725

ABSTRACT

Animals depend on sensory feedback from mechanosensory afferents for the dynamic control of movement. This sensory feedback needs to be selectively modulated in a task- and context-dependent manner. Here, we show that inhibitory interneurons (INs) expressing the RORß orphan nuclear receptor gate sensory feedback to the spinal motor system during walking and are required for the production of a fluid locomotor rhythm. Genetic manipulations that abrogate inhibitory RORß IN function result in an ataxic gait characterized by exaggerated flexion movements and marked alterations to the step cycle. Inactivation of RORß in inhibitory neurons leads to reduced presynaptic inhibition and changes to sensory-evoked reflexes, arguing that the RORß inhibitory INs function to suppress the sensory transmission pathways that activate flexor motor reflexes and interfere with the ongoing locomotor program. VIDEO ABSTRACT.


Subject(s)
Interneurons/physiology , Locomotion/physiology , Nuclear Receptor Subfamily 1, Group F, Member 2/metabolism , Spinal Cord/cytology , Walking/physiology , Afferent Pathways , Animals , Animals, Newborn , Electric Stimulation , Feedback, Sensory , GABA Agents/pharmacology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Glycine Plasma Membrane Transport Proteins/genetics , Glycine Plasma Membrane Transport Proteins/metabolism , Hip Joint/innervation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/physiology , Neural Inhibition/genetics , Neural Inhibition/physiology , Nuclear Receptor Subfamily 1, Group F, Member 2/genetics , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Reflex/genetics , Reflex/physiology , Sensory Thresholds/physiology
9.
Curr Biol ; 24(20): 2355-65, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25264256

ABSTRACT

BACKGROUND: Proteolytic processing of axon guidance receptors modulates their expression and functions. Contact repulsion by membrane-associated ephrins and Eph receptors was proposed to be facilitated by ectodomain cleavage, but whether this phenomenon is required for axon guidance in vivo is unknown. RESULTS: In support of established models, we find that cleavage of EphA4 promotes cell-cell and growth cone-cell detachment in vitro. Unexpectedly, however, a cleavage resistant isoform of EphA4 is as effective as a wild-type EphA4 in redirecting motor axons in limbs. Mice in which EphA4 cleavage is genetically abolished have motor axon guidance defects, suggesting an important role of EphA4 cleavage in nonneuronal tissues such as the limb mesenchyme target of spinal motor neurons. Indeed, we find that blocking EphA4 cleavage increases expression of full-length EphA4 in limb mesenchyme, which-via cis-attenuation-apparently reduces the effective concentration of ephrinAs capable of triggering EphA4 forward signaling in the motor axons. CONCLUSIONS: We propose that EphA4 cleavage is required to establish the concentration differential of active ephrins in the target tissue that is required for proper axon guidance. Our study reveals a novel mechanism to regulate guidance decision at an intermediate target based on the modulation of ligand availability by the proteolytic processing of the receptor.


Subject(s)
Axons/physiology , Receptor, EphA4/metabolism , Spinal Cord/cytology , Animals , Cell Adhesion , Ephrins/genetics , Ephrins/metabolism , Gene Expression Regulation/physiology , Mice , Mutation , Receptor, EphA4/genetics
10.
J Neurosci ; 33(12): 5399-410, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23516305

ABSTRACT

Axonal branches of the trigeminal ganglion (TG) display characteristic growth and arborization patterns during development. Subsets of TG neurons express different receptors for growth factors, but these are unlikely to explain the unique patterns of axonal arborizations. Intrinsic modulators may restrict or enhance cellular responses to specific ligands and thereby contribute to the development of axon growth patterns. Protein tyrosine phosphatase receptor type O (PTPRO), which is required for Eph receptor-dependent retinotectal development in chick and for development of subsets of trunk sensory neurons in mouse, may be such an intrinsic modulator of TG neuron development. PTPRO is expressed mainly in TrkB-expressing (TrkB(+)) and Ret(+) mechanoreceptors within the TG during embryogenesis. In PTPRO mutant mice, subsets of TG neurons grow longer and more elaborate axonal branches. Cultured PTPRO(-/-) TG neurons display enhanced axonal outgrowth and branching in response to BDNF and GDNF compared with control neurons, indicating that PTPRO negatively controls the activity of BDNF/TrkB and GDNF/Ret signaling. Mouse PTPRO fails to regulate Eph signaling in retinocollicular development and in hindlimb motor axon guidance, suggesting that chick and mouse PTPRO have different substrate specificities. PTPRO has evolved to fine tune growth factor signaling in a cell-type-specific manner and to thereby increase the diversity of signaling output of a limited number of receptor tyrosine kinases to control the branch morphology of developing sensory neurons. The regulation of Eph receptor-mediated developmental processes by protein tyrosine phosphatases has diverged between chick and mouse.


Subject(s)
Axons/physiology , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/metabolism , Pregnancy , Receptor, EphA1/metabolism , Receptor, trkA/metabolism , Receptor, trkC/metabolism , Signal Transduction/physiology , Trigeminal Ganglion/embryology , Trigeminal Nerve/cytology , Trigeminal Nerve/embryology , Trigeminal Nerve/metabolism
11.
Curr Biol ; 20(23): 2150-6, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21109439

ABSTRACT

Despite the abundance of guidance cues in vertebrate nervous systems, little is known about cooperation between them. Motor axons of the lateral motor column (LMC(L)) require two ligand/receptor systems, ephrinA/EphA4 and glial cell line-derived neurotrophic factor (GDNF)/Ret, to project to the dorsal limb. Deletion of either EphA4 or Ret in mice leads to rerouting of a portion of LMC(L) axons to the ventral limb, a phenotype enhanced in EphA4;Ret double mutants. The guidance errors in EphA4 knockouts were attributed to the lack of repulsion from ephrinAs in the ventral mesenchyme. However, it has remained unclear how GDNF, expressed dorsally next to the choice point, acts on motor axons and cooperates with ephrinAs. Here we show that GDNF induces attractive turning of LMC(L) axons. When presented in countergradients, GDNF and ephrinAs cooperate in axon turning, indicating that the receptors Ret and EphA4 invoke opposite effects within the same growth cone. GDNF also acts in a permissive manner by reducing ephrinA-induced collapse and keeping the axons in a growth-competent state. This is the first example of two opposing cues promoting the same trajectory choice at an intermediate target.


Subject(s)
Axons/physiology , Chemotactic Factors/metabolism , Ephrins/metabolism , Extremities/innervation , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Motor Neurons/cytology , Animals , Cells, Cultured , Chemotaxis/physiology , Ephrins/genetics , Female , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism
12.
Curr Biol ; 20(16): 1487-92, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20691595

ABSTRACT

The expression of the RNA-binding factor Fragile X mental retardation protein (FMRP) is disrupted in the most common inherited form of cognitive deficiency in humans. FMRP controls neuronal morphogenesis by mediating the translational regulation and localization of a large number of mRNA targets, and these functions are closely associated with transport of FMRP complexes within neurites by microtubule-based motors. However, the mechanisms that link FMRP to motors and regulate its transport are poorly understood. Here we show that FMRP is complexed with Bicaudal-D (BicD) through a domain in the latter protein that mediates linkage of cargoes with the minus-end-directed motor dynein. We demonstrate in Drosophila that the motility and, surprisingly, levels of FMRP protein are dramatically reduced in BicD mutant neurons, leading to a paucity of FMRP within processes. We also provide functional evidence that BicD and FMRP cooperate to control dendritic morphogenesis in the larval nervous system. Our findings open new perspectives for understanding localized mRNA functions in neurons.


Subject(s)
Drosophila Proteins/physiology , Drosophila/metabolism , Fragile X Mental Retardation Protein/metabolism , Gene Expression Regulation, Developmental , Morphogenesis , Neurons/metabolism , Animals , Brain/embryology , Brain/metabolism , Dendrites/metabolism , Drosophila/embryology , Drosophila Proteins/metabolism , Larva/metabolism , Neurogenesis , Protein Transport
13.
BMC Res Notes ; 3: 24, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20181027

ABSTRACT

BACKGROUND: The importance of non-coding RNAs (ncRNAs) as fine regulators of eukaryotic gene expression has emerged by several studies focusing on microRNAs (miRNAs). miRNAs represent a newly discovered family of non coding-RNAs. They are thought to be crucial players of human hematopoiesis and related tumorigenesis and to represent a potential tool to detect the early stages of cancer. More recently, the expression regulation of numerous long ncRNAs has been linked to cell growth, differentiation and cancer although the molecular mechanism of their function is still unknown.NB4 cells are promyelocytic cells that can be induced to differentiation upon retinoic acid (ATRA) treatment and represent a feasible model to study changes of non coding RNAs expression between cancer cells and their terminally differentiated counterpart. FINDINGS: we screened, by microarray analysis, the expression of 243 miRNAs and 492 human genes transcribing for putative long ncRNAs different from miRNAs in NB4 cells before and after ATRA induced differentiation. Our data show that 8 miRNAs, and 58 long ncRNAs were deregulated by ATRA induced NB4 differentiation. CONCLUSION: our data suggest that ATRA-induced differentiation lead to deregulation of a large number of the ncRNAs that can play regulatory roles in both tumorigenesis and differentiation.

14.
Nucleic Acids Res ; 36(20): 6608-19, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18940871

ABSTRACT

The Epstein-Barr virus (EBV)-encoded latent membrane protein-1 (LMP1), a functional homologue of the tumor necrosis factor receptor family, substantially contributes to EBV's oncogenic potential by activating nuclear factor-kappaB (NF-kappaB). miR-155 is an oncogenic miRNA critical for B-cell maturation and immunoglobulin production in response to antigen. We report that miR-155 expression is much higher in EBV-immortalized B cells than in EBV-negative B cells. LMP1, but not LMP2, up-regulated the expression of miR-155, when transfected in EBV-negative B cells. We analyzed two putative NF-kappaB binding sites in the miR-155 promoter; both sites recruited NF-kappaB complex, in nuclear extract from EBV-immortalized cells. The exogenous expression of LMP1, in EBV-negative background, is temporally correlated to induction of p65 with binding on both NF-kappaB sites and with miR-155 overexpression. The induction of p65 binding together with increased RNA polymerase II binding, confirms that LMP1-mediated activation of miR-155 occurs transcriptionally. In reporter assays, miR-155 promoter lacking NF-kappaB binding sites was no longer activated by LMP1 expression and an intact AP1 site is needed to attain maximum activation. Finally, we demonstrate that LMP1-mediated activation of miR-155 in an EBV-negative background correlates with reduction of protein PU.1, which is a possible miR target.


Subject(s)
B-Lymphocytes/virology , MicroRNAs/genetics , NF-kappa B/metabolism , Transcriptional Activation , Viral Matrix Proteins/metabolism , Animals , Binding Sites , Cell Line , Cells, Cultured , Herpesvirus 4, Human/physiology , Humans , Mice , MicroRNAs/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...