1
|
Ransdell JL, Carrasquillo Y, Bosch MK, Mellor RL, Ornitz DM, Nerbonne JM. Loss of intracellular FGF14 (iFGF14) increases excitability of mature hippocampal pyramidal neurons. J Gen Physiol 2025; 157:e202413597. [PMID: 40323232 PMCID: PMC12051480 DOI: 10.1085/jgp.202413597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 01/30/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Mutations in FGF14, which encodes intracellular fibroblast growth factor 14 (iFGF14), have been linked to spinocerebellar ataxia type 27 (SCA27), a multisystem disorder associated with deficits in motor coordination and cognitive function. Mice lacking iFGF14 (Fgf14-/-) display similar phenotypes, and we have previously shown that the deficits in motor coordination reflect reduced excitability of cerebellar Purkinje neurons, owing to a hyperpolarizing shift in the voltage-dependence of voltage-gated Na+ (Nav) current steady-state inactivation. Here, we present the results of experiments designed to test the hypothesis that loss of iFGF14 also attenuates the intrinsic excitability of mature hippocampal pyramidal neurons. Current-clamp recordings from CA1 pyramidal neurons in acute in vitro slices, however, revealed that evoked repetitive firing rates were higher in Fgf14-/- than in wild type (WT) cells. Also, in contrast with Purkinje neurons, voltage-clamp recordings demonstrated that the loss of iFGF14 did not affect the voltage dependence of steady-state inactivation of the Nav currents in CA1 pyramidal neurons. In addition, in contrast with results reported for neonatal (rat) hippocampal pyramidal neurons in dissociated cell culture, immunohistochemical experiments revealed that loss of iFGF14 does not disrupt the localization or alter the normalized distribution of α-Nav1.6 or α-ankyrin G labeling along the axon initial segments (AIS) of mature hippocampal CA1 neurons in situ. However, the integrated intensities of α-Nav1.6 labeling were significantly higher along the AIS of Fgf14-/-, compared with WT, adult hippocampal CA1 pyramidal neurons, consistent with the marked increase in the excitability of CA1 neurons with the loss of iFGF14.
Collapse
Affiliation(s)
- Joseph L. Ransdell
- Cardiovascular Division, Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yarimar Carrasquillo
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marie K. Bosch
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca L. Mellor
- Cardiovascular Division, Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - David M. Ornitz
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeanne M. Nerbonne
- Cardiovascular Division, Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
2
|
Brown SP, Jena AK, Osko JJ, Ransdell JL. Tsc1 deletion in Purkinje neurons disrupts the axon initial segment, impairing excitability and cerebellar function. Neurobiol Dis 2025; 207:106856. [PMID: 40015654 PMCID: PMC11997981 DOI: 10.1016/j.nbd.2025.106856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
Loss-of-function mutations in tuberous sclerosis 1 (TSC1) are prevalent monogenic causes of autism spectrum disorder (ASD). Selective deletion of Tsc1 from mouse cerebellar Purkinje neurons has been shown to cause several ASD-linked behavioral impairments, which are linked to reduced Purkinje neuron repetitive firing rates. We used electrophysiology methods to investigate why Purkinje neuron-specific Tsc1 deletion (Tsc1mut/mut) impairs Purkinje neuron firing. These studies revealed a depolarized shift in action potential threshold voltage, an effect that we link to reduced expression of the fast-transient voltage-gated sodium (Nav) current in Tsc1mut/mut Purkinje neurons. The reduced Nav currents in these cells was associated with diminished secondary immunofluorescence from anti-pan Nav channel labeling at Purkinje neuron axon initial segments (AIS). Anti-ankyrinG immunofluorescence was also found to be significantly reduced at the AIS of Tsc1mut/mut Purkinje neurons, suggesting Tsc1 is necessary for the organization and functioning of the Purkinje neuron AIS. An analysis of the 1st and 2nd derivative of the action potential voltage-waveform supported this hypothesis, revealing spike initiation and propagation from the AIS of Tsc1mut/mut Purkinje neurons is impaired compared to age-matched control Purkinje neurons. Heterozygous Tsc1 deletion resulted in no significant changes in the firing properties of adult Purkinje neurons, and slight reductions in anti-pan Nav and anti-ankyrinG labeling at the Purkinje neuron AIS, revealing deficits in Purkinje neuron firing due to Tsc1 haploinsufficiency are delayed compared to age-matched Tsc1mut/mut Purkinje neurons. Together, these data reveal that the loss of Tsc1 impairs Purkinje neuron firing and membrane excitability through the dysregulation of proteins essential for AIS organization and function.
Collapse
Affiliation(s)
- Samuel P Brown
- Department of Biology, Miami University, Oxford, OH 45056, United States
| | - Achintya K Jena
- Department of Biology, Miami University, Oxford, OH 45056, United States
| | - Joanna J Osko
- Department of Biology, Miami University, Oxford, OH 45056, United States
| | - Joseph L Ransdell
- Department of Biology, Miami University, Oxford, OH 45056, United States.
| |
Collapse
|
3
|
Wood DJ, Tsvetkov E, Comte-Walters S, Welsh CL, Bloyd M, Wood TG, Akiki RM, Anderson EM, Penrod RD, Madan LK, Ball LE, Taniguchi M, Cowan CW. Epigenetic Control of an Auxiliary Subunit of Voltage-Gated Sodium Channels Regulates the Strength of Drug-Cue Associations and Relapse-Like Cocaine Seeking. Biol Psychiatry 2025:S0006-3223(25)00075-7. [PMID: 39923817 DOI: 10.1016/j.biopsych.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Repeated use of addictive drugs produces long-lasting and prepotent drug-cue associations that increase vulnerability for relapse in individuals with a substance use disorder. Epigenetic factors, such as HDAC5 (histone deacetylase 5), play a key role in regulating the formation of drug-cue associations, but the underlying mechanisms remain unclear. METHODS We used a combination of molecular biology, cultured cells, tandem mass spectrometry, deacetylase activity measurements, co-immunoprecipitation, and molecular dynamics simulations to assess HDAC5 structure-activity relationships. In male and female Long Evans rats, we used viral-mediated expression of HDAC5 mutants in the nucleus accumbens (NAc) to test effects on cocaine intravenous self-administration and cue-reinstated cocaine seeking. We also used in silico analysis of single-nucleus RNA sequencing data, quantitative reverse transcriptase-polymerase chain reaction, viral-mediated expression of Scn4b short hairpin RNA, patch-clamp electrophysiology, and rat cocaine or sucrose SA to assess Scn4b's effects on NAc intrinsic excitability and cued reward seeking. RESULTS We discovered that 2 conserved cysteines located near HDAC5's catalytic domain were required for its intrinsic deacetylase activity and that HDAC5's deacetylase activity was required in NAc medium spiny neurons (MSNs) to limit relapse-like cue-reinstated cocaine seeking. Moreover, we found that HDAC5 limited cocaine-seeking, but not sucrose-seeking, behavior by reducing NAc MSN intrinsic excitability through the deacetylase-dependent repression of Scn4b, which codes for an auxiliary subunit of voltage-gated sodium channels. CONCLUSIONS Our findings suggest that HDAC5's control of NAc Scn4b expression governs the formation of cocaine-cue, but not sucrose-cue, associations through modulation of NAc MSN intrinsic excitability and drug-induced NAc plasticity mechanisms.
Collapse
Affiliation(s)
- Daniel J Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Susana Comte-Walters
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Colin L Welsh
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Michelle Bloyd
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina
| | - Timothy G Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina
| | - Ethan M Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Rachel D Penrod
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Lalima K Madan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren E Ball
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
4
|
Ransdell JL, Brown SP, Xiao M, Ornitz DM, Nerbonne JM. In Vivo Expression of an SCA27A-linked FGF14 Mutation Results in Haploinsufficiency and Impaired Firing of Cerebellar Purkinje Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620253. [PMID: 39484407 PMCID: PMC11527103 DOI: 10.1101/2024.10.25.620253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Autosomal dominant mutations in FGF14 , which encodes intracellular fibroblast growth factor 14 (iFGF14), underlie spinocerebellar ataxia type 27A (SCA27A), a devastating multisystem disorder resulting in progressive deficits in motor coordination and cognitive function. Mice lacking iFGF14 ( Fgf14 -/- ) exhibit similar phenotypes, which have been linked to iFGF14-mediated modulation of the voltage-gated sodium (Nav) channels that control the high frequency repetitive firing of Purkinje neurons, the main output neurons of the cerebellar cortex. To investigate the pathophysiological mechanisms underlying SCA27A, we developed a targeted knock-in strategy to introduce the first point mutation identified in FGF14 into the mouse Fgf14 locus ( Fgf14 F145S ), we determined the impact of in vivo expression of the mutant Fgf14 F145S allele on the motor performance of adult animals and on the firing properties of mature Purkinje neurons in acute cerebellar slices. Electrophysiological experiments revealed that repetitive firing rates are attenuated in adult Fgf14 F145S/+ cerebellar Purkinje neurons, attributed to a hyperpolarizing shift in the voltage-dependence of steady-state inactivation of Nav channels. More severe effects on firing properties and Nav channel inactivation were observed in homozygous Fgf14 F145S/F145S Purkinje neurons. Interestingly, the electrophysiological phenotypes identified in adult Fgf14 F145S/+ and Fgf14 F145S/F145S cerebellar Purkinje neurons mirror those observed in heterozygous Fgf14 +/- and homozygous Fgf14 -/- Purkinje neurons, respectively, suggesting that the mutation results in the loss of the iFGF14 protein. Western blot analysis of lysates from adult heterozygous Fgf14 F145S/+ and homozygous Fgf14 F145S/F145S animals revealed reduced or undetectable, respectively, iFGF14 expression, supporting the hypothesis that the mutant allele results in loss of the iFGF14 protein and that haploinsufficiency underlies SCA27A neurological phenotypes.
Collapse
|
5
|
Brown SP, Lawson RJ, Moreno JD, Ransdell JL. A Reinterpretation of the Relationship Between Persistent and Resurgent Sodium Currents. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.25.564042. [PMID: 38187680 PMCID: PMC10769191 DOI: 10.1101/2023.10.25.564042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The resurgent sodium current (INaR) activates on membrane repolarization, such as during the downstroke of neuronal action potentials. Due to its unique activation properties, INaR is thought to drive high rates of repetitive neuronal firing. However, INaR is often studied in combination with the persistent or non-inactivating portion of sodium currents (INaP). We used dynamic clamp to test how INaR and INaP individually affect repetitive firing in adult cerebellar Purkinje neurons from male and female mice. We learned INaR does not scale repetitive firing rates due to its rapid decay at subthreshold voltages, and that subthreshold INaP is critical in regulating neuronal firing rate. Adjustments to the Nav conductance model used in these studies revealed INaP and INaR can be inversely scaled by adjusting occupancy in the slow inactivated kinetic state. Together with additional dynamic clamp experiments, these data suggest the regulation of sodium channel slow inactivation can fine-tune INaP and Purkinje neuron repetitive firing rates.
Collapse
Affiliation(s)
| | - Ryan J. Lawson
- Department of Biology, Miami University, Oxford, OH 45056
| | - Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130
| | | |
Collapse
|
6
|
Sekerková G, Kilic S, Cheng YH, Fredrick N, Osmani A, Kim H, Opal P, Martina M. Phenotypical, genotypical and pathological characterization of the moonwalker mouse, a model of ataxia. Neurobiol Dis 2024; 195:106492. [PMID: 38575093 PMCID: PMC11089908 DOI: 10.1016/j.nbd.2024.106492] [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: 11/01/2023] [Revised: 03/13/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024] Open
Abstract
We performed a comprehensive study of the morphological, functional, and genetic features of moonwalker (MWK) mice, a mouse model of spinocerebellar ataxia caused by a gain of function of the TRPC3 channel. These mice show numerous behavioral symptoms including tremor, altered gait, circling behavior, impaired motor coordination, impaired motor learning and decreased limb strength. Cerebellar pathology is characterized by early and almost complete loss of unipolar brush cells as well as slowly progressive, moderate loss of Purkinje cell (PCs). Structural damage also includes loss of synaptic contacts from parallel fibers, swollen ER structures, and degenerating axons. Interestingly, no obvious correlation was observed between PC loss and severity of the symptoms, as the phenotype stabilizes around 2 months of age, while the cerebellar pathology is progressive. This is probably due to the fact that PC function is severely impaired much earlier than the appearance of PC loss. Indeed, PC firing is already impaired in 3 weeks old mice. An interesting feature of the MWK pathology that still remains to be explained consists in a strong lobule selectivity of the PC loss, which is puzzling considering that TRPC is expressed in every PC. Intriguingly, genetic analysis of MWK cerebella shows, among other alterations, changes in the expression of both apoptosis inducing and resistance factors possibly suggesting that damaged PCs initiate specific cellular pathways that protect them from overt cell loss.
Collapse
Affiliation(s)
- Gabriella Sekerková
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA.
| | - Sumeyra Kilic
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Yen-Hsin Cheng
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Natalie Fredrick
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Anne Osmani
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Haram Kim
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Puneet Opal
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA
| | - Marco Martina
- Department of Neuroscience, Northwestern University, Feinberg School of Medicine, 300 E. Superior, Chicago, IL 60611, USA.
| |
Collapse
|
7
|
Niewiadomska-Cimicka A, Fievet L, Surdyka M, Jesion E, Keime C, Singer E, Eisenmann A, Kalinowska-Poska Z, Nguyen HHP, Fiszer A, Figiel M, Trottier Y. AAV-Mediated CAG-Targeting Selectively Reduces Polyglutamine-Expanded Protein and Attenuates Disease Phenotypes in a Spinocerebellar Ataxia Mouse Model. Int J Mol Sci 2024; 25:4354. [PMID: 38673939 PMCID: PMC11050704 DOI: 10.3390/ijms25084354] [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: 03/26/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Polyglutamine (polyQ)-encoding CAG repeat expansions represent a common disease-causing mutation responsible for several dominant spinocerebellar ataxias (SCAs). PolyQ-expanded SCA proteins are toxic for cerebellar neurons, with Purkinje cells (PCs) being the most vulnerable. RNA interference (RNAi) reagents targeting transcripts with expanded CAG reduce the level of various mutant SCA proteins in an allele-selective manner in vitro and represent promising universal tools for treating multiple CAG/polyQ SCAs. However, it remains unclear whether the therapeutic targeting of CAG expansion can be achieved in vivo and if it can ameliorate cerebellar functions. Here, using a mouse model of SCA7 expressing a mutant Atxn7 allele with 140 CAGs, we examined the efficacy of short hairpin RNAs (shRNAs) targeting CAG repeats expressed from PHP.eB adeno-associated virus vectors (AAVs), which were introduced into the brain via intravascular injection. We demonstrated that shRNAs carrying various mismatches with the CAG target sequence reduced the level of polyQ-expanded ATXN7 in the cerebellum, albeit with varying degrees of allele selectivity and safety profile. An shRNA named A4 potently reduced the level of polyQ-expanded ATXN7, with no effect on normal ATXN7 levels and no adverse side effects. Furthermore, A4 shRNA treatment improved a range of motor and behavioral parameters 23 weeks after AAV injection and attenuated the disease burden of PCs by preventing the downregulation of several PC-type-specific genes. Our results show the feasibility of the selective targeting of CAG expansion in the cerebellum using a blood-brain barrier-permeable vector to attenuate the disease phenotype in an SCA mouse model. Our study represents a significant advancement in developing CAG-targeting strategies as a potential therapy for SCA7 and possibly other CAG/polyQ SCAs.
Collapse
Affiliation(s)
- Anna Niewiadomska-Cimicka
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France; (L.F.); (C.K.); (A.E.)
| | - Lorraine Fievet
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France; (L.F.); (C.K.); (A.E.)
| | - Magdalena Surdyka
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland; (M.S.); (E.J.); (Z.K.-P.); (M.F.)
| | - Ewelina Jesion
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland; (M.S.); (E.J.); (Z.K.-P.); (M.F.)
| | - Céline Keime
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France; (L.F.); (C.K.); (A.E.)
| | - Elisabeth Singer
- Centre for Rare Diseases (ZSE), University of Tuebingen, 72076 Tuebingen, Germany;
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, 72076 Tuebingen, Germany
- Department of Human Genetics, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany;
| | - Aurélie Eisenmann
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France; (L.F.); (C.K.); (A.E.)
| | - Zaneta Kalinowska-Poska
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland; (M.S.); (E.J.); (Z.K.-P.); (M.F.)
| | - Hoa Huu Phuc Nguyen
- Department of Human Genetics, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany;
| | - Agnieszka Fiszer
- Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland;
| | - Maciej Figiel
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland; (M.S.); (E.J.); (Z.K.-P.); (M.F.)
| | - Yvon Trottier
- Institute of Genetics and Molecular and Cellular Biology, INSERM U1258, CNRS UMR7104, University of Strasbourg, 67404 Illkirch, France; (L.F.); (C.K.); (A.E.)
| |
Collapse
|
8
|
Kelly JJ, Wen H, Brehm P. Single-cell RNAseq analysis of spinal locomotor circuitry in larval zebrafish. eLife 2023; 12:RP89338. [PMID: 37975797 PMCID: PMC10656102 DOI: 10.7554/elife.89338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Identification of the neuronal types that form the specialized circuits controlling distinct behaviors has benefited greatly from the simplicity offered by zebrafish. Electrophysiological studies have shown that in addition to connectivity, understanding of circuitry requires identification of functional specializations among individual circuit components, such as those that regulate levels of transmitter release and neuronal excitability. In this study, we use single-cell RNA sequencing (scRNAseq) to identify the molecular bases for functional distinctions between motoneuron types that are causal to their differential roles in swimming. The primary motoneuron, in particular, expresses high levels of a unique combination of voltage-dependent ion channel types and synaptic proteins termed functional 'cassettes.' The ion channel types are specialized for promoting high-frequency firing of action potentials and augmented transmitter release at the neuromuscular junction, both contributing to greater power generation. Our transcriptional profiling of spinal neurons further assigns expression of this cassette to specific interneuron types also involved in the central circuitry controlling high-speed swimming and escape behaviors. Our analysis highlights the utility of scRNAseq in functional characterization of neuronal circuitry, in addition to providing a gene expression resource for studying cell type diversity.
Collapse
Affiliation(s)
- Jimmy J Kelly
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Hua Wen
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Paul Brehm
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
9
|
Kelly JJ, Wen H, Brehm P. Single cell RNA-seq analysis of spinal locomotor circuitry in larval zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543939. [PMID: 37333232 PMCID: PMC10274715 DOI: 10.1101/2023.06.06.543939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Identification of the neuronal types that form the specialized circuits controlling distinct behaviors has benefited greatly from the simplicity offered by zebrafish. Electrophysiological studies have shown that additional to connectivity, understanding of circuitry requires identification of functional specializations among individual circuit components, such as those that regulate levels of transmitter release and neuronal excitability. In this study we use single cell RNA sequencing (scRNAseq) to identify the molecular bases for functional distinctions between motoneuron types that are causal to their differential roles in swimming. The primary motoneuron (PMn) in particular, expresses high levels of a unique combination of voltage-dependent ion channel types and synaptic proteins termed functional 'cassettes'. The ion channel types are specialized for promoting high frequency firing of action potentials and augmented transmitter release at the neuromuscular junction, both contributing to greater power generation. Our transcriptional profiling of spinal neurons further assigns expression of this cassette to specific interneuron types also involved in the central circuitry controlling high speed swimming and escape behaviors. Our analysis highlights the utility of scRNAseq in functional characterization of neuronal circuitry, in addition to providing a gene expression resource for studying cell type diversity.
Collapse
Affiliation(s)
- Jimmy J Kelly
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Hua Wen
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Paul Brehm
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
10
|
Jang DC, Chung G, Kim SK, Kim SJ. Dynamic alteration of intrinsic properties of the cerebellar Purkinje cell during the motor memory consolidation. Mol Brain 2023; 16:58. [PMID: 37430311 DOI: 10.1186/s13041-023-01043-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 06/10/2023] [Indexed: 07/12/2023] Open
Abstract
Intrinsic plasticity of the cerebellar Purkinje cell (PC) plays a critical role in motor memory consolidation. However, detailed changes in their intrinsic properties during memory consolidation are not well understood. Here, we report alterations in various properties involved in intrinsic excitability, such as the action potential (AP) threshold, AP width, afterhyperpolarization (AHP), and sag voltage, which are associated with the long-term depression of intrinsic excitability following the motor memory consolidation process. We analyzed data recorded from PCs before and 1, 4, and 24 h after cerebellum-dependent motor learning and found that these properties underwent dynamic changes during the consolidation process. We further analyzed data from PC-specific STIM1 knockout (STIM1PKO) mice, which show memory consolidation deficits, and derived intrinsic properties showing distinct change patterns compared with those of wild-type littermates. The levels of memory retention in the STIM1PKO mice were significantly different compared to wild-type mice between 1 and 4 h after training, and AP width, fast- and medium-AHP, and sag voltage showed different change patterns during this period. Our results provide information regarding alterations in intrinsic properties during a particular period that are critical for memory consolidation.
Collapse
Affiliation(s)
- Dong Cheol Jang
- Department of Physiology, Neuroscience Research Center, Wide River Institute of Immunology, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03087, Republic of Korea
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Department of East-West Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Sang Jeong Kim
- Department of Physiology, Neuroscience Research Center, Wide River Institute of Immunology, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03087, Republic of Korea.
| |
Collapse
|
11
|
Patel T, Hammelman J, Aziz S, Jang S, Closser M, Michaels TL, Blum JA, Gifford DK, Wichterle H. Transcriptional dynamics of murine motor neuron maturation in vivo and in vitro. Nat Commun 2022; 13:5427. [PMID: 36109497 PMCID: PMC9477853 DOI: 10.1038/s41467-022-33022-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/25/2022] [Indexed: 12/03/2022] Open
Abstract
Neurons born in the embryo can undergo a protracted period of maturation lasting well into postnatal life. How gene expression changes are regulated during maturation and whether they can be recapitulated in cultured neurons remains poorly understood. Here, we show that mouse motor neurons exhibit pervasive changes in gene expression and accessibility of associated regulatory regions from embryonic till juvenile age. While motifs of selector transcription factors, ISL1 and LHX3, are enriched in nascent regulatory regions, motifs of NFI factors, activity-dependent factors, and hormone receptors become more prominent in maturation-dependent enhancers. Notably, stem cell-derived motor neurons recapitulate ~40% of the maturation expression program in vitro, with neural activity playing only a modest role as a late-stage modulator. Thus, the genetic maturation program consists of a core hardwired subprogram that is correctly executed in vitro and an extrinsically-controlled subprogram that is dependent on the in vivo context of the maturing organism.
Collapse
Affiliation(s)
- Tulsi Patel
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Jennifer Hammelman
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
| | - Siaresh Aziz
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sumin Jang
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Michael Closser
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Theodore L Michaels
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jacob A Blum
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Gifford
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, 02139, USA
| | - Hynek Wichterle
- Departments of Pathology & Cell Biology, Neuroscience, and Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
12
|
Ransdell JL, Moreno JD, Bhagavan D, Silva JR, Nerbonne JM. Intrinsic mechanisms in the gating of resurgent Na + currents. eLife 2022; 11:e70173. [PMID: 35076394 PMCID: PMC8824471 DOI: 10.7554/elife.70173] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
The resurgent component of the voltage-gated sodium current (INaR) is a depolarizing conductance, revealed on membrane hyperpolarizations following brief depolarizing voltage steps, which has been shown to contribute to regulating the firing properties of numerous neuronal cell types throughout the central and peripheral nervous systems. Although mediated by the same voltage-gated sodium (Nav) channels that underlie the transient and persistent Nav current components, the gating mechanisms that contribute to the generation of INaR remain unclear. Here, we characterized Nav currents in mouse cerebellar Purkinje neurons, and used tailored voltage-clamp protocols to define how the voltage and the duration of the initial membrane depolarization affect the amplitudes and kinetics of INaR. Using the acquired voltage-clamp data, we developed a novel Markov kinetic state model with parallel (fast and slow) inactivation pathways and, we show that this model reproduces the properties of the resurgent, as well as the transient and persistent, Nav currents recorded in (mouse) cerebellar Purkinje neurons. Based on the acquired experimental data and the simulations, we propose that resurgent Na+ influx occurs as a result of fast inactivating Nav channels transitioning into an open/conducting state on membrane hyperpolarization, and that the decay of INaR reflects the slow accumulation of recovered/opened Nav channels into a second, alternative and more slowly populated, inactivated state. Additional simulations reveal that extrinsic factors that affect the kinetics of fast or slow Nav channel inactivation and/or impact the relative distribution of Nav channels in the fast- and slow-inactivated states, such as the accessory Navβ4 channel subunit, can modulate the amplitude of INaR.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Departments of Medicine, Cardiovascular Division, Washington UniversitySt. LouisUnited States
| | - Jonathan D Moreno
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Druv Bhagavan
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Jonathan R Silva
- Developmental Biomedical Engineering, Washington UniversitySt. LouisUnited States
| | - Jeanne M Nerbonne
- Departments of Medicine, Cardiovascular Division, Washington UniversitySt. LouisUnited States
- Developmental Biology, Washington UniversitySt. LouisUnited States
| |
Collapse
|
13
|
Le Cann K, Foerster A, Rösseler C, Erickson A, Hautvast P, Giesselmann S, Pensold D, Kurth I, Rothermel M, Mattis VB, Zimmer-Bensch G, von Hörsten S, Denecke B, Clarner T, Meents J, Lampert A. The difficulty to model Huntington's disease in vitro using striatal medium spiny neurons differentiated from human induced pluripotent stem cells. Sci Rep 2021; 11:6934. [PMID: 33767215 PMCID: PMC7994641 DOI: 10.1038/s41598-021-85656-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an expanded polyglutamine repeat in the huntingtin gene. The neuropathology of HD is characterized by the decline of a specific neuronal population within the brain, the striatal medium spiny neurons (MSNs). The origins of this extreme vulnerability remain unknown. Human induced pluripotent stem cell (hiPS cell)-derived MSNs represent a powerful tool to study this genetic disease. However, the differentiation protocols published so far show a high heterogeneity of neuronal populations in vitro. Here, we compared two previously published protocols to obtain hiPS cell-derived striatal neurons from both healthy donors and HD patients. Patch-clamp experiments, immunostaining and RT-qPCR were performed to characterize the neurons in culture. While the neurons were mature enough to fire action potentials, a majority failed to express markers typical for MSNs. Voltage-clamp experiments on voltage-gated sodium (Nav) channels revealed a large variability between the two differentiation protocols. Action potential analysis did not reveal changes induced by the HD mutation. This study attempts to demonstrate the current challenges in reproducing data of previously published differentiation protocols and in generating hiPS cell-derived striatal MSNs to model a genetic neurodegenerative disorder in vitro.
Collapse
Affiliation(s)
- Kim Le Cann
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Alec Foerster
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Corinna Rösseler
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Andelain Erickson
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Petra Hautvast
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany
| | | | - Daniel Pensold
- Institute of Biology II, Division of Functional Epigenetics in the Animal Model, RWTH Aachen University, 52074, Aachen, Germany
| | - Ingo Kurth
- Intitute of Human Genetic, RWTH Aachen University, 52074, Aachen, Germany
| | - Markus Rothermel
- Institute Für Biology II, Department Chemosensation, AG Neuromodulation, 52074, Aachen, Germany
| | - Virginia B Mattis
- Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Fujifilm Cellular Dynamics, Madison, WI, 53711, USA
| | - Geraldine Zimmer-Bensch
- Institute of Biology II, Division of Functional Epigenetics in the Animal Model, RWTH Aachen University, 52074, Aachen, Germany
| | - Stephan von Hörsten
- Intitute of Virology, Clinical and Molecular Virology, Animal Center of Preclinical Experiments (PETZ), 91054, Erlangen, Germany
| | | | - Tim Clarner
- Intitute for Neuroanatomy, MIT 1, 52074, Aachen, Germany
| | - Jannis Meents
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Multi Channel Systems MCS GmbH, Aspenhaustrasse 21, 72770, Reutlingen, Germany.
| | - Angelika Lampert
- Institute of Physiology, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
14
|
Canet-Pons J, Sen NE, Arsović A, Almaguer-Mederos LE, Halbach MV, Key J, Döring C, Kerksiek A, Picchiarelli G, Cassel R, René F, Dieterlé S, Fuchs NV, König R, Dupuis L, Lütjohann D, Gispert S, Auburger G. Atxn2-CAG100-KnockIn mouse spinal cord shows progressive TDP43 pathology associated with cholesterol biosynthesis suppression. Neurobiol Dis 2021; 152:105289. [PMID: 33577922 DOI: 10.1016/j.nbd.2021.105289] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/11/2020] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
Large polyglutamine expansions in Ataxin-2 (ATXN2) cause multi-system nervous atrophy in Spinocerebellar Ataxia type 2 (SCA2). Intermediate size expansions carry a risk for selective motor neuron degeneration, known as Amyotrophic Lateral Sclerosis (ALS). Conversely, the depletion of ATXN2 prevents disease progression in ALS. Although ATXN2 interacts directly with RNA, and in ALS pathogenesis there is a crucial role of RNA toxicity, the affected functional pathways remain ill defined. Here, we examined an authentic SCA2 mouse model with Atxn2-CAG100-KnockIn for a first definition of molecular mechanisms in spinal cord pathology. Neurophysiology of lower limbs detected sensory neuropathy rather than motor denervation. Triple immunofluorescence demonstrated cytosolic ATXN2 aggregates sequestrating TDP43 and TIA1 from the nucleus. In immunoblots, this was accompanied by elevated CASP3, RIPK1 and PQBP1 abundance. RT-qPCR showed increase of Grn, Tlr7 and Rnaset2 mRNA versus Eif5a2, Dcp2, Uhmk1 and Kif5a decrease. These SCA2 findings overlap well with known ALS features. Similar to other ataxias and dystonias, decreased mRNA levels for Unc80, Tacr1, Gnal, Ano3, Kcna2, Elovl5 and Cdr1 contrasted with Gpnmb increase. Preterminal stage tissue showed strongly activated microglia containing ATXN2 aggregates, with parallel astrogliosis. Global transcriptome profiles from stages of incipient motor deficit versus preterminal age identified molecules with progressive downregulation, where a cluster of cholesterol biosynthesis enzymes including Dhcr24, Msmo1, Idi1 and Hmgcs1 was prominent. Gas chromatography demonstrated a massive loss of crucial cholesterol precursor metabolites. Overall, the ATXN2 protein aggregation process affects diverse subcellular compartments, in particular stress granules, endoplasmic reticulum and receptor tyrosine kinase signaling. These findings identify new targets and potential biomarkers for neuroprotective therapies.
Collapse
Affiliation(s)
- Júlia Canet-Pons
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Nesli-Ece Sen
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany; Faculty of Biosciences, Goethe University, 60438 Frankfurt am Main, Germany
| | - Aleksandar Arsović
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Luis-Enrique Almaguer-Mederos
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany; Center for Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba
| | - Melanie V Halbach
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Jana Key
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany; Faculty of Biosciences, Goethe University, 60438 Frankfurt am Main, Germany
| | - Claudia Döring
- Dr. Senckenberg Institute of Pathology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Nordrhein-Westfalen, Germany
| | - Gina Picchiarelli
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Raphaelle Cassel
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Frédérique René
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Stéphane Dieterlé
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Nina V Fuchs
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Renate König
- Host-Pathogen Interactions, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Luc Dupuis
- UMRS-1118 INSERM, Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Nordrhein-Westfalen, Germany
| | - Suzana Gispert
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany
| | - Georg Auburger
- Experimental Neurology, Medical Faculty, Goethe University, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Moore S, Meschkat M, Ruhwedel T, Trevisiol A, Tzvetanova ID, Battefeld A, Kusch K, Kole MHP, Strenzke N, Möbius W, de Hoz L, Nave KA. A role of oligodendrocytes in information processing. Nat Commun 2020; 11:5497. [PMID: 33127910 PMCID: PMC7599337 DOI: 10.1038/s41467-020-19152-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Myelinating oligodendrocytes enable fast propagation of action potentials along the ensheathed axons. In addition, oligodendrocytes play diverse non-canonical roles including axonal metabolic support and activity-dependent myelination. An open question remains whether myelination also contributes to information processing in addition to speeding up conduction velocity. Here, we analyze the role of myelin in auditory information processing using paradigms that are also good predictors of speech understanding in humans. We compare mice with different degrees of dysmyelination using acute multiunit recordings in the auditory cortex, in combination with behavioral readouts. We find complex alterations of neuronal responses that reflect fatigue and temporal acuity deficits. We observe partially discriminable but similar deficits in well myelinated mice in which glial cells cannot fully support axons metabolically. We suggest a model in which myelination contributes to sustained stimulus perception in temporally complex paradigms, with a role of metabolically active oligodendrocytes in cortical information processing.
Collapse
Affiliation(s)
- Sharlen Moore
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- International Max Planck Research School for Neurosciences, Göttingen, Germany
- Göttingen Graduate Center for Neurosciences, Biophysics and Molecular Biosciences, Georg-August-Universität Göttingen, Göttingen, Germany
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, USA
| | - Martin Meschkat
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Andrea Trevisiol
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Iva D Tzvetanova
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Section of Pharmacology, School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Arne Battefeld
- Department of Axonal Signaling, Netherlands Institute for Neurosciences, Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
- Institut des Maladies Neurodégénératives, Université de Bordeaux, Bordeaux, France
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Maarten H P Kole
- Department of Axonal Signaling, Netherlands Institute for Neurosciences, Royal Netherlands Academy of Arts and Science, Amsterdam, The Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, University of Utrecht, Utrecht, The Netherlands
| | - Nicola Strenzke
- Institute for Auditory Neuroscience, University Medical Center, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Livia de Hoz
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
- Charité Medical University, Neuroscience Research Center, Berlin, Germany.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| |
Collapse
|
16
|
White HV, Brown ST, Bozza TC, Raman IM. Effects of FGF14 and Na Vβ4 deletion on transient and resurgent Na current in cerebellar Purkinje neurons. J Gen Physiol 2019; 151:1300-1318. [PMID: 31558566 PMCID: PMC6829560 DOI: 10.1085/jgp.201912390] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/03/2019] [Indexed: 11/20/2022] Open
Abstract
Voltage-gated Na channels of Purkinje cells are specialized to maintain high availability during high-frequency repetitive firing. They enter fast-inactivated states relatively slowly and undergo a voltage-dependent open-channel block by an intracellular protein (or proteins) that prevents stable fast inactivation and generates resurgent Na current. These properties depend on the pore-forming α subunits, as well as modulatory subunits within the Na channel complex. The identity of the factors responsible for open-channel block remains a question. Here we investigate the effects of genetic mutation of two Na channel auxiliary subunits highly expressed in Purkinje cells, NaVβ4 and FGF14, on modulating Na channel blocked as well as inactivated states. We find that although both NaVβ4 and the FGF14 splice variant FGF14-1a contain sequences that can generate resurgent-like currents when applied to Na channels in peptide form, deletion of either protein, or both proteins simultaneously, does not eliminate resurgent current in acutely dissociated Purkinje cell bodies. Loss of FGF14 expression does, however, reduce resurgent current amplitude and leads to an acceleration and stabilization of inactivation that is not reversed by application of the site-3 toxin, anemone toxin II (ATX). Tetrodotoxin (TTX) sensitivity is higher for resurgent than transient components of Na current, and loss of FGF14 preferentially affects a highly TTX-sensitive subset of Purkinje α subunits. The data suggest that NaV1.6 channels, which are known to generate the majority of Purkinje cell resurgent current, bind TTX with high affinity and are modulated by FGF14 to facilitate open-channel block.
Collapse
Affiliation(s)
- Hayley V White
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Spencer T Brown
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Thomas C Bozza
- Department of Neurobiology, Northwestern University, Evanston, IL.,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, IL .,Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL
| |
Collapse
|
17
|
Venugopal S, Seki S, Terman DH, Pantazis A, Olcese R, Wiedau-Pazos M, Chandler SH. Resurgent Na+ Current Offers Noise Modulation in Bursting Neurons. PLoS Comput Biol 2019; 15:e1007154. [PMID: 31226124 PMCID: PMC6608983 DOI: 10.1371/journal.pcbi.1007154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 07/03/2019] [Accepted: 06/04/2019] [Indexed: 01/20/2023] Open
Abstract
Neurons utilize bursts of action potentials as an efficient and reliable way to encode information. It is likely that the intrinsic membrane properties of neurons involved in burst generation may also participate in preserving its temporal features. Here we examined the contribution of the persistent and resurgent components of voltage-gated Na+ currents in modulating the burst discharge in sensory neurons. Using mathematical modeling, theory and dynamic-clamp electrophysiology, we show that, distinct from the persistent Na+ component which is important for membrane resonance and burst generation, the resurgent Na+ can help stabilize burst timing features including the duration and intervals. Moreover, such a physiological role for the resurgent Na+ offered noise tolerance and preserved the regularity of burst patterns. Model analysis further predicted a negative feedback loop between the persistent and resurgent gating variables which mediate such gain in burst stability. These results highlight a novel role for the voltage-gated resurgent Na+ component in moderating the entropy of burst-encoded neural information.
Collapse
Affiliation(s)
- Sharmila Venugopal
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Soju Seki
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - David H Terman
- Department of Mathematics, The Ohio State University, Columbus, OH, United States of America
| | - Antonios Pantazis
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America.,Division of Neurobiology Department of Clinical and Experimental Medicine (IKE) and Wallenberg Center for Molecular Medicine Linköping University 581 83 Linköping Sweden
| | - Riccardo Olcese
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Martina Wiedau-Pazos
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Scott H Chandler
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, United States of America
| |
Collapse
|
18
|
Deletion of Class II ADP-Ribosylation Factors in Mice Causes Tremor by the Nav1.6 Loss in Cerebellar Purkinje Cell Axon Initial Segments. J Neurosci 2019; 39:6339-6353. [PMID: 31201232 DOI: 10.1523/jneurosci.2002-18.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 05/14/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022] Open
Abstract
ADP-ribosylation factors (ARFs) are a family of small monomeric GTPases comprising six members categorized into three classes: class I (ARF1, 2, and 3), class II (ARF4 and 5), and class III (ARF6). In contrast to class I and III ARFs, which are the key regulators in vesicular membrane trafficking, the cellular function of class II ARFs remains unclear. In the present study, we generated class II ARF-deficient mice and found that ARF4+/-/ARF5-/- mice exhibited essential tremor (ET)-like behaviors. In vivo electrophysiological recordings revealed that ARF4+/-/ARF5-/- mice of both sexes exhibited abnormal brain activity when moving, raising the possibility of abnormal cerebellar excitability. Slice patch-clamp experiments demonstrated the reduced excitability of the cerebellar Purkinje cells (PCs) in ARF4+/-/ARF5-/- mice. Immunohistochemical and electrophysiological analyses revealed a severe and selective decrease of pore-forming voltage-dependent Na+ channel subunit Nav1.6, important for maintaining repetitive action potential firing, in the axon initial segment (AIS) of PCs. Importantly, this decrease in Nav1.6 protein localized in the AIS and the consequent tremors in ARF4+/-/ARF5-/- mice could be alleviated by the PC-specific expression of ARF5 using adeno-associated virus vectors. Together, our data demonstrate that the decreased expression of the class II ARF proteins in ARF4+/-/ARF5-/- mice, leading to a haploinsufficiency of ARF4 in the absence of ARF5, impairs the localization of Nav1.6 to the AIS and hence reduces the membrane excitability in PCs, resulting in the ET-like movement disorder. We suggest that class II ARFs function in localizing specific proteins, such as Nav1.6, to the AIS.SIGNIFICANCE STATEMENT We found that decreasing the expression of class II ARF proteins, through the generation of ARF4+/-/ARF5-/- mice, impairs Nav1.6 distribution to the axon initial segment (AIS) of cerebellar Purkinje cells (PCs), thereby resulting in the impairment of action potential firing of PCs. The ARF4+/-/ARF5-/- mutant mice exhibited movement-associated essential tremor (ET)-like behavior with pharmacological profiles similar to those in ET patients. The exogenous expression of ARF5 reduced the tremor phenotype and restored the localization of Nav1.6 immunoreactivity to the AIS in ARF4+/-/ARF5-/- mice. Thus, our results suggest that class II ARFs are involved in the localization of Nav1.6 to the AISs in cerebellar PCs and that the reduction of class II ARF activity leads to ET-like movement disorder.
Collapse
|
19
|
Hong H, Sanchez JT. Need for Speed and Precision: Structural and Functional Specialization in the Cochlear Nucleus of the Avian Auditory System. J Exp Neurosci 2018; 12:1179069518815628. [PMID: 30559595 PMCID: PMC6291874 DOI: 10.1177/1179069518815628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/06/2018] [Indexed: 11/17/2022] Open
Abstract
Birds such as the barn owl and zebra finch are known for their remarkable hearing abilities that are critical for survival, communication, and vocal learning functions. A key to achieving these hearing abilities is the speed and precision required for the temporal coding of sound-a process heavily dependent on the structural, synaptic, and intrinsic specializations in the avian auditory brainstem. Here, we review recent work from us and others focusing on the specialization of neurons in the chicken cochlear nucleus magnocellularis (NM)-a first-order auditory brainstem structure analogous to bushy cells in the mammalian anteroventral cochlear nucleus. Similar to their mammalian counterpart, NM neurons are mostly adendritic and receive auditory nerve input through large axosomatic endbulb of Held synapses. Axonal projections from NM neurons to their downstream auditory targets are sophisticatedly programmed regarding their length, caliber, myelination, and conduction velocity. Specialized voltage-dependent potassium and sodium channel properties also play important and unique roles in shaping the functional phenotype of NM neurons. Working synergistically with potassium channels, an atypical current known as resurgent sodium current promotes rapid and precise action potential firing for NM neurons. Interestingly, these structural and functional specializations vary dramatically along the tonotopic axis and suggest a plethora of encoding strategies for sounds of different acoustic frequencies, mechanisms likely shared across species.
Collapse
Affiliation(s)
- Hui Hong
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Jason Tait Sanchez
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA.,Department of Neurobiology, Northwestern University, Evanston, IL, USA.,The Hugh Knowles Hearing Research Center, Northwestern University, Evanston, IL, USA
| |
Collapse
|
20
|
Gu Y, Servello D, Han Z, Lalchandani RR, Ding JB, Huang K, Gu C. Balanced Activity between Kv3 and Nav Channels Determines Fast-Spiking in Mammalian Central Neurons. iScience 2018; 9:120-137. [PMID: 30390433 PMCID: PMC6218699 DOI: 10.1016/j.isci.2018.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/11/2018] [Accepted: 10/12/2018] [Indexed: 10/29/2022] Open
Abstract
Fast-spiking (FS) neurons can fire action potentials (APs) up to 1,000 Hz and play key roles in vital functions such as sound location, motor coordination, and cognition. Here we report that the concerted actions of Kv3 voltage-gated K+ (Kv) and Na+ (Nav) channels are sufficient and necessary for inducing and maintaining FS. Voltage-clamp analysis revealed a robust correlation between the Kv3/Nav current ratio and FS. Expressing Kv3 channels alone could convert ∼30%-60% slow-spiking (SS) neurons to FS in culture. In contrast, co-expression of either Nav1.2 or Nav1.6 together with Kv3.1 or Kv3.3, but not alone or with Kv1.2, converted SS to FS with 100% efficiency. Furthermore, RNA-sequencing-based genome-wide analysis revealed that the Kv3/Nav ratio and Kv3 expression levels strongly correlated with the maximal AP frequencies. Therefore, FS is established by the properly balanced activities of Kv3 and Nav channels and could be further fine-tuned by channel biophysical features and localization patterns.
Collapse
Affiliation(s)
- Yuanzheng Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 182 Rightmire Hall, 1060 Carmack Road, Columbus, OH 43210, USA
| | - Dustin Servello
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Zhi Han
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; College of Software, Nankai University, Tianjin 300071, China; Regenstrief Institute, Indianapolis, IN 46202, USA
| | - Rupa R Lalchandani
- Department of Neurosurgery, and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jun B Ding
- Department of Neurosurgery, and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Regenstrief Institute, Indianapolis, IN 46202, USA; School of Biomedical Engineering, Shenzhen University, Shenzhen 518037, China
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 182 Rightmire Hall, 1060 Carmack Road, Columbus, OH 43210, USA; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
21
|
Ransdell JL, Nerbonne JM. Voltage-gated sodium currents in cerebellar Purkinje neurons: functional and molecular diversity. Cell Mol Life Sci 2018; 75:3495-3505. [PMID: 29982847 PMCID: PMC6123253 DOI: 10.1007/s00018-018-2868-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 01/09/2023]
Abstract
Purkinje neurons, the sole output of the cerebellar cortex, deliver GABA-mediated inhibition to the deep cerebellar nuclei. To subserve this critical function, Purkinje neurons fire repetitively, and at high frequencies, features that have been linked to the unique properties of the voltage-gated sodium (Nav) channels expressed. In addition to the rapidly activating and inactivating, or transient, component of the Nav current (INaT) present in many types of central and peripheral neurons, Purkinje neurons, also expresses persistent (INaP) and resurgent (INaR) Nav currents. Considerable progress has been made in detailing the biophysical properties and identifying the molecular determinants of these discrete Nav current components, as well as defining their roles in the regulation of Purkinje neuron excitability. Here, we review this important work and highlight the remaining questions about the molecular mechanisms controlling the expression and the functioning of Nav currents in Purkinje neurons. We also discuss the impact of the dynamic regulation of Nav currents on the functioning of individual Purkinje neurons and cerebellar circuits.
Collapse
Affiliation(s)
- Joseph L Ransdell
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, Box 8086, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Jeanne M Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Medicine, Washington University School of Medicine, Box 8086, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
| |
Collapse
|
22
|
Hoxha E, Balbo I, Miniaci MC, Tempia F. Purkinje Cell Signaling Deficits in Animal Models of Ataxia. Front Synaptic Neurosci 2018; 10:6. [PMID: 29760657 PMCID: PMC5937225 DOI: 10.3389/fnsyn.2018.00006] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022] Open
Abstract
Purkinje cell (PC) dysfunction or degeneration is the most frequent finding in animal models with ataxic symptoms. Mutations affecting intrinsic membrane properties can lead to ataxia by altering the firing rate of PCs or their firing pattern. However, the relationship between specific firing alterations and motor symptoms is not yet clear, and in some cases PC dysfunction precedes the onset of ataxic signs. Moreover, a great variety of ionic and synaptic mechanisms can affect PC signaling, resulting in different features of motor dysfunction. Mutations affecting Na+ channels (NaV1.1, NaV1.6, NaVβ4, Fgf14 or Rer1) reduce the firing rate of PCs, mainly via an impairment of the Na+ resurgent current. Mutations that reduce Kv3 currents limit the firing rate frequency range. Mutations of Kv1 channels act mainly on inhibitory interneurons, generating excessive GABAergic signaling onto PCs, resulting in episodic ataxia. Kv4.3 mutations are responsible for a complex syndrome with several neurologic dysfunctions including ataxia. Mutations of either Cav or BK channels have similar consequences, consisting in a disruption of the firing pattern of PCs, with loss of precision, leading to ataxia. Another category of pathogenic mechanisms of ataxia regards alterations of synaptic signals arriving at the PC. At the parallel fiber (PF)-PC synapse, mutations of glutamate delta-2 (GluD2) or its ligand Crbl1 are responsible for the loss of synaptic contacts, abolishment of long-term depression (LTD) and motor deficits. At the same synapse, a correct function of metabotropic glutamate receptor 1 (mGlu1) receptors is necessary to avoid ataxia. Failure of climbing fiber (CF) maturation and establishment of PC mono-innervation occurs in a great number of mutant mice, including mGlu1 and its transduction pathway, GluD2, semaphorins and their receptors. All these models have in common the alteration of PC output signals, due to a variety of mechanisms affecting incoming synaptic signals or the way they are processed by the repertoire of ionic channels responsible for intrinsic membrane properties. Although the PC is a final common pathway of ataxia, the link between specific firing alterations and neurologic symptoms has not yet been systematically studied and the alterations of the cerebellar contribution to motor signals are still unknown.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Ilaria Balbo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy.,Department of Neuroscience, University of Torino, Turin, Italy.,National Institute of Neuroscience (INN), Turin, Italy
| |
Collapse
|
23
|
Abstract
Voltage-gated sodium channels are protein complexes comprised of one pore forming α subunit and two, non-pore forming, β subunits. The voltage-gated sodium channel β subunits were originally identified to function as auxiliary subunits, which modulate the gating, kinetics, and localization of the ion channel pore. Since that time, the five β subunits have been shown to play crucial roles as multifunctional signaling molecules involved in cell adhesion, cell migration, neuronal pathfinding, fasciculation, and neurite outgrowth. Here, we provide an overview of the evidence implicating the β subunits in their conducting and non-conducting roles. Mutations in the β subunit genes (SCN1B-SCN4B) have been linked to a variety of diseases. These include cancer, epilepsy, cardiac arrhythmias, sudden infant death syndrome/sudden unexpected death in epilepsy, neuropathic pain, and multiple neurodegenerative disorders. β subunits thus provide novel therapeutic targets for future drug discovery.
Collapse
Affiliation(s)
- Alexandra A Bouza
- Department of Pharmacology, University of Michigan Medical School, 2200 MSRBIII, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA.
| |
Collapse
|
24
|
Identification of Persistent and Resurgent Sodium Currents in Spiral Ganglion Neurons Cultured from the Mouse Cochlea. eNeuro 2017; 4:eN-NWR-0303-17. [PMID: 29138759 PMCID: PMC5684619 DOI: 10.1523/eneuro.0303-17.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 01/11/2023] Open
Abstract
In spiral ganglion neurons (SGNs), the afferent single units of the auditory nerve, high spontaneous and evoked firing rates ensure preservation of the temporal code describing the key features of incoming sound. During postnatal development, the spatiotemporal distribution of ion channel subtypes contributes to the maturation of action potential generation in SGNs, and to their ability to generate spike patterns that follow rapidly changing inputs. Here we describe tetrodotoxin (TTX)-sensitive Na+ currents in SGNs cultured from mice, whose properties may support this fast spiking behavior. A subthreshold persistent Na+ current (INaP) and a resurgent Na+ current (INaR) both emerged prior to the onset of hearing and became more prevalent as hearing matured. Navβ4 subunits, which are proposed to play a key role in mediating INaR elsewhere in the nervous system, were immunolocalized to the first heminode where spikes are generated in the auditory nerve, and to perisomatic nodes of Ranvier. ATX-II, a sea anemone toxin that slows classical Na+ channel inactivation selectively, enhanced INaP five-fold and INaR three-fold in voltage clamp recordings. In rapidly-adapting SGNs under current clamp, ATX-II increased the likelihood of firing additional action potentials. The data identify INaP and INaR as novel regulators of excitability in SGNs, and consistent with their roles in other neuronal types, we suggest that these nonclassical Na+ currents may contribute to the control of refractoriness in the auditory nerve.
Collapse
|
25
|
Enomoto A, Seki S, Tanaka S, Ishihama K, Yamanishi T, Kogo M, Hamada S. Development of resurgent and persistent sodium currents in mesencephalic trigeminal neurons. J Neurosci Res 2017; 96:305-312. [PMID: 28752895 DOI: 10.1002/jnr.24134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 12/18/2022]
Abstract
Sodium channels play multiple roles in the formation of neural membrane properties in mesencephalic trigeminal (Mes V) neurons and in other neural systems. Mes V neurons exhibit conditional robust high-frequency spike discharges. As previously reported, resurgent and persistent sodium currents (INaR and INaP , respectively) may carry small currents at subthreshold voltages that contribute to generation of spike firing. These currents play an important role in maintaining and allowing high-frequency spike discharge during a burst. In the present study, we investigated the developmental changes in tetrodotoxin-sensitive INaR and INaP underlying high-frequency spike discharges in Mes V neurons. Whole-cell patch-clamp recordings showed that both current densities increased one and a half times from postnatal day (P) 0-6 neurons to P7-14 neurons. Although these neurons do not exhibit subthreshold oscillations or burst discharges with high-frequency firing, INaR and INaP do exist in Mes V neurons at P0-6. When the spike frequency at rheobase was examined in firing Mes V neurons, the developmental change in firing frequency among P7-14 neurons was significant. INaR and INaP density at -40 mV also increased significantly among P7-14 neurons. The change to an increase in excitability in the P7-14 group could result from this quantitative change in INaP. In neurons older than P7 that exhibit repetitive firing, quantitative increases in INaR and INaP density may be major factors that facilitate and promote high-frequency firing as a function of age in Mes V neurons.
Collapse
Affiliation(s)
- Akifumi Enomoto
- Department of Oral and Maxillofacial Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- 1st Department of Oral Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Soju Seki
- 1st Department of Oral Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Susumu Tanaka
- 1st Department of Oral Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Kohji Ishihama
- 1st Department of Oral Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Tadashi Yamanishi
- 1st Department of Oral Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Mikihiko Kogo
- 1st Department of Oral Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Suguru Hamada
- Department of Oral and Maxillofacial Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
26
|
Molinarolo S, Granata D, Carnevale V, Ahern CA. Mining Protein Evolution for Insights into Mechanisms of Voltage-Dependent Sodium Channel Auxiliary Subunits. Handb Exp Pharmacol 2017; 246:33-49. [PMID: 29464397 DOI: 10.1007/164_2017_75] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Voltage-gated sodium channel (VGSC) beta (β) subunits have been called the "overachieving" auxiliary ion channel subunit. Indeed, these subunits regulate the trafficking of the sodium channel complex at the plasma membrane and simultaneously tune the voltage-dependent properties of the pore-forming alpha-subunit. It is now known that VGSC β-subunits are capable of similar modulation of multiple isoforms of related voltage-gated potassium channels, suggesting that their abilities extend into the broader voltage-gated channels. The gene family for these single transmembrane immunoglobulin beta-fold proteins extends well beyond the traditional VGSC β1-β4 subunit designation, with deep roots into the cell adhesion protein family and myelin-related proteins - where inherited mutations result in a myriad of electrical signaling disorders. Yet, very little is known about how VGSC β-subunits support protein trafficking pathways, the basis for their modulation of voltage-dependent gating, and, ultimately, their role in shaping neuronal excitability. An evolutionary approach can be useful in yielding new clues to such functions as it provides an unbiased assessment of protein residues, folds, and functions. An approach is described here which indicates the greater emergence of the modern β-subunits roughly 400 million years ago in the early neurons of Bilateria and bony fish, and the unexpected presence of distant homologues in bacteriophages. Recent structural breakthroughs containing α and β eukaryotic sodium channels containing subunits suggest a novel role for a highly conserved polar contact that occurs within the transmembrane segments. Overall, a mixture of approaches will ultimately advance our understanding of the mechanism for β-subunit interactions with voltage-sensor containing ion channels and membrane proteins.
Collapse
Affiliation(s)
- Steven Molinarolo
- Department of Molecular Physiology and Biophysics, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Daniele Granata
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| | - Christopher A Ahern
- Department of Molecular Physiology and Biophysics, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|