Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
J Neurosci ; 44(21)2024 May 22.
Article in English | MEDLINE | ID: mdl-38664011

ABSTRACT

Fragile X syndrome (FXS) arises from the loss of fragile X messenger ribonucleoprotein (FMRP) needed for normal neuronal excitability and circuit functions. Recent work revealed that FMRP contributes to mossy fiber long-term potentiation by adjusting the Kv4 A-type current availability through interactions with a Cav3-Kv4 ion channel complex, yet the mechanism has not yet been defined. In this study using wild-type and Fmr1 knock-out (KO) tsA-201 cells and cerebellar sections from male Fmr1 KO mice, we show that FMRP associates with all subunits of the Cav3.1-Kv4.3-KChIP3 complex and is critical to enabling calcium-dependent shifts in Kv4.3 inactivation to modulate the A-type current. Specifically, upon depolarization Cav3 calcium influx activates dual-specific phosphatase 1/6 (DUSP1/6) to deactivate ERK1/2 (ERK) and lower phosphorylation of Kv4.3, a signaling pathway that does not function in Fmr1 KO cells. In Fmr1 KO mouse tissue slices, cerebellar granule cells exhibit a hyperexcitable response to membrane depolarizations. Either incubating Fmr1 KO cells or in vivo administration of a tat-conjugated FMRP N-terminus fragment (FMRP-N-tat) rescued Cav3-Kv4 function and granule cell excitability, with a decrease in the level of DUSP6. Together these data reveal a Cav3-activated DUSP signaling pathway critical to the function of a FMRP-Cav3-Kv4 complex that is misregulated in Fmr1 KO conditions. Moreover, FMRP-N-tat restores function of this complex to rescue calcium-dependent control of neuronal excitability as a potential therapeutic approach to alleviating the symptoms of FXS.


Subject(s)
Calcium , Fragile X Mental Retardation Protein , Fragile X Syndrome , Mice, Knockout , Neurons , Animals , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Mice , Male , Fragile X Syndrome/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Neurons/metabolism , Calcium/metabolism , Mice, Inbred C57BL , Shal Potassium Channels/metabolism , Shal Potassium Channels/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism
2.
EBioMedicine ; 91: 104565, 2023 May.
Article in English | MEDLINE | ID: mdl-37088035

ABSTRACT

BACKGROUND: Extracellular signal-regulated kinase (ERK/MAPK) pathway in the brain is hypothesized to be a critical convergent node in the development of autism spectrum disorder. We reasoned that selectively targeting this pathway could reverse core autism-like phenotype in animal models. METHODS: Here we tested a clinically relevant, selective inhibitor of ERK pathway, PD325901 (Mirdametinib), in a mouse model of idiopathic autism, the BTBR mice. FINDINGS: We report that treating juvenile mice with PD325901 reduced ERK pathway activation, dose and duration-dependently reduced core disease-modeling deficits in sociability, vocalization and repetitive behavior, and reversed abnormal EEG signals. Further analysis revealed that subchronic treatment did not affect weight gain, locomotion, or neuronal density in the brain. Parallel treatment in the C57BL/6J mice did not alter their phenotype. INTERPRETATION: Our data indicate that selectively inhibiting ERK pathway using PD325901 is beneficial in the BTBR model, thus further support the notion that ERK pathway is critically involved in the pathophysiology of autism. These results suggest that a similar approach could be applied to animal models of syndromic autism with dysregulated ERK signaling, to further test selectively targeting ERK pathway as a new approach for treating autism. FUNDING: This has beenwork was supported by Alberta Children's Hospital Research Foundation (JMR & NC), University of Calgary Faculty of Veterinary Medicine (NC), Kids Brain Health Network (NC), and Natural Sciences and Engineering Research Council of Canada (NC).


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Mice , Animals , Autistic Disorder/drug therapy , Autistic Disorder/metabolism , Autism Spectrum Disorder/metabolism , Mice, Inbred C57BL , MAP Kinase Signaling System , Mice, Inbred Strains , Disease Models, Animal
3.
iScience ; 25(10): 105092, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36157585

ABSTRACT

Epilepsy and neurodevelopmental disorders can arise from pathogenic variants of KCNQ (Kv7) channels. A patient with developmental and epileptic encephalopathy exhibited an in-frame deletion of histidine 260 on Kv7.2. Coexpression of Kv7.2 mutant (mut) subunits with Kv7.3 invoked a decrease in current density, a depolarizing shift in voltage for activation, and a decrease in membrane conductance. Biotinylation revealed an increased level of surface Kv7.2mut compared to Kv7.3 with no change in total membrane protein expression. Super-resolution and FRET imaging confirmed heteromeric channel formation and a higher expression density of Kv7.2mut. Cannabidiol (1 µM) offset the effects of Kv7.2mut by inducing a hyperpolarizing shift in voltage for activation independent of CB1 or CB2 receptors. These data reveal that the ability for cannabidiol to reduce the effects of a pathogenic Kv7.2 variant supports its use as a potential therapeutic to reduce seizure activity.

4.
Alzheimers Dement ; 18(11): 2088-2098, 2022 11.
Article in English | MEDLINE | ID: mdl-34985200

ABSTRACT

INTRODUCTION: Neuronal hyperactivity is an early neuronal defect commonly observed in familial and sporadic Alzheimer's disease (AD), but the underlying mechanisms are unclear. METHODS: We employed a ryanodine receptor 2 (RyR2) mutant mouse model harboring the R4496C+/- mutation that markedly increases the channel's open probability (Po) to determine the impact of increased RyR2 activity in neuronal function without AD gene mutations. RESULTS: Genetically increasing RyR2 Po induced neuronal hyperactivity in vivo in anesthetized and awake mice. Increased RyR2 Po induced hyperactive behaviors, impaired learning and memory, defective dendritic spines, and neuronal cell death. Increased RyR2 Po exacerbated the onset of neuronal hyperexcitability and learning and memory impairments in 5xFAD mice. DISCUSSION: Increased RyR2 Po exacerbates the onset of familial AD-associated neuronal dysfunction, and induces AD-like defects in the absence of AD-causing gene mutations, suggesting that RyR2-associated neuronal hyperactivity represents a common target for combating AD with or without AD gene mutations.


Subject(s)
Alzheimer Disease , Ryanodine Receptor Calcium Release Channel , Mice , Animals , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Alzheimer Disease/genetics , Mutation/genetics , Memory Disorders/genetics , Amnesia , Probability , Disease Models, Animal
5.
Mol Brain ; 14(1): 115, 2021 07 17.
Article in English | MEDLINE | ID: mdl-34274007

ABSTRACT

T-type calcium channels are important regulators of neuronal excitability. The mammalian brain expresses three T-type channel isoforms (Cav3.1, Cav3.2 and Cav3.3) with distinct biophysical properties that are critically regulated by temperature. Here, we test the effects of how temperature affects spike output in a reduced firing neuron model expressing specific Cav3 channel isoforms. The modeling data revealed only a minimal effect on baseline spontaneous firing near rest, but a dramatic increase in rebound burst discharge frequency for Cav3.1 compared to Cav3.2 or Cav3.3 due to differences in window current or activation/recovery time constants. The reduced response by Cav3.2 could optimize its activity where it is expressed in peripheral tissues more subject to temperature variations than Cav3.1 or Cav3.3 channels expressed prominently in the brain. These tests thus reveal that aspects of neuronal firing behavior are critically dependent on both temperature and T-type calcium channel subtype.


Subject(s)
Action Potentials/physiology , Calcium Channels, T-Type/metabolism , Models, Neurological , Temperature , HEK293 Cells , Humans , Neurons/metabolism
6.
Front Physiol ; 12: 759707, 2021.
Article in English | MEDLINE | ID: mdl-35002757

ABSTRACT

Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.

7.
Cell Rep ; 32(12): 108169, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966798

ABSTRACT

Neuronal hyperactivity is an early primary dysfunction in Alzheimer's disease (AD) in humans and animal models, but effective neuronal hyperactivity-directed anti-AD therapeutic agents are lacking. Here we define a previously unknown mode of ryanodine receptor 2 (RyR2) control of neuronal hyperactivity and AD progression. We show that a single RyR2 point mutation, E4872Q, which reduces RyR2 open time, prevents hyperexcitability, hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset AD mouse model (5xFAD). The RyR2-E4872Q mutation upregulates hippocampal CA1-pyramidal cell A-type K+ current, a well-known neuronal excitability control that is downregulated in AD. Pharmacologically limiting RyR2 open time with the R-carvedilol enantiomer (but not racemic carvedilol) prevents and rescues neuronal hyperactivity, memory impairment, and neuron loss even in late stages of AD. These AD-related deficits are prevented even with continued ß-amyloid accumulation. Thus, limiting RyR2 open time may be a hyperactivity-directed, non-ß-amyloid-targeted anti-AD strategy.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Memory Disorders/complications , Memory Disorders/pathology , Neurons/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Alzheimer Disease/physiopathology , Animals , CA1 Region, Hippocampal/pathology , Carvedilol/pharmacology , Dendritic Spines/drug effects , Dendritic Spines/pathology , Ion Channel Gating , Long-Term Potentiation , Memory Disorders/physiopathology , Mice, Transgenic , Mutation/genetics , Neuroprotection/drug effects , Potassium Channels/metabolism , Pyramidal Cells/pathology , Ryanodine Receptor Calcium Release Channel/genetics , Time Factors , Up-Regulation
8.
Nat Commun ; 11(1): 2755, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488011

ABSTRACT

Fragile X Syndrome results from a loss of Fragile X Mental Retardation Protein (FMRP). We now show that FMRP is a member of a Cav3-Kv4 ion channel complex that is known to regulate A-type potassium current in cerebellar granule cells to produce mossy fiber LTP. Mossy fiber LTP is absent in Fmr1 knockout (KO) mice but is restored by FMRP(1-297)-tat peptide. This peptide further rapidly permeates the blood-brain barrier to enter cells across the cerebellar-cortical axis that restores the balance of protein translation for at least 24 h and transiently reduces elevated levels of activity of adult Fmr1 KO mice in the Open Field Test. These data reveal that FMRP(1-297)-tat can improve function from the levels of protein translation to synaptic efficacy and behaviour in a model of Fragile X syndrome, identifying a potential therapeutic strategy for this genetic disorder.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Ion Channels/metabolism , Animals , Brain/pathology , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Male , Mice , Mice, Knockout , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Protein Biosynthesis
9.
Cell Rep ; 28(9): 2427-2442.e6, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31461656

ABSTRACT

The excitability of CA1 hippocampal pyramidal cells is mediated by a slow AHP (sAHP) that responds to calcium increases by Cav1 calcium channels and ryanodine receptors (RyR). We used super-resolution and FRET microscopy to investigate the proximity and functional coupling among Cav1.3/Cav1.2, RyR2, and KCa3.1 potassium channels that contribute to the sAHP. dSTORM and FRET imaging shows that Cav1.3, RyR2, and KCa3.1 are organized as a triprotein complex that colocalizes with junctophilin (JPH) 3 and 4 proteins that tether the plasma membrane to the endoplasmic reticulum. JPH3 and JPH4 shRNAs dissociated a Cav1.3-RyR2-KCa3.1 complex and reduced the IsAHP. Infusing JPH3 and JPH4 antibodies into CA1 cells reduced IsAHP and spike accommodation. These data indicate that JPH3 and JPH4 proteins maintain a Cav1-RyR2-KCa3.1 complex that allows two calcium sources to act in tandem to define the activation properties of KCa3.1 channels and the IsAHP.


Subject(s)
Action Potentials , Calcium Channels, L-Type/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Female , Humans , Male , Membrane Proteins/genetics , Mice , Neurons/physiology , Rats , Rats, Sprague-Dawley
10.
Mol Pharmacol ; 96(1): 115-126, 2019 07.
Article in English | MEDLINE | ID: mdl-31048549

ABSTRACT

Three small conductance calcium-activated potassium channel (SK) subunits have been cloned and found to preferentially form heteromeric channels when expressed in a heterologous expression system. The original cloning of the gene encoding the intermediate conductance calcium-activated potassium channel (IKCa) was termed SK4 because of the high homology between channel subtypes. Recent immunovisualization suggests that IKCa is expressed in the same subcellular compartments of some neurons as SK channel subunits. Stochastic optical reconstruction microscopy super-resolution microscopy revealed that coexpressed IKCa and SK1 channel subunits were closely associated, a finding substantiated by measurement of fluorescence resonance energy transfer between coexpressed fluorophore-tagged subunits. Expression of homomeric SK1 channels produced current that displayed typical sensitivity to SK channel inhibitors, while expressed IKCa channel current was inhibited by known IKCa channel blockers. Expression of both SK1 and IKCa subunits gave a current that displayed no sensitivity to SK channel inhibitors and a decreased sensitivity to IKCa current inhibitors. Single channel recording indicated that coexpression of SK1 and IKCa subunits produced channels with properties intermediate between those observed for homomeric channels. These data indicate that SK1 and IKCa channel subunits preferentially combine to form heteromeric channels that display pharmacological and biophysical properties distinct from those seen with homomeric channels.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Multiprotein Complexes/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Cell Line , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Microscopy , Stochastic Processes
11.
J Neurosci ; 37(46): 11255-11270, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29038242

ABSTRACT

CaV1 L-type calcium channels are key to regulating neuronal excitability, with the range of functional roles enhanced by interactions with calmodulin, accessory proteins, or CaMKII that modulate channel activity. In hippocampal pyramidal cells, a prominent elevation of CaV1 activity is apparent in late channel openings that can last for seconds following a depolarizing stimulus train. The current study tested the hypothesis that a reported interaction among CaV1.3 channels, the scaffolding protein densin, and CaMKII could generate a facilitation of channel activity that outlasts a depolarizing stimulus. We found that CaV1.3 but not CaV1.2 channels exhibit a long-duration calcium-dependent facilitation (L-CDF) that lasts up to 8 s following a brief 50 Hz stimulus train, but only when coexpressed with densin and CaMKII. To test the physiological role for CaV1.3 L-CDF, we coexpressed the intermediate-conductance KCa3.1 potassium channel, revealing a strong functional coupling to CaV1.3 channel activity that was accentuated by densin and CaMKII. Moreover, the CaV1.3-densin-CaMKII interaction gave rise to an outward tail current of up to 8 s duration following a depolarizing stimulus in both tsA-201 cells and male rat CA1 pyramidal cells. A slow afterhyperpolarization in pyramidal cells was reduced by a selective block of CaV1 channels by isradipine, a CaMKII blocker, and siRNA knockdown of densin, and spike frequency increased upon selective block of CaV1 channel conductance. The results are important in revealing a CaV1.3-densin-CaMKII interaction that extends the contribution of CaV1.3 calcium influx to a time frame well beyond a brief input train.SIGNIFICANCE STATEMENT CaV1 L-type calcium channels play a key role in regulating the output of central neurons by providing calcium influx during repetitive inputs. This study identifies a long-duration calcium-dependent facilitation (L-CDF) of CaV1.3 channels that depends on the scaffolding protein densin and CaMKII and that outlasts a depolarizing stimulus by seconds. We further show a tight functional coupling between CaV1.3 calcium influx and the intermediate-conductance KCa3.1 potassium channel that promotes an outward tail current of up to 8 s following a depolarizing stimulus. Tests in CA1 hippocampal pyramidal cells reveal that a slow AHP is reduced by blocking different components of the CaV1.3-densin-CaMKII interaction, identifying an important role for CaV1.3 L-CDF in regulating neuronal excitability.


Subject(s)
Action Potentials/physiology , Calcium Channels/metabolism , Hippocampus/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Neurons/metabolism , Action Potentials/drug effects , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Hippocampus/drug effects , Male , Neurons/drug effects , Organ Culture Techniques , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/metabolism
12.
Mol Brain ; 10(1): 37, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28800734

ABSTRACT

Calmodulin (CaM) is an important signaling molecule that regulates a vast array of cellular functions by activating second messengers involved in cell function and plasticity. Low voltage-activated calcium channels of the Cav3 family have the important role of mediating low threshold calcium influx, but were not believed to interact with CaM. We find a constitutive association between CaM and the Cav3.1 channel at rest that is lost through an activity-dependent and Cav3.1 calcium-dependent CaM dissociation. Moreover, Cav3 calcium influx is sufficient to activate αCaMKII in the cytoplasm in a manner that depends on an intact Cav3.1 C-terminus needed to support the CaM interaction. Our findings thus establish that T-type channel calcium influx invokes a novel dynamic interaction between CaM and Cav3.1 channels to trigger a signaling cascade that leads to αCaMKII activation.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Animals , Calcium/metabolism , Enzyme Activation , Fluorescence Resonance Energy Transfer , Humans , Immunoprecipitation , Mice, Inbred C57BL , Neurons/metabolism , Phosphorylation , Protein Aggregates , Rats, Sprague-Dawley
13.
J Physiol ; 595(19): 6223, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28786112
14.
J Neurosci ; 36(44): 11196-11207, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807163

ABSTRACT

Mossy fiber afferents to cerebellar granule cells form the primary synaptic relay into cerebellum, providing an ideal site to process signal inputs differentially. Mossy fiber input is known to exhibit a long-term potentiation (LTP) of synaptic efficacy through a combination of presynaptic and postsynaptic mechanisms. However, the specific postsynaptic mechanisms contributing to LTP of mossy fiber input is unknown. The current study tested the hypothesis that LTP induces a change in intrinsic membrane excitability of rat cerebellar granule cells through modification of Kv4 A-type potassium channels. We found that theta-burst stimulation of mossy fiber input in lobule 9 granule cells lowered the current threshold to spike and increases the gain of spike firing by 2- to 3-fold. The change in postsynaptic excitability was traced to hyperpolarizing shifts in both the half-inactivation and half-activation potentials of Kv4 that occurred upon coactivating NMDAR and group I metabotropic glutamatergic receptors. The effects of theta-burst stimulation on Kv4 channel control of the gain of spike firing depended on a signaling cascade leading to extracellular signal-related kinase activation. Under physiological conditions, LTP of synaptically evoked spike output was expressed preferentially for short bursts characteristic of sensory input, helping to shape signal processing at the mossy fiber-granule cell relay. SIGNIFICANCE STATEMENT: Cerebellar granule cells receive mossy fiber inputs that convey information on different sensory modalities and feedback from descending cortical projections. Recent work suggests that signal processing across multiple cerebellar lobules is controlled differentially by postsynaptic ionic mechanisms at the level of granule cells. We found that long-term potentiation (LTP) of mossy fiber input invoked a large increase in granule cell excitability by modifying the biophysical properties of Kv4 channels through a specific signaling cascade. LTP of granule cell output became evident in response to bursts of mossy fiber input, revealing that Kv4 control of intrinsic excitability is modified to respond most effectively to patterns of afferent input that are characteristic of physiological sensory patterns.


Subject(s)
Long-Term Potentiation/physiology , Nerve Fibers/physiology , Neurons/physiology , Second Messenger Systems/physiology , Shal Potassium Channels/metabolism , Synaptic Transmission/physiology , Animals , Cells, Cultured , Ion Channel Gating/physiology , Male , Rats , Rats, Sprague-Dawley , Synaptic Potentials/physiology
15.
Channels (Austin) ; 10(4): 313-9, 2016 Jul 03.
Article in English | MEDLINE | ID: mdl-26950800

ABSTRACT

Our previous work reported that KCa3.1 (IKCa) channels are expressed in CA1 hippocampal pyramidal cells and contribute to the slow afterhyperpolarization that regulates spike accommodation in these cells. The current report presents data from single cell RT-PCR that further reveals mRNA in CA1 cells that corresponds to the sequence of an IKCa channel from transmembrane segments 5 through 6 including the pore region, revealing the established binding sites for 4 different IKCa channel blockers. A comparison of methods to internally apply the IKCa channel blocker TRAM-34 shows that including the drug in an electrode from the onset of an experiment is unviable given the speed of drug action upon gaining access for whole-cell recordings. Together the data firmly establish IKCa channel expression in CA1 neurons and clarify methodological requirements to obtain a block of IKCa channel activity through internal application of TRAM-34.


Subject(s)
CA1 Region, Hippocampal/cytology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Pyramidal Cells/physiology , Animals , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Membrane Potentials , Patch-Clamp Techniques , Polymerase Chain Reaction , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
16.
J Physiol ; 594(4): 985-1003, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26662168

ABSTRACT

KEY POINTS: Cerebellar Purkinje cells project GABAergic inhibitory input to neurons of the deep cerebellar nuclei (DCN) that generate a rebound increase in firing, but the specific patterns of input that might elicit a rebound response have not been established. We used recordings of Purkinje cell firing obtained during perioral whisker stimulation in vivo to create a physiological stimulus template to activate Purkinje cell afferents in vitro. DCN cell bursts were evoked by the stimulus pattern but not in relation to the perioral whisker stimulus, complex spikes or regular patterns within the Purkinje cell record. Reverse correlation revealed that bursts were triggered by an elevation-pause pattern of Purkinje cell firing, with pause duration a key factor in burst generation. Our data identify for the first time a physiological pattern of Purkinje cell input that can be encoded by the generation of rebound bursts in DCN cells. ABSTRACT: The end result of signal processing in cerebellar cortex is encoded in the output of Purkinje cells that project inhibitory input to deep cerebellar nuclear (DCN) neurons. DCN cells can respond to a period of inhibition in vitro with a rebound burst of firing, yet the optimal physiological pattern of Purkinje cell input that might evoke a rebound burst is unknown. The current study used spike trains recorded from rat Purkinje cells in response to perioral stimuli in vivo to create a physiological pattern to stimulate Purkinje cell axons in vitro. The perioral stimulus-evoked Purkinje cell firing pattern proved to be virtually ineffective in evoking a rebound burst despite the ability to reliably evoke rebounds using a traditional brief 100 Hz stimulus. Similarly, neither complex spike firing nor Purkinje cell patterns identified by CV2 analysis were reliably associated with rebound bursts. Reverse correlation revealed that the optimal Purkinje cell input to evoke a rebound burst was a sequential increase in mean firing rate of at least 30 Hz above baseline over 250 ms followed by a reduction of 40-60 Hz below baseline for up to 500 ms. The most important factor was the duration of a pause in Purkinje cell firing that allowed DCN cells to recover from a state of net inhibitory influence. These data indicate that physiological patterns of Purkinje cell firing can elicit rebound bursts in DCN cells in vitro, with pauses in Purkinje cell firing rate acting as a key stimulus for DCN cell rebound responses.


Subject(s)
Action Potentials , Cerebellar Nuclei/physiology , Evoked Potentials , Purkinje Cells/physiology , Animals , Axons/physiology , Rats , Rats, Sprague-Dawley
17.
Cell Rep ; 11(2): 175-82, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25865881

ABSTRACT

Control over the frequency and pattern of neuronal spike discharge depends on Ca2+-gated K+ channels that reduce cell excitability by hyperpolarizing the membrane potential. The Ca2+-dependent slow afterhyperpolarization (sAHP) is one of the most prominent inhibitory responses in the brain, with sAHP amplitude linked to a host of circuit and behavioral functions, yet the channel that underlies the sAHP has defied identification for decades. Here, we show that intermediate-conductance Ca2+-dependent K+ (IKCa) channels underlie the sAHP generated by trains of synaptic input or postsynaptic stimuli in CA1 hippocampal pyramidal cells. These findings are significant in providing a molecular identity for the sAHP of central neurons that will identify pharmacological tools capable of potentially modifying the several behavioral or disease states associated with the sAHP.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Neurons/physiology , Potassium Channels, Calcium-Activated/chemistry , Pyramidal Cells/physiology , Action Potentials/physiology , Animals , CA1 Region, Hippocampal/chemistry , CA1 Region, Hippocampal/physiology , Cell Polarity/physiology , Hippocampus/chemistry , Hippocampus/physiology , Mice , Neurons/chemistry , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/metabolism , Pyramidal Cells/chemistry
18.
Pflugers Arch ; 467(2): 311-28, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24797146

ABSTRACT

The expression pattern and functional roles for calcium-activated potassium channels of the KCa2.x family and KCa1.1 have been extensively examined in central neurons. Recent work indicates that intermediate conductance calcium-activated potassium channels (KCa3.1) are also expressed in central neurons of the cerebellum and spinal cord. The current study used immunocytochemistry and GFP linked to KCNN4 promoter activity in a transgenic mouse to determine the expression pattern of KCa3.1 channels in rat or mouse neocortex, hippocampus, thalamus, and cerebellum. KCa3.1 immunolabel and GFP expression were closely matched and detected in both excitatory and inhibitory cells of all regions examined. KCa3.1 immunolabel was localized primarily to the somatic region of excitatory cells in cortical structures but at the soma and over longer segments of dendrites of cells in deep cerebellar nuclei. More extensive labeling was apparent for inhibitory cells at the somatic and dendritic level with no detectable label associated with axon tracts or regions of intense synaptic innervation. The data indicate that KCa3.1 channels are expressed in the CNS with a differential pattern of distribution between cells, suggesting important functional roles for these calcium-activated potassium channels in regulating the excitability of central neurons.


Subject(s)
Brain/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Neurons/metabolism , Animals , Brain/cytology , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Rats , Rats, Sprague-Dawley , Species Specificity
19.
J Neurosci ; 34(26): 8800-12, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24966380

ABSTRACT

The cerebellum receives sensory information by mossy fiber input from a multitude of sources that require differential signal processing. A compartmentalization of function begins with the segregation of mossy fibers across 10 distinct lobules over the rostrocaudal axis, with tactile receptor afferents prevalent in anterior lobules and vestibular input in caudal lobules. However, it is unclear how these unique signals might be differentially processed at the circuit level across the cerebellum. As granule cells receive mossy fiber input, they represent a key stage at which postsynaptic mechanisms could influence signal processing. Granule cells express an A-type current mediated by Kv4 potassium channels that modify the latency and frequency of spike output. The current study examined the potential for a Cav3 calcium-Kv4 channel complex to regulate the response of granule cells to mossy fiber input in lobules 2 and 9 of the rat cerebellum. Similar A-type currents were recorded in both regions, but the Cav3 calcium current was expressed at a substantially higher density in lobule 9 cells, acting to increase A-type current availability through its influence on Kv4 voltage for inactivation. The difference in excitability imparted by Cav3-Kv4 interactions proves to allow lobule 2 granule cells to respond more effectively to tactile stimulus-like burst input and lobule 9 cells to slow shifts in input frequency characteristic of vestibular input. The expression pattern of Cav3 channels and its control of Kv4 availability thus provides a novel means of processing widely different forms of sensory input across cerebellar lobules.


Subject(s)
Action Potentials/physiology , Caveolin 3/metabolism , Cerebellum/physiology , Neurons/physiology , Shal Potassium Channels/metabolism , Animals , Caveolin 3/genetics , Cerebellum/cytology , Cerebellum/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Shal Potassium Channels/genetics , Synapses/metabolism , Synapses/physiology
20.
Pflugers Arch ; 466(4): 661-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24413887

ABSTRACT

The electrical output of neurons relies critically on voltage- and calcium-gated ion channels. The traditional view of ion channels is that they operate independently of each other in the plasma membrane in a manner that could be predicted according to biophysical characteristics of the isolated current. However, there is increasing evidence that channels interact with each other not just functionally but also physically. This is exemplified in the case of Cav3 T-type calcium channels, where new work indicates the ability to form signaling complexes with different types of calcium-gated and even voltage-gated potassium channels. The formation of a Cav3-K complex provides the calcium source required to activate KCa1.1 or KCa3.1 channels and, furthermore, to bestow a calcium-dependent regulation of Kv4 channels via associated KChIP proteins. Here, we review these interactions and discuss their significance in the context of neuronal firing properties.


Subject(s)
Calcium Channels, T-Type/physiology , Cell Membrane/physiology , Neurons/physiology , Potassium Channels/physiology , Animals , Humans , Membrane Potentials/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...