1
|
Harris-Warrick RM, Pecchi E, Drouillas B, Brocard F, Bos R. Effect of size on expression of bistability in mouse spinal motoneurons. J Neurophysiol 2024; 131:577-588. [PMID: 38380829 PMCID: PMC11305636 DOI: 10.1152/jn.00320.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 02/22/2024] Open
Abstract
Bistability in spinal motoneurons supports tonic spike activity in the absence of excitatory drive. Earlier work in adult preparations suggested that smaller motoneurons innervating slow antigravity muscle fibers are more likely to generate bistability for postural maintenance. However, whether large motoneurons innervating fast-fatigable muscle fibers display bistability is still controversial. To address this, we examined the relationship between soma size and bistability in lumbar (L4-L5) ventrolateral α-motoneurons of choline acetyltransferase (ChAT)-green fluorescent protein (GFP) and Hb9-GFP mice during the first 4 wk of life. We found that as neuron size increases, the prevalence of bistability rises. Smaller α-motoneurons lack bistability, whereas larger fast α-motoneurons [matrix metalloproteinase-9 (MMP-9)+/Hb9+] with a soma area ≥ 400 µm2 exhibit significantly higher bistability. Ionic currents associated with bistability, including the persistent Nav1.6 current, the thermosensitive Trpm5 Ca2+-activated Na+ current, and the slowly inactivating Kv1.2 current, also scale with cell size. Serotonin evokes full bistability in large motoneurons with partial bistable properties but not in small motoneurons. Our study provides important insights into the neural mechanisms underlying bistability and how motoneuron size correlates with bistability in mice.NEW & NOTEWORTHY Bistability is not a common feature of all mouse spinal motoneurons. It is absent in small, slow motoneurons but present in most large, fast motoneurons. This difference results from differential expression of ionic currents that enable bistability, which are highly expressed in large motoneurons but small or absent in small motoneurons. These results support a possible role for fast motoneurons in maintenance of tonic posture in addition to their known roles in fast movements.
Collapse
Affiliation(s)
- Ronald M Harris-Warrick
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, United States
| | - Emilie Pecchi
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Benoît Drouillas
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Frédéric Brocard
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Rémi Bos
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| |
Collapse
|
2
|
Harris-Warrick RM, Pecchi E, Drouillas B, Brocard F, Bos R. A size principle for bistability in mouse spinal motoneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.559784. [PMID: 37808773 PMCID: PMC10557784 DOI: 10.1101/2023.09.29.559784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Bistability in spinal motoneurons supports tonic spike activity in the absence of excitatory drive. Earlier work in adult preparations suggested that smaller motoneurons innervating slow antigravity muscle fibers are more likely to generate bistability for postural maintenance. However, whether large motoneurons innervating fast-fatigable muscle fibers display bistability related to postural tone is still controversial. To address this, we examined the relationship between soma size and bistability in lumbar ventrolateral α-motoneurons of ChAT-GFP and Hb9-GFP mice across different developmental stages: neonatal (P2-P7), young (P7-P14) and mature (P21-P25). We found that as neuron size increases, the prevalence of bistability rises. Smaller α-motoneurons lack bistability, while larger fast α-motoneurons (MMP-9 + /Hb9 + ) with a soma area ≥ 400µm 2 exhibit significantly higher bistability. Ionic currents associated with bistability, including the persistent Nav1.6 current, thermosensitive Trpm5 Ca 2+ -activated Na + current and the slowly inactivating Kv1.2 current, also scale with cell size. Serotonin evokes full bistability in large motoneurons with partial bistable properties, but not in small motoneurons. Our study provides important insights into the neural mechanisms underlying bistability and how motoneuron size dictates this process. New and Noteworthy Bistability is not a common feature of all mouse spinal motoneurons. It is absent in small, slow motoneurons but present in most large, fast motoneurons. This difference results from differential expression of ionic currents that enable bistability, which are highly expressed in large motoneurons but small or absent in small motoneurons. These results support a possible role for fast motoneurons in maintenance of tonic posture in addition to their known roles in fast movements.
Collapse
|
3
|
Drouillas B, Brocard C, Zanella S, Bos R, Brocard F. Persistent Nav1.1 and Nav1.6 currents drive spinal locomotor functions through nonlinear dynamics. Cell Rep 2023; 42:113085. [PMID: 37665666 DOI: 10.1016/j.celrep.2023.113085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
Persistent sodium current (INaP) in the spinal locomotor network promotes two distinct nonlinear firing patterns: a self-sustained spiking triggered by a brief excitation in bistable motoneurons and bursting oscillations in interneurons of the central pattern generator (CPG). Here, we identify the NaV channels responsible for INaP and their role in motor behaviors. We report the axonal Nav1.6 as the main molecular player for INaP in lumbar motoneurons. The inhibition of Nav1.6, but not of Nav1.1, in motoneurons impairs INaP, bistability, postural tone, and locomotor performance. In interneurons of the rhythmogenic CPG region, both Nav1.6 and Nav1.1 equally mediate INaP. Inhibition of both channels is required to abolish oscillatory bursting activities and the locomotor rhythm. Overall, Nav1.6 plays a significant role both in posture and locomotion by governing INaP-dependent bistability in motoneurons and working in tandem with Nav1.1 to provide INaP-dependent rhythmogenic properties of the CPG.
Collapse
Affiliation(s)
- Benoît Drouillas
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Cécile Brocard
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Sébastien Zanella
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Rémi Bos
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone, UMR 7289, Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France.
| |
Collapse
|
4
|
Bédécarrats A, Simmers J, Nargeot R. Sodium-mediated plateau potentials in an identified decisional neuron contribute to feeding-related motor pattern genesis in Aplysia. Front Neural Circuits 2023; 17:1200902. [PMID: 37361713 PMCID: PMC10288323 DOI: 10.3389/fncir.2023.1200902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Motivated behaviors such as feeding depend on the functional properties of decision neurons to provide the flexibility required for behavioral adaptation. Here, we analyzed the ionic basis of the endogenous membrane properties of an identified decision neuron (B63) that drive radula biting cycles underlying food-seeking behavior in Aplysia. Each spontaneous bite cycle arises from the irregular triggering of a plateau-like potential and resultant bursting by rhythmic subthreshold oscillations in B63's membrane potential. In isolated buccal ganglion preparations, and after synaptic isolation, the expression of B63's plateau potentials persisted after removal of extracellular calcium, but was completely suppressed in a tetrodotoxin (TTX)- containing bath solution, thereby indicating the contribution of a transmembrane Na+ influx. Potassium outward efflux through tetraethylammonium (TEA)- and calcium-sensitive channels was found to contribute to each plateau's active termination. This intrinsic plateauing capability, in contrast to B63's membrane potential oscillation, was blocked by the calcium-activated non-specific cationic current (ICAN) blocker flufenamic acid (FFA). Conversely, the SERCA blocker cyclopianozic acid (CPA), which abolished the neuron's oscillation, did not prevent the expression of experimentally evoked plateau potentials. These results therefore indicate that the dynamic properties of the decision neuron B63 rely on two distinct mechanisms involving different sub-populations of ionic conductances.
Collapse
|
5
|
Cotinat M, Boquet I, Ursino M, Brocard C, Jouve E, Alberti C, Bensoussan L, Viton JM, Brocard F, Blin O. Riluzole for treating spasticity in patients with chronic traumatic spinal cord injury: Study protocol in the phase ib/iib adaptive multicenter randomized controlled RILUSCI trial. PLoS One 2023; 18:e0276892. [PMID: 36662869 PMCID: PMC9858801 DOI: 10.1371/journal.pone.0276892] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/15/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Satisfactory treatment is often lacking for spasticity, a highly prevalent motor disorder in patients with spinal cord injury (SCI). Low concentrations of riluzole potently reduce the persistent sodium current, the post-SCI increase in which contributes to spasticity. The repurposing of this drug may therefore constitute a useful potential therapeutic option for relieving SCI patients suffering from chronic traumatic spasticity. OBJECTIVE RILUSCI is a phase 1b-2b trial designed to assess whether riluzole is a safe and biologically effective means of managing spasticity in adult patients with traumatic chronic SCI. METHODS In this multicenter double-blind trial, adults (aged 18-65 years) suffering from spasticity after SCI (target enrollment: 90 participants) will be randomly assigned to be given either a placebo or a recommended daily oral dose of riluzole for two weeks. The latter dose will be previously determined in phase 1b of the study by performing double-blind dose-finding tests using a Bayesian continuous reassessment method. The primary endpoint of the trial will be an improvement in the Modified Ashworth Score (MAS) or the Numerical Rating Score (NRS) quantifying spasticity. The secondary outcomes will be based on the safety and pharmacokinetics of riluzole as well as its impact on muscle spasms, pain, bladder dysfunction and quality of life. Analyses will be performed before, during and after the treatment and the placebo-controlled period. CONCLUSION To the best of our knowledge, this clinical trial will be the first to document the safety and efficacy of riluzole as a means of reducing spasticity in patients with chronic SCI. TRIAL REGISTRATION The clinical trial, which is already in progress, was registered on the ClinicalTrials.gov website on August 9, 2016 under the registration number NCT02859792. TRIAL SPONSOR Assistance Publique-Hôpitaux de Marseille.
Collapse
Affiliation(s)
- Maëva Cotinat
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
- Department of Physical and Rehabilitation Medicine, Sainte Marguerite University Hospital, APHM, Marseille, France
| | - Isabelle Boquet
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Moreno Ursino
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Robert Debré, FCRIN PARTNERS Platform, Université de Paris, Sorbonne Paris-Cité, INSERM U1123 and CIC-EC 1426, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université de Paris, F-75006 Paris, France
- Inria, Paris, France
| | - Cécile Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Elisabeth Jouve
- Aix Marseille University, APHM, INSERM, Inst Neurosci Syst, UMR1106, Service de Pharmacologie Clinique et Pharmacovigilance, Marseille, France
| | - Corinne Alberti
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, Centre Hospitalier Universitaire Robert Debré, FCRIN PARTNERS Platform, Université de Paris, Sorbonne Paris-Cité, INSERM U1123 and CIC-EC 1426, Paris, France
| | - Laurent Bensoussan
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
- Institut Universitaire de Réadaptation de Valmante Sud, UGECAM, Marseille, France
| | - Jean-Michel Viton
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
- Department of Physical and Rehabilitation Medicine, Sainte Marguerite University Hospital, APHM, Marseille, France
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Olivier Blin
- Aix Marseille University, APHM, INSERM, Inst Neurosci Syst, UMR1106, Service de Pharmacologie Clinique et Pharmacovigilance, Marseille, France
| |
Collapse
|
6
|
Chalif JI, Mentis GZ. Normal Development and Pathology of Motoneurons: Anatomy, Electrophysiological Properties, Firing Patterns and Circuit Connectivity. ADVANCES IN NEUROBIOLOGY 2022; 28:63-85. [PMID: 36066821 DOI: 10.1007/978-3-031-07167-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This chapter will provide an introduction into motoneuron anatomy, electrophysiological properties, firing patterns focusing on development and also describing several pathological conditions that affect mononeurons. It starts with a historical retrospective describing the early landmark work into motoneurons. The next section lays out the various types of motoneurons (alpha, beta, and gamma) and their subclasses (fast-twitch fatigable, fast-twitch fatigue-resistant, and slow-twitch fatigue resistant), highlighting the functional relevance of this classification scheme. The third section describes the development of motoneurons' passive and active electrophysiological properties. This section also defines the major terms one uses in describing how a neuron functions electrophysiologically. The electrophysiological aspects of a neuron is critical to understanding how it behaves within a circuit and contributes to behavior since the firing of an action potential is how neurons communicate with each other and with muscles. The electrophysiological changes of motoneurons over development underlies how their function changes over the lifetime of an organism. After describing the properties of individual motoneurons, the chapter then turns to revealing how motoneurons interact within complex neural circuits, with other motoneurons as well as sensory neurons, and how these circuits change over development. Finally, this chapter ends with highlighting some recent advances made in motoneuron pathology, focusing on spinal muscular atrophy, amyotrophic lateral sclerosis, and axotomy.
Collapse
Affiliation(s)
- Joshua I Chalif
- Departments of Neurology and Pathology & Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - George Z Mentis
- Departments of Neurology and Pathology & Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA.
| |
Collapse
|
7
|
Trpm5 channels encode bistability of spinal motoneurons and ensure motor control of hindlimbs in mice. Nat Commun 2021; 12:6815. [PMID: 34819493 PMCID: PMC8613399 DOI: 10.1038/s41467-021-27113-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Bistable motoneurons of the spinal cord exhibit warmth-activated plateau potential driven by Na+ and triggered by a brief excitation. The thermoregulating molecular mechanisms of bistability and their role in motor functions remain unknown. Here, we identify thermosensitive Na+-permeable Trpm5 channels as the main molecular players for bistability in mouse motoneurons. Pharmacological, genetic or computational inhibition of Trpm5 occlude bistable-related properties (slow afterdepolarization, windup, plateau potentials) and reduce spinal locomotor outputs while central pattern generators for locomotion operate normally. At cellular level, Trpm5 is activated by a ryanodine-mediated Ca2+ release and turned off by Ca2+ reuptake through the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump. Mice in which Trpm5 is genetically silenced in most lumbar motoneurons develop hindlimb paresis and show difficulties in executing high-demanding locomotor tasks. Overall, by encoding bistability in motoneurons, Trpm5 appears indispensable for producing a postural tone in hindlimbs and amplifying the locomotor output.
Collapse
|
8
|
Sharples SA, Miles GB. Maturation of persistent and hyperpolarization-activated inward currents shapes the differential activation of motoneuron subtypes during postnatal development. eLife 2021; 10:e71385. [PMID: 34783651 PMCID: PMC8641952 DOI: 10.7554/elife.71385] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
The size principle underlies the orderly recruitment of motor units; however, motoneuron size is a poor predictor of recruitment amongst functionally defined motoneuron subtypes. Whilst intrinsic properties are key regulators of motoneuron recruitment, the underlying currents involved are not well defined. Whole-cell patch-clamp electrophysiology was deployed to study intrinsic properties, and the underlying currents, that contribute to the differential activation of delayed and immediate firing motoneuron subtypes. Motoneurons were studied during the first three postnatal weeks in mice to identify key properties that contribute to rheobase and may be important to establish orderly recruitment. We find that delayed and immediate firing motoneurons are functionally homogeneous during the first postnatal week and are activated based on size, irrespective of subtype. The rheobase of motoneuron subtypes becomes staggered during the second postnatal week, which coincides with the differential maturation of passive and active properties, particularly persistent inward currents. Rheobase of delayed firing motoneurons increases further in the third postnatal week due to the development of a prominent resting hyperpolarization-activated inward current. Our results suggest that motoneuron recruitment is multifactorial, with recruitment order established during postnatal development through the differential maturation of passive properties and sequential integration of persistent and hyperpolarization-activated inward currents.
Collapse
Affiliation(s)
- Simon A Sharples
- School of Psychology and Neuroscience, University of St AndrewsSt AndrewsUnited Kingdom
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St AndrewsSt AndrewsUnited Kingdom
| |
Collapse
|
9
|
Buntschu S, Tscherter A, Heidemann M, Streit J. Critical Components for Spontaneous Activity and Rhythm Generation in Spinal Cord Circuits in Culture. Front Cell Neurosci 2020; 14:81. [PMID: 32410961 PMCID: PMC7198714 DOI: 10.3389/fncel.2020.00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/19/2020] [Indexed: 11/23/2022] Open
Abstract
Neuronal excitability contributes to rhythm generation in central pattern generating networks (CPGs). In spinal cord CPGs, such intrinsic excitability partly relies on persistent sodium currents (INaP), whereas respiratory CPGs additionally depend on calcium-activated cation currents (ICAN). Here, we investigated the contributions of INaP and ICAN to spontaneous rhythm generation in neuronal networks of the spinal cord and whether they mainly involve Hb9 neurons. We used cultures of ventral and transverse slices from the E13-14 embryonic rodent lumbar spinal cord on multielectrode arrays (MEAs). All cultures showed spontaneous bursts of network activity. Blocking synaptic excitation with the AMPA receptor antagonist CNQX suppressed spontaneous network bursts and left asynchronous intrinsic activity at about 30% of the electrodes. Such intrinsic activity was completely blocked at all electrodes by both the INaP blocker riluzole as well as by the ICAN blocker flufenamic acid (FFA) and the more specific TRPM4 channel antagonist 9-phenanthrol. All three antagonists also suppressed spontaneous bursting completely and strongly reduced stimulus-evoked bursts. Also, FFA reduced repetitive spiking that was induced in single neurons by injection of depolarizing current pulses to few spikes. Other antagonists of unspecific cation currents or calcium currents had no suppressing effects on either intrinsic activity (gadolinium chloride) or spontaneous bursting (the TRPC channel antagonists clemizole and ML204 and the T channel antagonist TTA-P2). Combined patch-clamp and MEA recordings showed that Hb9 interneurons were activated by network bursts but could not initiate them. Together these findings suggest that both INaP through Na+-channels and ICAN through putative TRPM4 channels contribute to spontaneous intrinsic and repetitive spiking in spinal cord neurons and thereby to the generation of network bursts.
Collapse
Affiliation(s)
| | | | | | - Jürg Streit
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
10
|
Goyal R, Spencer KA, Borodinsky LN. From Neural Tube Formation Through the Differentiation of Spinal Cord Neurons: Ion Channels in Action During Neural Development. Front Mol Neurosci 2020; 13:62. [PMID: 32390800 PMCID: PMC7193536 DOI: 10.3389/fnmol.2020.00062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022] Open
Abstract
Ion channels are expressed throughout nervous system development. The type and diversity of conductances and gating mechanisms vary at different developmental stages and with the progressive maturational status of neural cells. The variety of ion channels allows for distinct signaling mechanisms in developing neural cells that in turn regulate the needed cellular processes taking place during each developmental period. These include neural cell proliferation and neuronal differentiation, which are crucial for developmental events ranging from the earliest steps of morphogenesis of the neural tube through the establishment of neuronal circuits. Here, we compile studies assessing the ontogeny of ionic currents in the developing nervous system. We then review work demonstrating a role for ion channels in neural tube formation, to underscore the necessity of the signaling downstream ion channels even at the earliest stages of neural development. We discuss the function of ion channels in neural cell proliferation and neuronal differentiation and conclude with how the regulation of all these morphogenetic and cellular processes by electrical activity enables the appropriate development of the nervous system and the establishment of functional circuits adapted to respond to a changing environment.
Collapse
Affiliation(s)
- Raman Goyal
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Kira A Spencer
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
11
|
Kocovic DM, Limaye PV, Colburn LCH, Singh MB, Milosevic MM, Tadic J, Petronijevic M, Vrzic-Petronijevic S, Andjus PR, Antic SD. Cadmium versus Lanthanum Effects on Spontaneous Electrical Activity and Expression of Connexin Isoforms Cx26, Cx36, and Cx45 in the Human Fetal Cortex. Cereb Cortex 2020; 30:1244-1259. [PMID: 31408166 PMCID: PMC7132928 DOI: 10.1093/cercor/bhz163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 12/29/2022] Open
Abstract
Electrical activity is important for brain development. In brain slices, human subplate neurons exhibit spontaneous electrical activity that is highly sensitive to lanthanum. Based on the results of pharmacological experiments in human fetal tissue, we hypothesized that hemichannel-forming connexin (Cx) isoforms 26, 36, and 45 would be expressed on neurons in the subplate (SP) zone. RNA sequencing of dissected human cortical mantles at ages of 17-23 gestational weeks revealed that Cx45 has the highest expression, followed by Cx36 and Cx26. The levels of Cx and pannexin expression between male and female fetal cortices were not significantly different. Immunohistochemical analysis detected Cx45- and Cx26-expressing neurons in the upper segment of the SP zone. Cx45 was present on the cell bodies of human SP neurons, while Cx26 was found on both cell bodies and dendrites. Cx45, Cx36, and Cx26 were strongly expressed in the cortical plate, where newborn migrating neurons line up to form cortical layers. New information about the expression of 3 "neuronal" Cx isoforms in each cortical layer/zone (e.g., SP, cortical plate) and pharmacological data with cadmium and lanthanum may improve our understanding of the cellular mechanisms underlying neuronal development in human fetuses and potential vulnerabilities.
Collapse
Affiliation(s)
- Dusica M Kocovic
- Faculty of Biology, University of Belgrade, Belgrade 11000, Serbia
| | - Pallavi V Limaye
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Lauren C H Colburn
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Mandakini B Singh
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Milena M Milosevic
- Faculty of Biology, University of Belgrade, Belgrade 11000, Serbia
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| | - Jasmina Tadic
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | | | | | - Pavle R Andjus
- Faculty of Biology, University of Belgrade, Belgrade 11000, Serbia
| | - Srdjan D Antic
- Institute for Systems Genomics, Stem Cell Institute, Department of Neuroscience, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
12
|
Plantier V, Sanchez-Brualla I, Dingu N, Brocard C, Liabeuf S, Gackière F, Brocard F. Calpain fosters the hyperexcitability of motoneurons after spinal cord injury and leads to spasticity. eLife 2019; 8:e51404. [PMID: 31815668 PMCID: PMC6927741 DOI: 10.7554/elife.51404] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Up-regulation of the persistent sodium current (INaP) and down-regulation of the potassium/chloride extruder KCC2 lead to spasticity after spinal cord injury (SCI). We here identified calpain as the driver of the up- and down-regulation of INaP and KCC2, respectively, in neonatal rat lumbar motoneurons. Few days after SCI, neonatal rats developed behavioral signs of spasticity with the emergence of both hyperreflexia and abnormal involuntary muscle contractions on hindlimbs. At the same time, in vitro isolated lumbar spinal cords became hyperreflexive and displayed numerous spontaneous motor outputs. Calpain-I expression paralleled with a proteolysis of voltage-gated sodium (Nav) channels and KCC2. Acute inhibition of calpains reduced this proteolysis, restored the motoneuronal expression of Nav and KCC2, normalized INaP and KCC2 function, and curtailed spasticity. In sum, by up- and down-regulating INaP and KCC2, the calpain-mediated proteolysis of Nav and KCC2 drives the hyperexcitability of motoneurons which leads to spasticity after SCI.
Collapse
Affiliation(s)
- Vanessa Plantier
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Irene Sanchez-Brualla
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Nejada Dingu
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Cécile Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Sylvie Liabeuf
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Florian Gackière
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRSMarseilleFrance
| |
Collapse
|
13
|
Hagger-Vaughan N, Storm JF. Synergy of Glutamatergic and Cholinergic Modulation Induces Plateau Potentials in Hippocampal OLM Interneurons. Front Cell Neurosci 2019; 13:508. [PMID: 31780902 PMCID: PMC6861217 DOI: 10.3389/fncel.2019.00508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/28/2019] [Indexed: 01/18/2023] Open
Abstract
Oriens-lacunosum moleculare (OLM) cells are hippocampal inhibitory interneurons that are implicated in the regulation of information flow in the CA1 circuit, inhibiting cortical inputs to distal pyramidal cell dendrites, whilst disinhibiting CA3 inputs to pyramidal cells. OLM cells express metabotropic cholinergic (mAChR) and glutamatergic (mGluR) receptors, so modulation of these cells via these receptors may contribute to switching between functional modes of the hippocampus. Using a transgenic mouse line to identify OLM cells, we found that both mAChR and mGluR activation caused the cells to exhibit long-lasting depolarizing plateau potentials following evoked spike trains. Both mAChR- and mGluR-induced plateau potentials were eliminated by blocking transient receptor potential (TRP) channels, and were dependent on intracellular calcium concentration and calcium entry. Pharmacological tests indicated that Group I mGluRs are responsible for the glutamatergic induction of plateaus. There was also a pronounced synergy between the cholinergic and glutamatergic modulation, plateau potentials being generated by agonists applied together at concentrations too low to elicit any change when applied individually. This synergy could enable OLM cells to function as coincidence detectors of different neuromodulatory systems, leading to their enhanced and prolonged activation and a functional change in information flow within the hippocampus.
Collapse
Affiliation(s)
| | - Johan F. Storm
- Brain Signaling Laboratory, Section for Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Growth at Cold Temperature Increases the Number of Motor Neurons to Optimize Locomotor Function. Curr Biol 2019; 29:1787-1799.e5. [PMID: 31130453 PMCID: PMC7501754 DOI: 10.1016/j.cub.2019.04.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/26/2019] [Accepted: 04/29/2019] [Indexed: 01/18/2023]
Abstract
During vertebrate development, spinal neurons differentiate and connect to generate a system that performs sensorimotor functions critical for survival. Spontaneous Ca2+ activity regulates different aspects of spinal neuron differentiation. It is unclear whether environmental factors can modulate this Ca2+ activity in developing spinal neurons to alter their specialization and ultimately adjust sensorimotor behavior to fit the environment. Here, we show that growing Xenopus laevis embryos at cold temperatures results in an increase in the number of spinal motor neurons in larvae. This change in spinal cord development optimizes the escape response to gentle touch of animals raised in and tested at cold temperatures. The cold-sensitive channel TRPM8 increases Ca2+ spike frequency of developing ventral spinal neurons, which in turn regulates expression of the motor neuron master transcription factor HB9. TRPM8 is necessary for the increase in motor neuron number of animals raised in cold temperatures and for their enhanced sensorimotor behavior when tested at cold temperatures. These findings suggest the environment modulates neuronal differentiation to optimize the behavior of the developing organism. Spencer et al. discover that Xenopus larvae reared in cold temperature are better equipped to escape upon touch at cold temperature relative to warm-grown siblings. This advantage is dependent on the cold-sensitive channel TRPM8, which is necessary for increased Ca2+ spike frequency in embryonic spinal neurons, their differentiation, and survival.
Collapse
|
15
|
|
16
|
Abstract
Motoneurons are known to be an essential component of central pattern generators in invertebrates, but it is only recently that they have been shown to play a similar role in vertebrate locomotor circuits. Here, we review early experiments implicating motoneurons in the genesis of spontaneous motor activity in development and more recent experiments identifying motoneurons as important regulators of locomotor activity in the adult zebrafish and in the neonatal mouse spinal cord. We discuss the mechanisms responsible for these actions, the experimental challenges in studying the role of motoneurons in the mammalian spinal cord and the functional significance of the excitatory influence of motoneuron activity on locomotor behavior.
Collapse
|
17
|
Dingu N, Deumens R, Taccola G. Afferent Input Induced by Rhythmic Limb Movement Modulates Spinal Neuronal Circuits in an Innovative Robotic In Vitro Preparation. Neuroscience 2018; 394:44-59. [PMID: 30342198 DOI: 10.1016/j.neuroscience.2018.10.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/30/2022]
Abstract
Locomotor patterns are mainly modulated by afferent feedback, but its actual contribution to spinal network activity during continuous passive limb training is still unexplored. To unveil this issue, we devised a robotic in vitro setup (Bipedal Induced Kinetic Exercise, BIKE) to induce passive pedaling, while simultaneously recording low-noise ventral and dorsal root (VR and DR) potentials in isolated neonatal rat spinal cords with hindlimbs attached. As a result, BIKE evoked rhythmic afferent volleys from DRs, reminiscent of pedaling speed. During BIKE, spontaneous VR activity remained unchanged, while a DR rhythmic component paired the pedaling pace. Moreover, BIKE onset rarely elicited brief episodes of fictive locomotion (FL) and, when trains of electrical pulses were simultaneously applied to a DR, it increased the amplitude, but not the number, of FL cycles. When BIKE was switched off after a 30-min training, the number of electrically induced FL oscillations was transitorily facilitated, without affecting VR reflexes or DR potentials. However, 90 min of BIKE no longer facilitated FL, but strongly depressed area of VR reflexes and stably increased antidromic DR discharges. Patch clamp recordings from single motoneurons after 90-min sessions indicated an increased frequency of both fast- and slow-decaying synaptic input to motoneurons. In conclusion, hindlimb rhythmic and alternated pedaling for different durations affects distinct dorsal and ventral spinal networks by modulating excitatory and inhibitory input to motoneurons. These results suggest defining new parameters for effective neurorehabilitation that better exploits spinal circuit activity.
Collapse
Affiliation(s)
- Nejada Dingu
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, TS, Italy; SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione (IMFR), via Gervasutta 48, Udine, UD, Italy
| | - Ronald Deumens
- Institute of Neuroscience, Université catholique de Louvain, Av. Hippocrate 54, Brussels, Belgium
| | - Giuliano Taccola
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, TS, Italy; SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione (IMFR), via Gervasutta 48, Udine, UD, Italy.
| |
Collapse
|
18
|
Dai Y, Cheng Y, Fedirchuk B, Jordan LM, Chu J. Motoneuron output regulated by ionic channels: a modeling study of motoneuron frequency-current relationships during fictive locomotion. J Neurophysiol 2018; 120:1840-1858. [PMID: 30044677 DOI: 10.1152/jn.00068.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cat lumbar motoneurons display changes in membrane properties during fictive locomotion. These changes include reduction of input resistance and afterhyperpolarization, hyperpolarization of voltage threshold, and voltage-dependent excitation of the motoneurons. The state-dependent alteration of membrane properties leads to dramatic changes in frequency-current (F-I) relationship. The mechanism underlying these changes remains unknown. Using a motoneuron model combined with electrophysiological data, we investigated the channel mechanisms underlying the regulation of motoneuronal excitability and motor output. Simulation results showed that upregulation of transient sodium, persistent sodium, or Cav1.3 calcium conductances or downregulation of calcium-activated potassium or KCNQ/Kv7 potassium conductances could increase motoneuronal excitability and motor output through hyperpolarizing (left shifting) the F-I relationships or increasing the F-I slopes, whereas downregulation of input resistance or upregulation of potassium-mediated leak conductance produced the opposite effects. The excitatory phase of locomotor drive potentials (LDPs) also substantially hyperpolarized the F-I relationships and increased the F-I slopes, whereas the inhibitory phase of the LDPs had opposite effects to a similar extent. The simulation results also showed that none of the individual channel modulations could produce all the changes in the F-I relationships. The effects of modulation of Cav1.3 and KCNQ/Kv7 on F-I relationships were supported by slice experiments with the Cav1.3 agonist Bay K8644 and the KCNQ/Kv7 antagonist XE-991. The conclusion is that the varying changes in F-I relationships during fictive locomotion could be regulated by multichannel modulations. This study provides insight into the ionic basis for control of motor output in walking. NEW & NOTEWORTHY Mammalian spinal motoneurons have their excitability adapted to facilitate recruitment and firing during locomotion. Cat lumbar motoneurons display dramatic changes in membrane properties during fictive locomotion. These changes lead to a varying alteration of frequency-current relationship. The mechanisms underlying the changes remain unknown. In particular, little is known about the ionic basis for regulation of motoneuronal excitability and thus control of the motor output for walking by the spinal motor system.
Collapse
Affiliation(s)
- Yue Dai
- Shanghai Key Laboratory of Multidimensional Information Processing, School of Information Science Technology, East China Normal University , Shanghai , People's Republic of China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education and Health Care, East China Normal University , Shanghai , People's Republic of China
| | - Yi Cheng
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education and Health Care, East China Normal University , Shanghai , People's Republic of China
| | - Brent Fedirchuk
- Department of Physiology, University of Manitoba , Winnipeg, Manitoba , Canada
| | - Larry M Jordan
- Department of Physiology, University of Manitoba , Winnipeg, Manitoba , Canada
| | - Junhao Chu
- Shanghai Key Laboratory of Multidimensional Information Processing, School of Information Science Technology, East China Normal University , Shanghai , People's Republic of China
| |
Collapse
|
19
|
Persistent Sodium Current Drives Excitability of Immature Renshaw Cells in Early Embryonic Spinal Networks. J Neurosci 2018; 38:7667-7682. [PMID: 30012693 DOI: 10.1523/jneurosci.3203-17.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 06/14/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022] Open
Abstract
Spontaneous network activity (SNA) emerges in the spinal cord (SC) before the formation of peripheral sensory inputs and central descending inputs. SNA is characterized by recurrent giant depolarizing potentials (GDPs). Because GDPs in motoneurons (MNs) are mainly evoked by prolonged release of GABA, they likely necessitate sustained firing of interneurons. To address this issue we analyzed, as a model, embryonic Renshaw cell (V1R) activity at the onset of SNA (E12.5) in the embryonic mouse SC (both sexes). V1R are one of the interneurons known to contact MNs, which are generated early in the embryonic SC. Here, we show that V1R already produce GABA in E12.5 embryo, and that V1R make synaptic-like contacts with MNs and have putative extrasynaptic release sites, while paracrine release of GABA occurs at this developmental stage. In addition, we discovered that V1R are spontaneously active during SNA and can already generate several intrinsic activity patterns including repetitive-spiking and sodium-dependent plateau potential that rely on the presence of persistent sodium currents (INap). This is the first demonstration that INap is present in the embryonic SC and that this current can control intrinsic activation properties of newborn interneurons in the SC of mammalian embryos. Finally, we found that 5 μm riluzole, which is known to block INaP, altered SNA by reducing episode duration and increasing inter-episode interval. Because SNA is essential for neuronal maturation, axon pathfinding, and synaptogenesis, the presence of INaP in embryonic SC neurons may play a role in the early development of mammalian locomotor networks.SIGNIFICANCE STATEMENT The developing spinal cord (SC) exhibits spontaneous network activity (SNA) involved in the building of nascent locomotor circuits in the embryo. Many studies suggest that SNA depends on the rhythmic release of GABA, yet intracellular recordings of GABAergic neurons have never been performed at the onset of SNA in the SC. We first discovered that embryonic Renshaw cells (V1R) are GABAergic at E12.5 and spontaneously active during SNA. We uncover a new role for persistent sodium currents (INaP) in driving plateau potential in V1R and in SNA patterning in the embryonic SC. Our study thus sheds light on a role for INaP in the excitability of V1R and the developing SC.
Collapse
|
20
|
Bos R, Harris-Warrick RM, Brocard C, Demianenko LE, Manuel M, Zytnicki D, Korogod SM, Brocard F. Kv1.2 Channels Promote Nonlinear Spiking Motoneurons for Powering Up Locomotion. Cell Rep 2018; 22:3315-3327. [PMID: 29562186 PMCID: PMC5907934 DOI: 10.1016/j.celrep.2018.02.093] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/02/2018] [Accepted: 02/23/2018] [Indexed: 01/15/2023] Open
Abstract
Spinal motoneurons are endowed with nonlinear spiking behaviors manifested by a spike acceleration whose functional significance remains uncertain. Here, we show in rodent lumbar motoneurons that these nonlinear spiking properties do not rely only on activation of dendritic nifedipine-sensitive L-type Ca2+ channels, as assumed for decades, but also on the slow inactivation of a nifedipine-sensitive K+ current mediated by Kv1.2 channels that are highly expressed in axon initial segments. Specifically, the pharmacological and computational inhibition of Kv1.2 channels occluded the spike acceleration of rhythmically active motoneurons and the correlated slow buildup of rhythmic motor output recorded at the onset of locomotor-like activity. This study demonstrates that slow inactivation of Kv1.2 channels provides a potent gain control mechanism in mammalian spinal motoneurons and has a behavioral role in enhancing locomotor drive during the transition from immobility to steady-state locomotion.
Collapse
Affiliation(s)
- Rémi Bos
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | | | - Cécile Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Liliia E Demianenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Marin Manuel
- Centre de Neurophysique, Physiologie et Pathologie, UMR 8119, CNRS/Université Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Daniel Zytnicki
- Centre de Neurophysique, Physiologie et Pathologie, UMR 8119, CNRS/Université Paris Descartes, 45 rue des Saints-Pères, 75270 Paris Cedex 06, France
| | - Sergiy M Korogod
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and Centre National de la Recherche Scientifique (CNRS), Marseille, France.
| |
Collapse
|
21
|
White SH, Sturgeon RM, Gu Y, Nensi A, Magoski NS. Tyrosine Phosphorylation Determines Afterdischarge Initiation by Regulating an Ionotropic Cholinergic Receptor. Neuroscience 2018; 372:273-288. [PMID: 29306054 DOI: 10.1016/j.neuroscience.2017.12.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/30/2017] [Accepted: 12/26/2017] [Indexed: 12/12/2022]
Abstract
Changes to neuronal activity often involve a rapid and precise transition from low to high excitability. In the marine snail, Aplysia, the bag cell neurons control reproduction by undergoing an afterdischarge, which begins with synaptic input releasing acetylcholine to open an ionotropic cholinergic receptor. Gating of this receptor causes depolarization and a shift from silence to continuous action potential firing, leading to the neuroendocrine secretion of egg-laying hormone and ovulation. At the onset of the afterdischarge, there is a rise in intracellular Ca2+, followed by both protein kinase C (PKC) activation and tyrosine dephosphorylation. To determine whether these signals influence the acetylcholine ionotropic receptor, we examined the bag cell neuron cholinergic response both in culture and isolated clusters using whole-cell and/or sharp-electrode electrophysiology. The acetylcholine-induced current was not altered by increasing intracellular Ca2+ via voltage-gated Ca2+ channels, clamping intracellular Ca2+ with exogenous Ca2+ buffers, or activating PKC with phorbol esters. However, lowering phosphotyrosine levels by inhibiting tyrosine kinases both reduced the cholinergic current and prevented acetylcholine from triggering action potentials or afterdischarge-like bursts. In other systems, acetylcholine receptors are often modulated by multiple signals, but bag cell neurons appear to be more restrictive in this regard. Prior work finds that, as the afterdischarge proceeds, tyrosine dephosphorylation leads to biophysical alterations that promote persistent firing. Because this firing is subsequent to the cholinergic input, inhibiting the acetylcholine receptor may represent a means of properly orchestrating synaptically induced changes in excitability.
Collapse
Affiliation(s)
- Sean H White
- Department of Biomedical and Molecular Sciences, Physiology and Neuroscience Graduate Programs, Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Raymond M Sturgeon
- Department of Biomedical and Molecular Sciences, Physiology and Neuroscience Graduate Programs, Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Yueling Gu
- Department of Biomedical and Molecular Sciences, Physiology and Neuroscience Graduate Programs, Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Alysha Nensi
- Department of Biomedical and Molecular Sciences, Physiology and Neuroscience Graduate Programs, Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Neil S Magoski
- Department of Biomedical and Molecular Sciences, Physiology and Neuroscience Graduate Programs, Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
22
|
Coslovich T, Brumley MR, D'Angelo G, Della Mora A, Swann HE, Ortolani F, Taccola G. Histamine modulates spinal motoneurons and locomotor circuits. J Neurosci Res 2017; 96:889-900. [PMID: 29114923 DOI: 10.1002/jnr.24195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/01/2017] [Accepted: 10/16/2017] [Indexed: 12/20/2022]
Abstract
Spinal motoneurons and locomotor networks are regulated by monoamines, among which, the contribution of histamine has yet to be fully addressed. The present study investigates histaminergic regulation of spinal activity, combining intra- and extracellular electrophysiological recordings from neonatal rat spinal cord in vitro preparations. Histamine dose-dependently and reversibly generated motoneuron depolarization and action potential firing. Histamine (20 µM) halved the area of dorsal root reflexes and always depolarized motoneurons. The majority of cells showed a transitory repolarization, while 37% showed a sustained depolarization maintained with intense firing. Extracellularly, histamine depolarized ventral roots (VRs), regardless of blockage of ionotropic glutamate receptors. Initial, transient glutamate-mediated bursting was synchronous among VRs, with some bouts of locomotor activity in a subgroup of preparations. After washout, the amplitude of spontaneous tonic discharges increased. No desensitization or tachyphylaxis appeared after long perfusion or serial applications of histamine. On the other hand, histamine induced single motoneuron and VR depolarization, even in the presence of tetrodotoxin (TTX). During chemically induced fictive locomotion (FL), histamine depolarized VRs. Histamine dose-dependently increased rhythm periodicity and reduced cycle amplitude until near suppression. This study demonstrates that histamine induces direct motoneuron membrane depolarization and modulation of locomotor output, indicating new potential targets for locomotor neurorehabilitation.
Collapse
Affiliation(s)
- Tamara Coslovich
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265 Trieste, (TS), Italy.,SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione (IMFR), via Gervasutta 48 Udine (UD), Italy
| | | | - Giuseppe D'Angelo
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265 Trieste, (TS), Italy.,SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione (IMFR), via Gervasutta 48 Udine (UD), Italy
| | - Alberto Della Mora
- Department of Experimental Clinical Medicine, University of Udine, Piazzale Kolbe 3 Udine, Italy
| | | | - Fulvia Ortolani
- Department of Experimental Clinical Medicine, University of Udine, Piazzale Kolbe 3 Udine, Italy
| | - Giuliano Taccola
- Neuroscience Department, International School for Advanced Studies (SISSA), via Bonomea 265 Trieste, (TS), Italy.,SPINAL (Spinal Person Injury Neurorehabilitation Applied Laboratory), Istituto di Medicina Fisica e Riabilitazione (IMFR), via Gervasutta 48 Udine (UD), Italy
| |
Collapse
|
23
|
Jean-Xavier C, Sharples SA, Mayr KA, Lognon AP, Whelan PJ. Retracing your footsteps: developmental insights to spinal network plasticity following injury. J Neurophysiol 2017; 119:521-536. [PMID: 29070632 DOI: 10.1152/jn.00575.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
During development of the spinal cord, a precise interaction occurs between descending projections and sensory afferents, with spinal networks that lead to expression of coordinated motor output. In the rodent, during the last embryonic week, motor output first occurs as regular bursts of spontaneous activity, progressing to stochastic patterns of episodes that express bouts of coordinated rhythmic activity perinatally. Locomotor activity becomes functionally mature in the 2nd postnatal wk and is heralded by the onset of weight-bearing locomotion on the 8th and 9th postnatal day. Concomitantly, there is a maturation of intrinsic properties and key conductances mediating plateau potentials. In this review, we discuss spinal neuronal excitability, descending modulation, and afferent modulation in the developing rodent spinal cord. In the adult, plastic mechanisms are much more constrained but become more permissive following neurotrauma, such as spinal cord injury. We discuss parallel mechanisms that contribute to maturation of network function during development to mechanisms of pathological plasticity that contribute to aberrant motor patterns, such as spasticity and clonus, which emerge following central injury.
Collapse
Affiliation(s)
- C Jean-Xavier
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Comparative Biology and Experimental Medicine, University of Calgary , Calgary, Alberta , Canada
| | - S A Sharples
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Neuroscience, University of Calgary , Calgary, Alberta , Canada
| | - K A Mayr
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Neuroscience, University of Calgary , Calgary, Alberta , Canada
| | - A P Lognon
- Department of Comparative Biology and Experimental Medicine, University of Calgary , Calgary, Alberta , Canada
| | - P J Whelan
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada.,Department of Comparative Biology and Experimental Medicine, University of Calgary , Calgary, Alberta , Canada
| |
Collapse
|
24
|
Dendritic and Axonal L-Type Calcium Channels Cooperate to Enhance Motoneuron Firing Output during Drosophila Larval Locomotion. J Neurosci 2017; 37:10971-10982. [PMID: 28986465 DOI: 10.1523/jneurosci.1064-17.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/18/2017] [Accepted: 09/26/2017] [Indexed: 11/21/2022] Open
Abstract
Behaviorally adequate neuronal firing patterns are critically dependent on the specific types of ion channel expressed and on their subcellular localization. This study combines in situ electrophysiology with genetic and pharmacological intervention in larval Drosophila melanogaster of both sexes to address localization and function of L-type like calcium channels in motoneurons. We demonstrate that Dmca1D (Cav1 homolog) L-type like calcium channels localize to both the somatodendritic and the axonal compartment of larval crawling motoneurons. In situ patch-clamp recordings in genetic mosaics reveal that Dmca1D channels increase burst duration and maximum intraburst firing frequencies during crawling-like motor patterns in semi-intact animals. Genetic and acute pharmacological manipulations suggest that prolonged burst durations are caused by dendritically localized Dmca1D channels, which activate upon cholinergic synaptic input and amplify EPSPs, thus indicating a conserved function of dendritic L-type channels from Drosophila to vertebrates. By contrast, maximum intraburst firing rates require axonal calcium influx through Dmca1D channels, likely to enhance sodium channel de-inactivation via a fast afterhyperpolarization through BK channel activation. Therefore, in unmyelinated Drosophila motoneurons different functions of axonal and dendritic L-type like calcium channels likely operate synergistically to maximize firing output during locomotion.SIGNIFICANCE STATEMENT Nervous system function depends on the specific excitabilities of different types of neurons. Excitability is largely shaped by different combinations of voltage-dependent ion channels. Despite a high degree of conservation, the huge diversity of ion channel types and their differential localization pose challenges in assigning distinct functions to specific channels across species. We find a conserved role, from fruit flies to mammals, for L-type calcium channels in augmenting motoneuron excitability. As in spinal cord, dendritic L-type channels amplify excitatory synaptic input. In contrast to spinal motoneurons, axonal L-type channels enhance firing rates in unmyelinated Drosophila motoraxons. Therefore, enhancing motoneuron excitability by L-type channels seems an old strategy, but localization and interactions with other channels are tuned to species-specific requirements.
Collapse
|
25
|
Plantier V, Brocard F. [Calpain as a new therapeutic target for treating spasticity after a spinal cord injury]. Med Sci (Paris) 2017; 33:629-636. [PMID: 28990565 DOI: 10.1051/medsci/20173306020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
After a spinal cord injury (SCI), patients develop spasticity, a motor disorder characterized by hyperreflexia and stiffness of muscles. Spasticity results from alterations in motoneurons with an upregulation of their persistent sodium current (I NaP), simultaneously with a disinhibition caused by a reduction of expression of chloride (Cl-) co-transporters KCC2. Until recently the origin of alterations was unknown. After reviewing pathophysiology of spasticity, the manuscript relates our recent work showing a tight relationship between the calpain-dependent proteolysis of voltage-gated sodium channels, the upregulation of I NaP and spasticity following SCI. We also discuss KCC2 as a substrate of calpains which may contribute to the disinhibition of motoneurons below the lesion. This led us to consider the proteolytic cleavage of both sodium channels and KCC2 as the upstream mechanism contributing to the development of spasticity after SCI.
Collapse
Affiliation(s)
- Vanessa Plantier
- Équipe P3M, Institut de Neurosciences de la Timone, UMR7289, Aix Marseille Université et Centre National de la Recherche Scientifique (CNRS), Marseille, France
| | - Frédéric Brocard
- Équipe P3M, Institut de Neurosciences de la Timone, UMR7289, Aix Marseille Université et Centre National de la Recherche Scientifique (CNRS), Marseille, France
| |
Collapse
|
26
|
Svensson E, Jeffreys H, Li WC. The modulation of two motor behaviors by persistent sodium currents in Xenopus laevis tadpoles. J Neurophysiol 2017; 118:121-130. [PMID: 28331009 PMCID: PMC5494356 DOI: 10.1152/jn.00755.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/22/2017] [Accepted: 03/22/2017] [Indexed: 12/12/2022] Open
Abstract
We have characterized persistent sodium currents in three groups of spinal neurons and their role in shaping spiking activity in the Xenopus tadpole. We then attempted to evaluate the role of persistent sodium currents in regulating tadpole swimming and struggling motor outputs by using low concentrations of the persistent sodium current antagonist riluzole. Persistent sodium currents (INaP) are common in neuronal circuitries and have been implicated in several diseases, such as amyotrophic lateral sclerosis (ALS) and epilepsy. However, the role of INaP in the regulation of specific behaviors is still poorly understood. In this study we have characterized INaP and investigated its role in the swimming and struggling behavior of Xenopus tadpoles. INaP was identified in three groups of neurons, namely, sensory Rohon-Beard neurons (RB neurons), descending interneurons (dINs), and non-dINs (neurons rhythmically active in swimming). All groups of neurons expressed INaP, but the currents differed in decay time constants, amplitudes, and the membrane potential at which INaP peaked. Low concentrations (1 µM) of the INaP blocker riluzole blocked INaP ~30% and decreased the excitability of the three neuron groups without affecting spike amplitudes or cellular input resistances. Riluzole reduced the number of rebound spikes in dINs and depressed repetitive firing in RB neurons and non-dINs. At the behavior level, riluzole at 1 µM shortened fictive swimming episodes. It also reduced the number of action potentials neurons fired on each struggling cycle. The results show that INaP may play important modulatory roles in motor behaviors. NEW & NOTEWORTHY We have characterized persistent sodium currents in three groups of spinal neurons and their role in shaping spiking activity in the Xenopus tadpole. We then attempted to evaluate the role of persistent sodium currents in regulating tadpole swimming and struggling motor outputs by using low concentrations of the persistent sodium current antagonist riluzole.
Collapse
Affiliation(s)
- Erik Svensson
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, United Kingdom
| | - Hugo Jeffreys
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, United Kingdom
| | - Wen-Chang Li
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, Fife, United Kingdom
| |
Collapse
|
27
|
Côté MP, Murray M, Lemay MA. Rehabilitation Strategies after Spinal Cord Injury: Inquiry into the Mechanisms of Success and Failure. J Neurotrauma 2016; 34:1841-1857. [PMID: 27762657 DOI: 10.1089/neu.2016.4577] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Body-weight supported locomotor training (BWST) promotes recovery of load-bearing stepping in lower mammals, but its efficacy in individuals with a spinal cord injury (SCI) is limited and highly dependent on injury severity. While animal models with complete spinal transections recover stepping with step-training, motor complete SCI individuals do not, despite similarly intensive training. In this review, we examine the significant differences between humans and animal models that may explain this discrepancy in the results obtained with BWST. We also summarize the known effects of SCI and locomotor training on the muscular, motoneuronal, interneuronal, and supraspinal systems in human and non-human models of SCI and address the potential causes for failure to translate to the clinic. The evidence points to a deficiency in neuronal activation as the mechanism of failure, rather than muscular insufficiency. While motoneuronal and interneuronal systems cannot be directly probed in humans, the changes brought upon by step-training in SCI animal models suggest a beneficial re-organization of the systems' responsiveness to descending and afferent feedback that support locomotor recovery. The literature on partial lesions in humans and animal models clearly demonstrate a greater dependency on supraspinal input to the lumbar cord in humans than in non-human mammals for locomotion. Recent results with epidural stimulation that activates the lumbar interneuronal networks and/or increases the overall excitability of the locomotor centers suggest that these centers are much more dependent on the supraspinal tonic drive in humans. Sensory feedback shapes the locomotor output in animal models but does not appear to be sufficient to drive it in humans.
Collapse
Affiliation(s)
- Marie-Pascale Côté
- 1 Department of Neurobiology and Anatomy, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Marion Murray
- 1 Department of Neurobiology and Anatomy, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Michel A Lemay
- 2 Department of Bioengineering, Temple University , Philadelphia, Pennsylvania
| |
Collapse
|
28
|
Leijon SC, Peyda S, Magnusson AK. Temporal processing capacity in auditory-deprived superior paraolivary neurons is rescued by sequential plasticity during early development. Neuroscience 2016; 337:315-330. [DOI: 10.1016/j.neuroscience.2016.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/01/2016] [Accepted: 09/09/2016] [Indexed: 01/04/2023]
|
29
|
Brocard C, Plantier V, Boulenguez P, Liabeuf S, Bouhadfane M, Viallat-Lieutaud A, Vinay L, Brocard F. Cleavage of Na+ channels by calpain increases persistent Na+ current and promotes spasticity after spinal cord injury. Nat Med 2016; 22:404-11. [DOI: 10.1038/nm.4061] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 02/08/2016] [Indexed: 12/13/2022]
|
30
|
Depolarizing GABA/glycine synaptic events switch from excitation to inhibition during frequency increases. Sci Rep 2016; 6:21753. [PMID: 26912194 PMCID: PMC4766471 DOI: 10.1038/srep21753] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/27/2016] [Indexed: 01/04/2023] Open
Abstract
By acting on their ionotropic chloride channel receptors, GABA and glycine represent the major inhibitory transmitters of the central nervous system. Nevertheless, in various brain structures, depolarizing GABAergic/glycinergic postsynaptic potentials (dGPSPs) lead to dual inhibitory (shunting) and excitatory components, the functional consequences of which remain poorly acknowledged. Indeed, the extent to which each component prevails during dGPSP is unclear. Understanding the mechanisms predicting the dGPSP outcome on neural network activity is therefore a major issue in neurobiology. By combining electrophysiological recordings of spinal embryonic mouse motoneurons and modelling study, we demonstrate that increasing the chloride conductance (gCl) favors inhibition either during a single dGPSP or during trains in which gCl summates. Finally, based on this summation mechanism, the excitatory effect of EPSPs is overcome by dGPSPs in a frequency-dependent manner. These results reveal an important mechanism by which dGPSPs protect against the overexcitation of neural excitatory circuits.
Collapse
|
31
|
Durand J, Filipchuk A, Pambo-Pambo A, Amendola J, Borisovna Kulagina I, Guéritaud JP. Developing electrical properties of postnatal mouse lumbar motoneurons. Front Cell Neurosci 2015; 9:349. [PMID: 26388736 PMCID: PMC4557103 DOI: 10.3389/fncel.2015.00349] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/20/2015] [Indexed: 11/13/2022] Open
Abstract
We studied the rapid changes in electrical properties of lumbar motoneurons between postnatal days 3 and 9 just before mice weight-bear and walk. The input conductance and rheobase significantly increased up to P8. A negative correlation exists between the input resistance (Rin) and rheobase. Both parameters are significantly correlated with the total dendritic surface area of motoneurons, the largest motoneurons having the lowest Rin and the highest rheobase. We classified the motoneurons into three groups according to their discharge firing patterns during current pulse injection (transient, delayed onset, sustained). The delayed onset firing type has the highest rheobase and the fastest action potential (AP) whereas the transient firing group has the lowest rheobase and the less mature AP. We found 32 and 10% of motoneurons with a transient firing at P3-P5 and P8, respectively. About 20% of motoneurons with delayed onset firing were detected at P8. At P9, all motoneurons exhibit a sustained firing. We defined five groups of motoneurons according to their discharge firing patterns in response to ascending and descending current ramps. In addition to the four classical types, we defined a fifth type called transient for the quasi-absence of discharge during the descending phase of the ramp. This transient type represents about 40% between P3-P5 and tends to disappear with age. Types 1 and 2 (linear and clockwise hysteresis) are the most preponderant at P6-P7. Types 3 and 4 (prolonged sustained and counter clockwise hysteresis) emerge at P8-P9. The emergence of types 3 and 4 probably depends on the maturation of L type calcium channels in the dendrites of motoneurons. No correlation was found between groups defined by step or triangular ramp of currents with the exception of transient firing patterns. Our data support the idea that a switch in the electrical properties of lumbar motoneurons might exist in the second postnatal week of life in mice.
Collapse
Affiliation(s)
- Jacques Durand
- Institut de Neurosciences de la Timone, Aix Marseille Université - CNRS, UMR 7289 Marseille, France
| | - Anton Filipchuk
- Institut de Neurosciences de la Timone, Aix Marseille Université - CNRS, UMR 7289 Marseille, France
| | - Arnaud Pambo-Pambo
- Institut de Neurosciences de la Timone, Aix Marseille Université - CNRS, UMR 7289 Marseille, France
| | - Julien Amendola
- Institut de Neurosciences de la Timone, Aix Marseille Université - CNRS, UMR 7289 Marseille, France
| | | | - Jean-Patrick Guéritaud
- Institut de Neurosciences de la Timone, Aix Marseille Université - CNRS, UMR 7289 Marseille, France
| |
Collapse
|
32
|
Kurowski P, Gawlak M, Szulczyk P. Muscarinic receptor control of pyramidal neuron membrane potential in the medial prefrontal cortex (mPFC) in rats. Neuroscience 2015; 303:474-88. [PMID: 26186898 DOI: 10.1016/j.neuroscience.2015.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/08/2015] [Indexed: 11/19/2022]
Abstract
Damage to the cholinergic input to the prefrontal cortex has been implicated in neuropsychiatric disorders. Cholinergic endings release acetylcholine, which activates nicotinic and/or G-protein-coupled muscarinic receptors. Muscarinic receptors activate transduction systems, which control cellular effectors that regulate the membrane potential in medial prefrontal cortex (mPFC) neurons. The mechanisms responsible for the cholinergic-dependent depolarization of mPFC layer V pyramidal neurons in slices obtained from young rats were elucidated in this study. Glutamatergic and GABAergic transmission as well as tetrodotoxin (TTX)-sensitive Na(+) and voltage-dependent Ca(++) currents were eliminated. Cholinergic receptor stimulation by carbamoylcholine chloride (CCh; 100 μM) evoked depolarization (10.0 ± 1.3 mV), which was blocked by M1/M4 (pirenzepine dihydrochloride, 2 μM) and M1 (VU 0255035, 5 μM) muscarinic receptor antagonists and was not affected by a nicotinic receptor antagonist (mecamylamine hydrochloride, 10 μM). CCh-dependent depolarization was attenuated by extra- (20 μM) or intracellular (50 μM) application of an inhibitor of the βγ-subunit-dependent transduction system (gallein). It was also inhibited by intracellular application of a βγ-subunit-binding peptide (GRK2i, 10μM). mPFC pyramidal neurons express Nav1.9 channels. CCh-dependent depolarization was abolished in the presence of antibodies against Nav1.9 channels in the intracellular solution and augmented by the presence of ProTx-I toxin (100 nM) in the extracellular solution. CCh-induced depolarization was not affected by the following reagents: intracellular transduction system blockers, including U-73122 (10 μM), chelerythrine chloride (5 μM), SQ 22536 (100 μM) and H-89 (2 μM); channel blockers, including Ba(++) ions (200 μM), apamin (100 nM), flufenamic acid (200 μM), 2-APB (200 μM), SKF 96365 (50 μM), and ZD 7288 (50 μM); and a Na(+)/Ca(++) exchanger blocker, benzamil (20 μM). We conclude that muscarinic M1 receptor-dependent depolarization in mPFC pyramidal neurons is evoked by the activation of Nav1.9 channels and that the signal transduction pathway involves G-protein βγ subunits.
Collapse
Affiliation(s)
- P Kurowski
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Banacha 1B, Warsaw 02-097, Poland
| | - M Gawlak
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Banacha 1B, Warsaw 02-097, Poland
| | - P Szulczyk
- Laboratory of Physiology and Pathophysiology, Centre for Preclinical Research and Technology, The Medical University of Warsaw, Banacha 1B, Warsaw 02-097, Poland.
| |
Collapse
|
33
|
Bouhadfane M, Kaszás A, Rózsa B, Harris-Warrick RM, Vinay L, Brocard F. Sensitization of neonatal rat lumbar motoneuron by the inflammatory pain mediator bradykinin. eLife 2015; 4:e06195. [PMID: 25781633 PMCID: PMC4410746 DOI: 10.7554/elife.06195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 03/16/2015] [Indexed: 11/27/2022] Open
Abstract
Bradykinin (Bk) is a potent inflammatory mediator that causes hyperalgesia. The action of Bk on the sensory system is well documented but its effects on motoneurons, the final pathway of the motor system, are unknown. By a combination of patch-clamp recordings and two-photon calcium imaging, we found that Bk strongly sensitizes spinal motoneurons. Sensitization was characterized by an increased ability to generate self-sustained spiking in response to excitatory inputs. Our pharmacological study described a dual ionic mechanism to sensitize motoneurons, including inhibition of a barium-sensitive resting K+ conductance and activation of a nonselective cationic conductance primarily mediated by Na+. Examination of the upstream signaling pathways provided evidence for postsynaptic activation of B2 receptors, G protein activation of phospholipase C, InsP3 synthesis, and calmodulin activation. This study questions the influence of motoneurons in the assessment of hyperalgesia since the withdrawal motor reflex is commonly used as a surrogate pain model. DOI:http://dx.doi.org/10.7554/eLife.06195.001 When we accidentally place our hand on a hot stove, we normally experience a painful sensation that starts with the sensory nerves under our skin. These nerves respond by transmitting electrical impulses to our brain, where the painful sensation is then processed. At the same time, these impulses are also transmitted to the motor nerves that control the muscles in our hand to trigger an immediate reflex to withdraw the hand from the hot stove. Pain therefore has a useful role as it can reduce how bad an injury is. People with a condition called hyperalgesia have an increased sensitivity to pain. This condition can result from a chemical called bradykinin ‘sensitizing’ the sensory nerves, causing them to transmit more electrical impulses in response to pain than normal. This makes the injury feel much more painful, and can make the pain last for longer than is beneficial. It was less clear whether bradykinin also affects motor nerves and so triggers a withdrawal reflex. By recording the electrical activity of motor nerve cells taken from the spinal cords of newborn rats, Bouhadfane et al. now show that these motor nerves become more active when exposed to bradykinin. Nerve cells generate electrical signals when ions—such as potassium, sodium, and calcium ions—move through channels in the membranes of the cell. Therefore, to investigate how bradykinin influences the electrical activity of motor nerves, Bouhadfane et al. exposed the cells to drugs that inhibit particular ion channels. This revealed that bradykinin sensitizes the motor nerves by blocking a type of potassium ion channel and activating another ion channel that mainly transports sodium ions. Furthermore, Bouhadfane et al. were able to identify the signaling pathways that allow bradykinin to affect the motor nerve cells. The study implies that the neuronal circuitry for pain does not rely exclusively on sensory nerve cells but should also integrate motor nerve cells. A future challenge remains in developing a protocol to resolve the contribution of motor nerve cells to hyperalgesia assessed by reflex withdrawal. DOI:http://dx.doi.org/10.7554/eLife.06195.002
Collapse
Affiliation(s)
- Mouloud Bouhadfane
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Attila Kaszás
- Institut de Neuroscience des Systèmes (UMR1106), Aix Marseille Université and INSERM, Marseille, France
| | - Balázs Rózsa
- Two-Photon Imaging Center, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Laurent Vinay
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| | - Frédéric Brocard
- Institut de Neurosciences de la Timone (UMR7289), Aix-Marseille Université and CNRS, Marseille, France
| |
Collapse
|
34
|
Husch A, Dietz SB, Hong DN, Harris-Warrick RM. Adult spinal V2a interneurons show increased excitability and serotonin-dependent bistability. J Neurophysiol 2014; 113:1124-34. [PMID: 25520435 DOI: 10.1152/jn.00741.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In mice, most studies of the organization of the spinal central pattern generator (CPG) for locomotion, and its component neuron classes, have been performed on neonatal [postnatal day (P)2-P4] animals. While the neonatal spinal cord can generate a basic locomotor pattern, it is often argued that the CPG network is in an immature form whose detailed properties mature with postnatal development. Here, we compare intrinsic properties and serotonergic modulation of the V2a class of excitatory spinal interneurons in behaviorally mature (older than P43) mice to those in neonatal mice. Using perforated patch recordings from genetically tagged V2a interneurons, we revealed an age-dependent increase in excitability. The input resistance increased, the rheobase values decreased, and the relation between injected current and firing frequency (F/I plot) showed higher excitability in the adult neurons, with almost all neurons firing tonically during a current step. The adult action potential (AP) properties became narrower and taller, and the AP threshold hyperpolarized. While in neonates the AP afterhyperpolarization was monophasic, most adult V2a interneurons showed a biphasic afterhyperpolarization. Serotonin increased excitability and depolarized most neonatal and adult V2a interneurons. However, in ∼30% of adult V2a interneurons, serotonin additionally elicited spontaneous intrinsic membrane potential bistability, resulting in alternations between hyperpolarized and depolarized states with a dramatically decreased membrane input resistance and facilitation of evoked plateau potentials. This was never seen in younger animals. Our findings indicate a significant postnatal development of the properties of locomotor-related V2a interneurons, which could alter their interpretation of synaptic inputs in the locomotor CPG.
Collapse
Affiliation(s)
- Andreas Husch
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York
| | - Shelby B Dietz
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York
| | - Diana N Hong
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York
| | | |
Collapse
|
35
|
Yan Z, Xu Y, Liang M, Ren X. Expression and functional role of Nav1.9 sodium channel in cartwheel cells of the dorsal cochlear nucleus. Mol Med Rep 2014; 11:1833-6. [PMID: 25385328 DOI: 10.3892/mmr.2014.2922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 06/17/2014] [Indexed: 11/06/2022] Open
Abstract
In the central auditory system, cartwheel cells (CWCs) are a group of interneurons in the dorsal cochlear nucleus (DCN). While other DCN neurons respond to stimuli with a simple discharge pattern of single action potentials (SAPs), CWCs respond with complex action potentials (CAPs), consisting of SAPs superimposed on a slow depolarization. The CAPs in CWCs may participate in various auditory or non‑auditory signaling processing but its intrinsic mechanisms are largely unknown. In the present study, in vitro whole‑cell current clamp recordings on neonatal mice brain slices were used to demonstrate that CWCs respond to brief voltage stimulation with CAPs. Western blotting and immunohistochemistry were also utilized to demonstrate that Nav1.9 was expressed in the CWCs. Finally, when Nav1.9 was genetically silenced, CWCs responded to voltage stimulation with SAPs, not CAPs. The results strongly suggested that Nav1.9 was expressed and functionally contributed to the signaling processing in the central auditory pathway.
Collapse
Affiliation(s)
- Zhiyu Yan
- Depatment of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| | - Yanjun Xu
- Depatment of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| | - Min Liang
- Depatment of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| | - Xiaowei Ren
- Depatment of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| |
Collapse
|
36
|
Alvares TS, Revill AL, Huxtable AG, Lorenz CD, Funk GD. P2Y1 receptor-mediated potentiation of inspiratory motor output in neonatal rat in vitro. J Physiol 2014; 592:3089-111. [PMID: 24879869 DOI: 10.1113/jphysiol.2013.268136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
PreBötzinger complex inspiratory rhythm-generating networks are excited by metabotropic purinergic receptor subtype 1 (P2Y1R) activation. Despite this, and the fact that inspiratory MNs express P2Y1Rs, the role of P2Y1Rs in modulating motor output is not known for any MN pool. We used rhythmically active brainstem-spinal cord and medullary slice preparations from neonatal rats to investigate the effects of P2Y1R signalling on inspiratory output of phrenic and XII MNs that innervate diaphragm and airway muscles, respectively. MRS2365 (P2Y1R agonist, 0.1 mm) potentiated XII inspiratory burst amplitude by 60 ± 9%; 10-fold higher concentrations potentiated C4 burst amplitude by 25 ± 7%. In whole-cell voltage-clamped XII MNs, MRS2365 evoked small inward currents and potentiated spontaneous EPSCs and inspiratory synaptic currents, but these effects were absent in TTX at resting membrane potential. Voltage ramps revealed a persistent inward current (PIC) that was attenuated by: flufenamic acid (FFA), a blocker of the Ca(2+)-dependent non-selective cation current ICAN; high intracellular concentrations of BAPTA, which buffers Ca(2+) increases necessary for activation of ICAN; and 9-phenanthrol, a selective blocker of TRPM4 channels (candidate for ICAN). Real-time PCR analysis of mRNA extracted from XII punches and laser-microdissected XII MNs revealed the transcript for TRPM4. MRS2365 potentiated the PIC and this potentiation was blocked by FFA, which also blocked the MRS2365 potentiation of glutamate currents. These data suggest that XII MNs are more sensitive to P2Y1R modulation than phrenic MNs and that the P2Y1R potentiation of inspiratory output occurs in part via potentiation of TRPM4-mediated ICAN, which amplifies inspiratory inputs.
Collapse
Affiliation(s)
- T S Alvares
- Department of Physiology, Centre for Neuroscience, Women and Children's Health Research Institute (WCHRI), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - A L Revill
- Department of Physiology, Centre for Neuroscience, Women and Children's Health Research Institute (WCHRI), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - A G Huxtable
- Department of Physiology, Centre for Neuroscience, Women and Children's Health Research Institute (WCHRI), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - C D Lorenz
- Department of Physiology, Centre for Neuroscience, Women and Children's Health Research Institute (WCHRI), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - G D Funk
- Department of Physiology, Centre for Neuroscience, Women and Children's Health Research Institute (WCHRI), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
37
|
Hasreiter J, Goldnagl L, Böhm S, Kubista H. Cav1.2 and Cav1.3 L-type calcium channels operate in a similar voltage range but show different coupling to Ca(2+)-dependent conductances in hippocampal neurons. Am J Physiol Cell Physiol 2014; 306:C1200-13. [PMID: 24760982 DOI: 10.1152/ajpcell.00329.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the central nervous system, L-type voltage-gated calcium channels (LTCCs) come in two isoforms, namely Cav1.2 and Cav1.3 channels. It has been shown previously that these channels differ in biophysical properties, in subcellular localization, and in the coupling to the gene transcription machinery. In previous work on rat hippocampal neurons we have identified an excitatory cation conductance and an inhibitory potassium conductance as important LTCC coupling partners. Notably, a stimulus-dependent interplay of LTCC-mediated Ca(2+) influx and activation of these Ca(2+)-dependent conductances was found to give rise to characteristic voltage responses. However, the contribution of Cav1.2 and Cav1.3 to these voltage responses remained unknown. Hence, the relative contribution of the LTCC isoforms therein was the focus of the current study on hippocampal neurons derived from genetically modified mice, which either lack a LTCC isoform (Cav1.3 knockout mice) or express a dihydropyridine-insensitive LTCC isoform (Cav1.2DHP(-)-knockin mice). We identified common and alternate ion channel couplings of Cav1.2 and Cav1.3 channels. Whereas hyperpolarizing Ca(2+)-dependent conductances were coupled to both Cav1.2 and Cav1.3 channels, an afterdepolarizing potential was only induced by the activity of Cav1.2 channels. Unexpectedly, the activity of Cav1.2 channels was found at relatively hyperpolarized membrane voltages. Our data add important information about the differences between Cav1.2 and Cav1.3 channels that furthers our understanding of the physiological and pathophysiological neuronal roles of these calcium channels. Moreover, our findings suggest that Cav1.3 knockout mice together with Cav1.2DHP(-)-knockin mice provide valuable models for future investigation of hippocampal LTCC-dependent afterdepolarizations.
Collapse
Affiliation(s)
- Julia Hasreiter
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lena Goldnagl
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Böhm
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Center of Physiology and Pharmacology, Department of Neurophysiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Delestrée N, Manuel M, Iglesias C, Elbasiouny SM, Heckman CJ, Zytnicki D. Adult spinal motoneurones are not hyperexcitable in a mouse model of inherited amyotrophic lateral sclerosis. J Physiol 2014; 592:1687-703. [PMID: 24445319 DOI: 10.1113/jphysiol.2013.265843] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In amyotrophic lateral sclerosis (ALS), an adult onset disease in which there is progressive degeneration of motoneurones, it has been suggested that an intrinsic hyperexcitability of motoneurones (i.e. an increase in their firing rates), contributes to excitotoxicity and to disease onset. Here we show that there is no such intrinsic hyperexcitability in spinal motoneurones. Our studies were carried out in an adult mouse model of ALS with a mutated form of superoxide dismutase 1 around the time of the first muscle fibre denervations. We showed that the recruitment current, the voltage threshold for spiking and the frequency-intensity gain in the primary range are all unchanged in most spinal motoneurones, despite an increased input conductance. On its own, increased input conductance would decrease excitability, but the homeostasis for excitability is maintained due to an upregulation of a depolarizing current that is activated just below the spiking threshold. However, this homeostasis failed in a substantial fraction of motoneurones, which became hypoexcitable and unable to produce sustained firing in response to ramps of current. We found similar results both in lumbar motoneurones recorded in anaesthetized mice, and in sacrocaudal motoneurones recorded in vitro, indicating that the lack of hyperexcitability is not caused by anaesthetics. Our results suggest that, if excitotoxicity is indeed a mechanism leading to degeneration in ALS, it is not caused by the intrinsic electrical properties of motoneurones but by extrinsic factors such as excessive synaptic excitation.
Collapse
Affiliation(s)
- Nicolas Delestrée
- Laboratoire de Neurophysique et Physiologie, UMR CNRS 8119, Université Paris Descartes, 45 rue des Saints-Pères, 75006 Paris, France.
| | | | | | | | | | | |
Collapse
|