1
|
Gupta S, Gee MM, Newton AJH, Kuttippurathu L, Moss A, Tompkins JD, Schwaber JS, Vadigepalli R, Lytton WW. Biophysical modelling of intrinsic cardiac nervous system neuronal electrophysiology based on single-cell transcriptomics. J Physiol 2025; 603:2119-2138. [PMID: 40077928 PMCID: PMC11955872 DOI: 10.1113/jp287595] [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: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
The intrinsic cardiac nervous system (ICNS), termed as the heart's 'little brain', is the final point of neural regulation of cardiac function. Studying the dynamic behaviour of these ICNS neurons via multiscale neuronal computer models has been limited by the sparsity of electrophysiological data. We developed and analysed a computational library of neuronal electrophysiological models based on single neuron transcriptomic data obtained from ICNS neurons. Each neuronal genotype was characterized by a unique combination of ion channels identified from the transcriptomic data, using a cycle threshold cutoff that ensured the electrical excitability of the neuronal models. The parameters of the ion channel models were grounded based on passive properties (resting membrane potential, input impedance and rheobase) to avoid biasing the dynamic behaviour of the model. Consistent with experimental observations, the emergent model dynamics showed phasic activity in response to the current clamp stimulus in a majority of neuronal genotypes (61%). Additionally, 24% of the ICNS neurons showed a tonic response, 11% were phasic-to-tonic with increasing current stimulation and 3% showed tonic-to-phasic behaviour. The computational approach and the library of models bridge the gap between widely available molecular-level gene expression and sparse cellular-level electrophysiology for studying the functional role of the ICNS in cardiac regulation and pathology. KEY POINTS: Computational models were developed of neuron electrophysiology from single-cell transcriptomic data from neurons in the heart's 'little brain': the intrinsic cardiac nervous system. The single-cell transcriptomic data were thresholded to select the ion channel combinations in each neuronal model. The library of neuronal models was constrained by the passive electrical properties of the neurons and predicted a distribution of phasic and tonic responses that aligns with experimental observations. The ratios of model-predicted conductance values are correlated with the gene expression ratios from transcriptomic data. These neuron models are a first step towards connecting single-cell transcriptomic data to dynamic, predictive physiology-based models.
Collapse
Affiliation(s)
- Suranjana Gupta
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNYUSA
| | - Michelle M. Gee
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDEUSA
| | - Adam J. H. Newton
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNYUSA
| | - Lakshmi Kuttippurathu
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
| | - Alison Moss
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
| | - John D. Tompkins
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| | - James S. Schwaber
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDEUSA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDEUSA
| | - William W. Lytton
- Department of Physiology and PharmacologySUNY Downstate Health Sciences UniversityBrooklynNYUSA
- Department of NeurologyKings County HospitalBrooklynNYUSA
| |
Collapse
|
2
|
Malloy C, Ahern M, Lin L, Hoffman DA. Neuronal Roles of the Multifunctional Protein Dipeptidyl Peptidase-like 6 (DPP6). Int J Mol Sci 2022; 23:9184. [PMID: 36012450 PMCID: PMC9409431 DOI: 10.3390/ijms23169184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The concerted action of voltage-gated ion channels in the brain is fundamental in controlling neuronal physiology and circuit function. Ion channels often associate in multi-protein complexes together with auxiliary subunits, which can strongly influence channel expression and function and, therefore, neuronal computation. One such auxiliary subunit that displays prominent expression in multiple brain regions is the Dipeptidyl aminopeptidase-like protein 6 (DPP6). This protein associates with A-type K+ channels to control their cellular distribution and gating properties. Intriguingly, DPP6 has been found to be multifunctional with an additional, independent role in synapse formation and maintenance. Here, we feature the role of DPP6 in regulating neuronal function in the context of its modulation of A-type K+ channels as well as its independent involvement in synaptic development. The prevalence of DPP6 in these processes underscores its importance in brain function, and recent work has identified that its dysfunction is associated with host of neurological disorders. We provide a brief overview of these and discuss research directions currently underway to advance our understanding of the contribution of DPP6 to their etiology.
Collapse
Affiliation(s)
| | | | | | - Dax A. Hoffman
- Molecular Neurophysiology and Biophysics Section, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Lamothe SM, Kurata HT. Slc7a5 alters Kvβ-mediated regulation of Kv1.2. J Gen Physiol 2021; 152:151687. [PMID: 32311044 PMCID: PMC7335012 DOI: 10.1085/jgp.201912524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/22/2020] [Accepted: 03/13/2020] [Indexed: 01/15/2023] Open
Abstract
The voltage-gated potassium channel Kv1.2 plays a pivotal role in neuronal excitability and is regulated by a variety of known and unknown extrinsic factors. The canonical accessory subunit of Kv1.2, Kvβ, promotes N-type inactivation and cell surface expression of the channel. We recently reported that a neutral amino acid transporter, Slc7a5, alters the function and expression of Kv1.2. In the current study, we investigated the effects of Slc7a5 on Kv1.2 in the presence of Kvβ1.2 subunits. We observed that Slc7a5-induced suppression of Kv1.2 current and protein expression was attenuated with cotransfection of Kvβ1.2. However, gating effects mediated by Slc7a5, including disinhibition and a hyperpolarizing shift in channel activation, were observed together with Kvβ-mediated inactivation, indicating convergent regulation of Kv1.2 by both regulatory proteins. Slc7a5 influenced several properties of Kvβ-induced inactivation of Kv1.2, including accelerated inactivation, a hyperpolarizing shift and greater extent of steady-state inactivation, and delayed recovery from inactivation. These modified inactivation properties were also apparent in altered deactivation of the Kv1.2/Kvβ/Slc7a5 channel complex. Taken together, these findings illustrate a functional interaction arising from simultaneous regulation of Kv1.2 by Kvβ and Slc7a5, leading to powerful effects on Kv1.2 expression, gating, and overall channel function.
Collapse
Affiliation(s)
- Shawn M Lamothe
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Harley T Kurata
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
Deutsch M, Günther A, Lerchundi R, Rose CR, Balfanz S, Baumann A. AAV-Mediated CRISPRi and RNAi Based Gene Silencing in Mouse Hippocampal Neurons. Cells 2021; 10:324. [PMID: 33557342 PMCID: PMC7915209 DOI: 10.3390/cells10020324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 12/12/2022] Open
Abstract
Uncovering the physiological role of individual proteins that are part of the intricate process of cellular signaling is often a complex and challenging task. A straightforward strategy of studying a protein's function is by manipulating the expression rate of its gene. In recent years, the Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/Cas9-based technology was established as a powerful gene-editing tool for generating sequence specific changes in proliferating cells. However, obtaining homogeneous populations of transgenic post-mitotic neurons by CRISPR/Cas9 turned out to be challenging. These constraints can be partially overcome by CRISPR interference (CRISPRi), which mediates the inhibition of gene expression by competing with the transcription machinery for promoter binding and, thus, transcription initiation. Notably, CRISPR/Cas is only one of several described approaches for the manipulation of gene expression. Here, we targeted neurons with recombinant Adeno-associated viruses to induce either CRISPRi or RNA interference (RNAi), a well-established method for impairing de novo protein biosynthesis by using cellular regulatory mechanisms that induce the degradation of pre-existing mRNA. We specifically targeted hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels, which are widely expressed in neuronal tissues and play essential physiological roles in maintaining biophysical characteristics in neurons. Both of the strategies reduced the expression levels of three HCN isoforms (HCN1, 2, and 4) with high specificity. Furthermore, detailed analysis revealed that the knock-down of just a single HCN isoform (HCN4) in hippocampal neurons did not affect basic electrical parameters of transduced neurons, whereas substantial changes emerged in HCN-current specific properties.
Collapse
Affiliation(s)
- Matthias Deutsch
- Forschungszentrum Jülich, Institute of Biological Information Processing, IBI-1, Leo-Brandt-Straße, 52428 Jülich, Germany; (M.D.); (S.B.)
- Department of Biology, University of California, San Diego, La Jolla, CA 92083, USA
| | - Anne Günther
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany;
| | - Rodrigo Lerchundi
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany; (R.L.); (C.R.R.)
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany; (R.L.); (C.R.R.)
| | - Sabine Balfanz
- Forschungszentrum Jülich, Institute of Biological Information Processing, IBI-1, Leo-Brandt-Straße, 52428 Jülich, Germany; (M.D.); (S.B.)
| | - Arnd Baumann
- Forschungszentrum Jülich, Institute of Biological Information Processing, IBI-1, Leo-Brandt-Straße, 52428 Jülich, Germany; (M.D.); (S.B.)
| |
Collapse
|
5
|
Noh W, Pak S, Choi G, Yang S, Yang S. Transient Potassium Channels: Therapeutic Targets for Brain Disorders. Front Cell Neurosci 2019; 13:265. [PMID: 31263403 PMCID: PMC6585177 DOI: 10.3389/fncel.2019.00265] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/28/2019] [Indexed: 01/04/2023] Open
Abstract
Transient potassium current channels (IA channels), which are expressed in most brain areas, have a central role in modulating feedforward and feedback inhibition along the dendroaxonic axis. Loss of the modulatory channels is tightly associated with a number of brain diseases such as Alzheimer’s disease, epilepsy, fragile X syndrome (FXS), Parkinson’s disease, chronic pain, tinnitus, and ataxia. However, the functional significance of IA channels in these diseases has so far been underestimated. In this review, we discuss the distribution and function of IA channels. Particularly, we posit that downregulation of IA channels results in neuronal (mostly dendritic) hyperexcitability accompanied by the imbalanced excitation and inhibition ratio in the brain’s networks, eventually causing the brain diseases. Finally, we propose a potential therapeutic target: the enhanced action of IA channels to counteract Ca2+-permeable channels including NMDA receptors could be harnessed to restore dendritic excitability, leading to a balanced neuronal state.
Collapse
Affiliation(s)
- Wonjun Noh
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| | - Sojeong Pak
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Geunho Choi
- Department of Computer Science and Engineering, Incheon National University, Incheon, South Korea
| | - Sungchil Yang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Sunggu Yang
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| |
Collapse
|
6
|
Zobeiri M, Chaudhary R, Datunashvili M, Heuermann RJ, Lüttjohann A, Narayanan V, Balfanz S, Meuth P, Chetkovich DM, Pape HC, Baumann A, van Luijtelaar G, Budde T. Modulation of thalamocortical oscillations by TRIP8b, an auxiliary subunit for HCN channels. Brain Struct Funct 2018; 223:1537-1564. [PMID: 29168010 PMCID: PMC5869905 DOI: 10.1007/s00429-017-1559-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels have important functions in controlling neuronal excitability and generating rhythmic oscillatory activity. The role of tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b) in regulation of hyperpolarization-activated inward current, I h, in the thalamocortical system and its functional relevance for the physiological thalamocortical oscillations were investigated. A significant decrease in I h current density, in both thalamocortical relay (TC) and cortical pyramidal neurons was found in TRIP8b-deficient mice (TRIP8b-/-). In addition basal cAMP levels in the brain were found to be decreased while the availability of the fast transient A-type K+ current, I A, in TC neurons was increased. These changes were associated with alterations in intrinsic properties and firing patterns of TC neurons, as well as intrathalamic and thalamocortical network oscillations, revealing a significant increase in slow oscillations in the delta frequency range (0.5-4 Hz) during episodes of active-wakefulness. In addition, absence of TRIP8b suppresses the normal desynchronization response of the EEG during the switch from slow-wave sleep to wakefulness. It is concluded that TRIP8b is necessary for the modulation of physiological thalamocortical oscillations due to its direct effect on HCN channel expression in thalamus and cortex and that mechanisms related to reduced cAMP signaling may contribute to the present findings.
Collapse
Affiliation(s)
- Mehrnoush Zobeiri
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany.
| | - Rahul Chaudhary
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany
| | - Maia Datunashvili
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany
| | - Robert J Heuermann
- Davee Department of Neurology and Clinical Neurosciences and Department of Physiology, Feinberg School of Medicine, Northwestern University, 60611Chicago, USA
| | - Annika Lüttjohann
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany
| | - Venu Narayanan
- Department of Neurology and Institute of Translational Neurology, Westfälische Wilhelms-Universität, 48149, Münster, Germany
| | - Sabine Balfanz
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Patrick Meuth
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany
| | - Dane M Chetkovich
- Davee Department of Neurology and Clinical Neurosciences and Department of Physiology, Feinberg School of Medicine, Northwestern University, 60611Chicago, USA
| | - Hans-Christian Pape
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany
| | - Arnd Baumann
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | | | - Thomas Budde
- Institut für Physiologie I, Westfälische Wilhelms-Universität, 48149, Münster, Germany.
| |
Collapse
|
7
|
A novel role of the antitumor agent tricyclodecan-9-yl-xanthogenate as an open channel blocker of KCNQ1/KCNE1. Eur J Pharmacol 2018; 824:99-107. [DOI: 10.1016/j.ejphar.2018.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/26/2022]
|
8
|
Wolff M, Czorlich P, Nagaraj C, Schnöbel-Ehehalt R, Li Y, Kwapiszewska G, Olschewski H, Heschl S, Olschewski A. Amitriptyline and carbamazepine utilize voltage-gated ion channel suppression to impair excitability of sensory dorsal horn neurons in thin tissue slice: An in vitro study. Neurosci Res 2016; 109:16-27. [DOI: 10.1016/j.neures.2016.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 11/27/2022]
|
9
|
Rainnie DG, Hazra R, Dabrowska J, Guo JD, Li CC, Dewitt S, Muly EC. Distribution and functional expression of Kv4 family α subunits and associated KChIP β subunits in the bed nucleus of the stria terminalis. J Comp Neurol 2014; 522:609-25. [PMID: 24037673 DOI: 10.1002/cne.23435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 06/28/2013] [Accepted: 07/02/2013] [Indexed: 12/22/2022]
Abstract
Regulation of BNSTALG neuronal firing activity is tightly regulated by the opposing actions of the fast outward potassium current, IA , mediated by α subunits of the Kv4 family of ion channels, and the transient inward calcium current, IT . Together, these channels play a critical role in regulating the latency to action potential onset, duration, and frequency, as well as dendritic back-propagation and synaptic plasticity. Previously we have shown that Type I-III BNSTALG neurons express mRNA transcripts for each of the Kv4 α subunits. However, the biophysical properties of native IA channels are critically dependent on the formation of macromolecular complexes of Kv4 channels with a family of chaperone proteins, the potassium channel-interacting proteins (KChIP1-4). Here we used a multidisciplinary approach to investigate the expression and function of Kv4 channels and KChIPs in neurons of the rat BNSTALG . Using immunofluorescence we demonstrated the pattern of localization of Kv4.2, Kv4.3, and KChIP1-4 proteins in the BNSTALG . Moreover, our single-cell reverse-transcription polymerase chain reaction (scRT-PCR) studies revealed that mRNA transcripts for Kv4.2, Kv4.3, and all four KChIPs were differentially expressed in Type I-III BNSTALG neurons. Furthermore, immunoelectron microscopy revealed that Kv4.2 and Kv4.3 channels were primarily localized to the dendrites and spines of BNSTALG neurons, and are thus ideally situated to modulate synaptic transmission. Consistent with this observation, in vitro patch clamp recordings showed that reducing postsynaptic IA in these neurons lowered the threshold for long-term potentiation (LTP) induction. These results are discussed in relation to potential modulation of IA channels by chronic stress.
Collapse
Affiliation(s)
- Donald G Rainnie
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia; Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, Georgia
| | | | | | | | | | | | | |
Collapse
|
10
|
Kuczewski N, Fourcaud-Trocmé N, Savigner A, Thevenet M, Aimé P, Garcia S, Duchamp-Viret P, Palouzier-Paulignan B. Insulin modulates network activity in olfactory bulb slices: impact on odour processing. J Physiol 2014; 592:2751-69. [PMID: 24710056 DOI: 10.1113/jphysiol.2013.269639] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Odour perception depends closely on nutritional status, in animals as in humans. Insulin, the principal anorectic hormone, appears to be one of the major candidates for ensuring the link between olfactory abilities and nutritional status, by modifying processing in the olfactory bulb (OB), one of its main central targets. The present study investigates whether and how insulin can act in OB, by evaluating its action on the main output neurons activities, mitral cells (MCs), in acute rat OB slices. Insulin was found to act at two OB network levels: (1) on MCs, by increasing their excitability, probably by inhibiting two voltage-gated potassium (K(+)) channels; (2) on interneurons by modifying the GABAergic and on glutamatergic synaptic activity impinging on MCs, mainly reducing them. Insulin also altered the olfactory nerve (ON)-evoked excitatory postsynaptic currents in 60% of MCs. Insulin decreased or increased the ON-evoked responses in equal proportion and the direction of its effect depended on the initial neuron ON-evoked firing rate. Indeed, insulin tended to decrease the high and to increase the low ON-evoked firing rates, thereby reducing inter-MC response firing variability. Therefore, the effects of insulin on the evoked firing rates were not carried out indiscriminately in the MC population. By constructing a mathematical model, the impact of insulin complex effects on OB was assessed at the population activity level. The model shows that the reduction of variability across cells could affect MC detection and discrimination abilities, mainly by decreasing and, less frequently, increasing them, depending on odour quality. Thus, as previously proposed, this differential action of insulin on MCs across odours would allow this hormone to put the olfactory function under feeding signal control, given the discerning valence of an odour as a function of nutritional status.
Collapse
Affiliation(s)
- Nicola Kuczewski
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Nicolas Fourcaud-Trocmé
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Agnès Savigner
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Marc Thevenet
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Pascaline Aimé
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Samuel Garcia
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Patricia Duchamp-Viret
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| | - Brigitte Palouzier-Paulignan
- Université Lyon1, Centre de Recherche en Neurosciences de Lyon INSERM U1028/CNRS UMR5292 - Equipe Olfaction: Du codage à la mémoire, F-69366, Lyon, France
| |
Collapse
|
11
|
Yang YS, Kim KD, Eun SY, Jung SC. Roles of somatic A-type K(+) channels in the synaptic plasticity of hippocampal neurons. Neurosci Bull 2014; 30:505-14. [PMID: 24526657 DOI: 10.1007/s12264-013-1399-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 07/19/2013] [Indexed: 01/11/2023] Open
Abstract
In the mammalian brain, information encoding and storage have been explained by revealing the cellular and molecular mechanisms of synaptic plasticity at various levels in the central nervous system, including the hippocampus and the cerebral cortices. The modulatory mechanisms of synaptic excitability that are correlated with neuronal tasks are fundamental factors for synaptic plasticity, and they are dependent on intracellular Ca(2+)-mediated signaling. In the present review, the A-type K(+) (IA) channel, one of the voltage-dependent cation channels, is considered as a key player in the modulation of Ca(2+) influx through synaptic NMDA receptors and their correlated signaling pathways. The cellular functions of IA channels indicate that they possibly play as integral parts of synaptic and somatic complexes, completing the initiation and stabilization of memory.
Collapse
Affiliation(s)
- Yoon-Sil Yang
- Department of Physiology, School of Medicine, Jeju National University, Jeju, 690756, Republic of Korea
| | | | | | | |
Collapse
|
12
|
Lovell PV, Carleton JB, Mello CV. Genomics analysis of potassium channel genes in songbirds reveals molecular specializations of brain circuits for the maintenance and production of learned vocalizations. BMC Genomics 2013; 14:470. [PMID: 23845108 PMCID: PMC3711925 DOI: 10.1186/1471-2164-14-470] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 06/19/2013] [Indexed: 02/08/2023] Open
Abstract
Background A fundamental question in molecular neurobiology is how genes that determine basic neuronal properties shape the functional organization of brain circuits underlying complex learned behaviors. Given the growing availability of complete vertebrate genomes, comparative genomics represents a promising approach to address this question. Here we used genomics and molecular approaches to study how ion channel genes influence the properties of the brain circuitry that regulates birdsong, a learned vocal behavior with important similarities to human speech acquisition. We focused on potassium (K-)Channels, which are major determinants of neuronal cell excitability. Starting with the human gene set of K-Channels, we used cross-species mRNA/protein alignments, and syntenic analysis to define the full complement of orthologs, paralogs, allelic variants, as well as novel loci not previously predicted in the genome of zebra finch (Taeniopygia guttata). We also compared protein coding domains in chicken and zebra finch orthologs to identify genes under positive selective pressure, and those that contained lineage-specific insertions/deletions in functional domains. Finally, we conducted comprehensive in situ hybridizations to determine the extent of brain expression, and identify K-Channel gene enrichments in nuclei of the avian song system. Results We identified 107 K-Channel finch genes, including 6 novel genes common to non-mammalian vertebrate lineages. Twenty human genes are absent in songbirds, birds, or sauropsids, or unique to mammals, suggesting K-Channel properties may be lineage-specific. We also identified specific family members with insertions/deletions and/or high dN/dS ratios compared to chicken, a non-vocal learner. In situ hybridization revealed that while most K-Channel genes are broadly expressed in the brain, a subset is selectively expressed in song nuclei, representing molecular specializations of the vocal circuitry. Conclusions Together, these findings shed new light on genes that may regulate biophysical and excitable properties of the song circuitry, identify potential targets for the manipulation of the song system, and reveal genomic specializations that may relate to the emergence of vocal learning and associated brain areas in birds.
Collapse
|
13
|
Yasuda T, Cuny H, Adams DJ. Kv3.1 channels stimulate adult neural precursor cell proliferation and neuronal differentiation. J Physiol 2013; 591:2579-91. [PMID: 23478135 DOI: 10.1113/jphysiol.2012.249151] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Adult neural stem/precursor cells (NPCs) play a pivotal role in neuronal plasticity throughout life. Among ion channels identified in adult NPCs, voltage-gated delayed rectifier K(+) (KDR) channels are dominantly expressed. However, the KDR channel subtype and its physiological role are still undefined. We used real-time quantitative RT-PCR and gene knockdown techniques to identify a major functional KDR channel subtype in adult NPCs. Dominant mRNA expression of Kv3.1, a high voltage-gated KDR channel, was quantitatively confirmed. Kv3.1 gene knockdown with specific small interfering RNAs (siRNA) for Kv3.1 significantly inhibited Kv3.1 mRNA expression by 63.9% (P < 0.001) and KDR channel currents by 52.2% (P < 0.001). This indicates that Kv3.1 is the subtype responsible for producing KDR channel outward currents. Resting membrane properties, such as resting membrane potential, of NPCs were not affected by Kv3.1 expression. Kv3.1 knockdown with 300 nm siRNA inhibited NPC growth (increase in cell numbers) by 52.9% (P < 0.01). This inhibition was attributed to decreased cell proliferation, not increased cell apoptosis. We also established a convenient in vitro imaging assay system to evaluate NPC differentiation using NPCs from doublecortin-green fluorescent protein transgenic mice. Kv3.1 knockdown also significantly reduced neuronal differentiation by 31.4% (P < 0.01). We have demonstrated that Kv3.1 is a dominant functional KDR channel subtype expressed in adult NPCs and plays key roles in NPC proliferation and neuronal lineage commitment during differentiation.
Collapse
Affiliation(s)
- Takahiro Yasuda
- Center for Clinical Research, School of Medicine, Kobe University Hospital, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | | | | |
Collapse
|
14
|
Abstract
Potassium (K(+)) channels are essential to neuronal signaling and survival. Here we show that these proteins are targets of reactive oxygen species in mammalian brain and that their oxidation contributes to neuropathy. Thus, the KCNB1 (Kv2.1) channel, which is abundantly expressed in cortex and hippocampus, formed oligomers upon exposure to oxidizing agents. These oligomers were ∼10-fold more abundant in the brain of old than young mice. Oxidant-induced oligomerization of wild-type KCNB1 enhanced apoptosis in neuronal cells subject to oxidative insults. Consequently, a KCNB1 variant resistant to oxidation, obtained by mutating a conserved cysteine to alanine, (C73A), was neuroprotective. The fact that oxidation of KCNB1 is toxic, argues that this mechanism may contribute to neuropathy in conditions characterized by high levels of oxidative stress, such as Alzheimer's disease (AD). Accordingly, oxidation of KCNB1 channels was exacerbated in the brain of a triple transgenic mouse model of AD (3xTg-AD). The C73A variant protected neuronal cells from apoptosis induced by incubation with β-amyloid peptide (Aβ(1-42)). In an invertebrate model (Caenorhabditis elegans) that mimics aspects of AD, a C73A-KCNB1 homolog (C113S-KVS-1) protected specific neurons from apoptotic death induced by ectopic expression of human Aβ(1-42). Together, these data underscore a novel mechanism of toxicity in neurodegenerative disease.
Collapse
|
15
|
Garrido-Sanabria ER, Perez-Cordova MG, Colom LV. Differential expression of voltage-gated K+ currents in medial septum/diagonal band complex neurons exhibiting distinct firing phenotypes. Neurosci Res 2011; 70:361-9. [PMID: 21624401 PMCID: PMC3150140 DOI: 10.1016/j.neures.2011.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 04/22/2011] [Accepted: 05/09/2011] [Indexed: 01/28/2023]
Abstract
The medial septum/diagonal band complex (MSDB) controls hippocampal excitability, rhythms and plastic processes. Medial septal neuronal populations display heterogeneous firing patterns. In addition, some of these populations degenerate during age-related disorders (e.g. cholinergic neurons). Thus, it is particularly important to examine the intrinsic properties of theses neurons in order to create new agents that effectively modulate hippocampal excitability and enhance memory processes. Here, we have examined the properties of voltage-gated, K(+) currents in electrophysiologically-identified neurons. These neurons were taken from young rat brain slices containing the MS/DB complex. Whole-cell, patch recordings of outward currents were obtained from slow firing, fast-spiking, regular-firing and burst-firing neurons. Slow firing neurons showed depolarization-activated K(+) current peaks and densities larger than in other neuronal subtypes. Slow firing total current exhibited an inactivating A-type current component that activates at subthreshold depolarization and was reliably blocked by high concentrations of 4-AP. In addition, slow firing neurons expressed a low-threshold delayed rectifier K(+) current component with slow inactivation and intermediate sensitivity to tetraethylammonium. Fast-spiking neurons exhibited the smaller I(K) and I(A) current densities. Burst and regular firing neurons displayed an intermediate firing phenotype with I(K) and I(A) current densities that were larger than the ones observed in fast-spiking neurons but smaller than the ones observed in slow-firing neurons. In addition, the prevalence of each current differed among electrophysiological groups with slow firing and regular firing neurons expressing mostly I(A) and fast spiking and bursting neurons exhibiting mostly delayer rectifier K(+) currents with only minimal contributions of the I(A). The pharmacological or genetic modulations of these currents constitute an important target for the treatment of age-related disorders.
Collapse
Affiliation(s)
- Emilio R. Garrido-Sanabria
- Department of Biological Sciences, The Center for Biomedical Studies, The University of Texas at Brownsville, 80 Fort Brown, Brownsville, Texas 78520
| | - Miriam G. Perez-Cordova
- Department of Biological Sciences, The Center for Biomedical Studies, The University of Texas at Brownsville, 80 Fort Brown, Brownsville, Texas 78520
| | - Luis V. Colom
- Department of Biological Sciences, The Center for Biomedical Studies, The University of Texas at Brownsville, 80 Fort Brown, Brownsville, Texas 78520
| |
Collapse
|
16
|
Action potentials in primary osteoblasts and in the MG-63 osteoblast-like cell line. J Bioenerg Biomembr 2011; 43:311-22. [PMID: 21523406 DOI: 10.1007/s10863-011-9354-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Accepted: 04/03/2011] [Indexed: 01/25/2023]
Abstract
Whole-cell patch-clamp analysis revealed a resting membrane potential of -60 mV in primary osteoblasts and in the MG-63 osteoblast-like cells. Depolarization-induced action potentials were characterized by duration of 60 ms, a minimal peak-to-peak distance of 180 ms, a threshold value of -20 mV and a repolarization between the spikes to -45 mV. Expressed channels were characterized by application of voltage pulses between -150 mV and 90 mV in 10 mV steps, from a holding potential of -40 mV. Voltages below -60 mV induced an inward current. Depolarizing voltages above -30 mV evoked two currents: (a) a fast activated and inactivated inward current at voltages between -30 and 30 mV, and (b) a delayed-activated outward current that was induced by voltages above -30 mV. Electrophysiological and pharmacological parameters indicated that hyperpolarization activated strongly rectifying K(+) (K(ir)) channels, whereas depolarization activated tetrodotoxin sensitive voltage gated Na(+) (Na(v)) channels as well as delayed, slowly activated, non-inactivating, and tetraethylammonium sensitive voltage gated K(+) (K(v)) channels. In addition, RT-PCR showed expression of Na(v)1.3, Na(v)1.4, Na(v)1.5, Na(v)1.6, Na(v)1.7, and K(ir)2.1, K(ir)2.3, and K(ir)2.4 as well as K(v)2.1. We conclude that osteoblasts express channels that allow firing of action potentials.
Collapse
|
17
|
Hazra R, Guo JD, Ryan SJ, Jasnow AM, Dabrowska J, Rainnie DG. A transcriptomic analysis of type I-III neurons in the bed nucleus of the stria terminalis. Mol Cell Neurosci 2011; 46:699-709. [PMID: 21310239 PMCID: PMC3066260 DOI: 10.1016/j.mcn.2011.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 01/14/2011] [Accepted: 01/31/2011] [Indexed: 10/18/2022] Open
Abstract
The activity of neurons in the anterolateral cell group of the bed nucleus of the stria terminalis (BNST(ALG)) plays a critical role in anxiety- and stress-related behaviors. Histochemical studies have suggested that multiple distinct neuronal phenotypes exist in the BNST(ALG). Consistent with this observation, the physiological properties of BNST(ALG) neurons are also heterogeneous, and three distinct cell types can be defined (Types I-III) based primarily on their expression of four key membrane currents, namely I(h), I(A), I(T), and I(K(IR)). Significantly, all four channels are multimeric proteins and can comprise of more than one pore-forming α subunit. Hence, differential expression of α subunits may further diversify the neuronal population. However, nothing is known about the relative expression of these ion channel α subunits in BNST(ALG) neurons. We have addressed this lacuna by combining whole-cell patch-clamp recording together with single-cell reverse transcriptase polymerase chain reaction (scRT-PCR) to assess the mRNA transcript expression for each of the subunits for the four key ion channels in Type I-III neurons of the BNST(ALG.) Here, cytosolic mRNA from single neurons was probed for the expression of transcripts for each of the α subunits of I(h) (HCN1-HCN4), I(T) (Ca(v)3.1-Ca(v)3.3), I(A) (K(v)1.4, K(v)3.4, K(v)4.1-K(v) 4.3) and I(K(IR)) (Kir2.1-Kir2.4). An unbiased hierarchical cluster analysis followed by discriminant function analysis revealed that a positive correlation exists between the physiological and genetic phenotype of BNST(ALG) neurons. Thus, the analysis segregated BNST(ALG) neurons into 3 distinct groups, based on their α subunit mRNA expression profile, which positively correlated with our existing electrophysiological classification (Types I-III). Furthermore, analysis of mRNA transcript expression in Type I-Type III neurons suggested that, whereas Type I and III neurons appear to represent genetically homologous cell populations, Type II neurons may be further subdivided into three genetically distinct subgroups. These data not only validate our original classification scheme, but further refine the classification at the molecular level, and thus identifies novel targets for potential disruption and/or pharmacotherapeutic intervention in stress-related anxiety-like behaviors.
Collapse
Affiliation(s)
- Rimi Hazra
- Department of Psychiatry and Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
| | - Ji-Dong Guo
- Department of Psychiatry and Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
| | - Steven J Ryan
- Graduate program in Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
| | - Aaron M Jasnow
- Department of Psychiatry and Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
| | - Joanna Dabrowska
- Department of Psychiatry and Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
| | - Donald G Rainnie
- Department of Psychiatry and Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329
| |
Collapse
|
18
|
Kanyshkova T, Broicher T, Meuth SG, Pape HC, Budde T. A-type K+ currents in intralaminar thalamocortical relay neurons. Pflugers Arch 2011; 461:545-56. [PMID: 21437601 DOI: 10.1007/s00424-011-0953-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 02/22/2011] [Indexed: 10/18/2022]
Abstract
Transient A-type K+ currents (I(A)) are known to influence the firing pattern of a number of thalamic cell types, but have not been investigated in intralaminar thalamocortical (TC) relay neurons yet. We therefore combined whole-cell patch-clamp techniques, PCR analysis, and immunohistochemistry to investigate the voltage-dependent and pharmacological properties of I(A) and to determine its molecular basis in TC neurons from the centrolateral, paracentral, and centromedial thalamic nuclei. I(A) revealed half-maximal (V (h)) activation and inactivation at about -17 and -67 mV, respectively. At a concentration of 5-10 mM 4-aminopyridine (4-AP) completely blocked I(A). Furthermore, I(A) was nearly unaffected by two sea anemone toxins (blood depressing substances 1 and 2, BDS1 and BDS2; 6-8% block at a concentration of 1 μM) but strongly sensitive to the K(V)4 channel blocker Heteropoda venatoria toxin 2 (HpTx2; about 45% block at a concentration of 5 μM). PCR screening revealed the expression of K(V)4.1-4.3, with strongest expression for K(V)4.2 and weak expression for K(V)4.1. Accordingly K(V)4.1 was not detected in immunohistochemical staining. Furthermore, K(V)4.2 and K(V)4.3 revealed mainly dendritic and somatic staining, respectively. Together with current clamp recordings, these findings point to a scenario where the fast transient I(A) in intralaminar TC neurons has a depolarized threshold at potentials negative to -50 mV, is substantially generated by K(V)4.2 and K(V)4.3 channels, allows prominent burst firing at hyperpolarized potentials, prevents the generation of high-threshold potentials, generates a delayed onset of firing at more depolarized potentials, and allows fast tonic firing.
Collapse
Affiliation(s)
- Tatyana Kanyshkova
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, 48149, Münster, Germany
| | | | | | | | | |
Collapse
|
19
|
Luján R. Organisation of potassium channels on the neuronal surface. J Chem Neuroanat 2010; 40:1-20. [PMID: 20338235 DOI: 10.1016/j.jchemneu.2010.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 03/10/2010] [Accepted: 03/11/2010] [Indexed: 11/30/2022]
Abstract
Potassium channels are a family of ion channels that govern the intrinsic electrical properties of neurons in the brain. Molecular cloning has revealed over 100 genes encoding the pore-forming alpha subunits of potassium channels in mammals, making them the most diverse subset of ion channels. Multiplicity in this ion channel family is further generated through alternative splicing. The precise location of potassium channels along the dendro-somato-axonic surface of the neurons is an important factor in determining its functional impact. Today, it is widely accepted that potassium channels can be located at any subcellular compartment on the neuronal surface, at synaptic and extrasynaptic sites, from somata to dendritic shafts, dendritic spines, axons or axon terminals. However, they are not evenly distributed on the neuronal surface and depending on the potassium channel subtype, are instead concentrated at different compartments. This selective localization of ion channels to specific neuronal compartments has many different functional implications. One factor necessary to understand the role of potassium channels in neuronal function is to unravel their specialized distribution and subcellular localization within a cell, and this can only be achieved by electron microscopy. In this review, I summarize anatomical findings, describing their distribution in the central nervous system. The distinct regional, cellular and subcellular distribution of potassium channels in the brain will be discussed in view of their possible functional implications.
Collapse
Affiliation(s)
- Rafael Luján
- Departamento de Ciencias Médicas, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Campus Biosanitario, C/Almansa 14, 02006 Albacete, Spain.
| |
Collapse
|
20
|
Shen QJ, Zhao YM, Cao DX, Wang XL. Contribution of Kv channel subunits to glutamate-induced apoptosis in cultured rat hippocampal neurons. J Neurosci Res 2009; 87:3153-60. [PMID: 19472219 DOI: 10.1002/jnr.22136] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Potassium channel dysfunction has been implicated in apoptosis in many pathological conditions. However, which Kv channel subunit is involved in glutamate-induced apoptosis remains unknown. In this study, the contributions of nine Kv alpha and three Kv beta subunits to glutamate-induced hippocampal neuronal apoptosis were investigated. Results showed that neuronal apoptosis was not obvious with 12 hr incubation of glutamate but increased markedly after 18 hr, which was attenuated by the Kv channel blocker TEA. Among all the Kv subunits investigated, gene and protein expression of Kv2.1 increased significantly before the appearance of neuronal apoptosis, whereas the Kv1.1 mRNA level decreased quickly, and protein expression was reduced gradually after the insult. Seven other Kv alpha subunits and three Kv beta subunits were not obviously affected over time. In addition, Kv1.1 overexpression could reduce glutamate-induced hippocampal neuronal apoptosis. Therefore, the alterations of Kv1.1 and Kv2.1 might contribute to glutamate-induced toxicity in hippocampal neurons. These findings suggest that these two Kv channel subunits may represent potential therapeutic targets for neuropathological conditions in which glutamate-induced toxicity is thought to contribute to neuronal dysfunction.
Collapse
Affiliation(s)
- Qi-Jun Shen
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
21
|
McNicholas K, Chen T, Abbott CA. Dipeptidyl peptidase (DP) 6 and DP10: novel brain proteins implicated in human health and disease. Clin Chem Lab Med 2009; 47:262-7. [PMID: 19676137 DOI: 10.1515/cclm.2009.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Dipeptidyl peptidase (DP) 6 and DP10 are non-enzyme members of the dipeptidyl peptidase IV family, which includes fibroblast activation protein, DP8, and DP9. DP6 and DP10 proteins have been shown to be critical components of voltage-gated potassium (Kv) channels important in determining cellular excitability. The aim of this paper was to review the research to date on DP6 and DP10 structure, expression, and functions. To date, the protein region responsible for modulating Kv4 channels has not been conclusively identified and the significance of the splice variants has not been resolved. Resolution of these issues will improve our overall knowledge of DP6 and DP10 and lead to a better understanding of their role in diseases, such as asthma and Alzheimer's disease.
Collapse
Affiliation(s)
- Kym McNicholas
- School of Biological Sciences, Flinders University, Adelaide, South Australia, Australia
| | | | | |
Collapse
|
22
|
Bocksteins E, Raes AL, Van de Vijver G, Bruyns T, Van Bogaert PP, Snyders DJ. Kv2.1 and silent Kv subunits underlie the delayed rectifier K+ current in cultured small mouse DRG neurons. Am J Physiol Cell Physiol 2009; 296:C1271-8. [PMID: 19357235 DOI: 10.1152/ajpcell.00088.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Silent voltage-gated K(+) (K(v)) subunits interact with K(v)2 subunits and primarily modulate the voltage dependence of inactivation of these heterotetrameric channels. Both K(v)2 and silent K(v) subunits are expressed in the mammalian nervous system, but little is known about their expression and function in sensory neurons. This study reports the presence of K(v)2.1, K(v)2.2, and silent subunit K(v)6.1, K(v)8.1, K(v)9.1, K(v)9.2, and K(v)9.3 mRNA in mouse dorsal root ganglia (DRG). Immunocytochemistry confirmed the protein expression of K(v)2.x and K(v)9.x subunits in cultured small DRG neurons. To investigate if K(v)2 and silent K(v) subunits are underlying the delayed rectifier K(+) current (I(K)) in these neurons, K(v)2-mediated currents were isolated by the extracellular application of rStromatoxin-1 (ScTx) or by the intracellular application of K(v)2 antibodies. Both ScTx- and anti-K(v)2.1-sensitive currents displayed two components in their voltage dependence of inactivation. Together, both components accounted for approximately two-thirds of I(K). A comparison with results obtained in heterologous expression systems suggests that one component reflects homotetrameric K(v)2.1 channels, whereas the other component represents heterotetrameric K(v)2.1/silent K(v) channels. These observations support a physiological role for silent K(v) subunits in small DRG neurons.
Collapse
Affiliation(s)
- Elke Bocksteins
- Laboratory for Molecular Biophysics, Physiology and Pharmacology, Department of Biomedical Sciences, University of Antwerp, Antwerpen 2610, Belgium
| | | | | | | | | | | |
Collapse
|
23
|
Gu Q, Lim ME, Gleich GJ, Lee LY. Mechanisms of eosinophil major basic protein-induced hyperexcitability of vagal pulmonary chemosensitive neurons. Am J Physiol Lung Cell Mol Physiol 2009; 296:L453-61. [PMID: 19136577 DOI: 10.1152/ajplung.90467.2008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We have reported recently that eosinophil-derived basic proteins directly enhance the capsaicin- and electrical stimulation-evoked whole cell responses in rat pulmonary sensory neurons (19). Our present study further elucidates the mechanisms underlying the sensitization of pulmonary afferent nerves induced by these cationic proteins. Our results show that pretreatment with eosinophil major basic protein (MBP; 2 microM, 60 s) significantly enhanced the excitability of isolated rat vagal pulmonary chemosensitive neurons to acid and ATP in the current-clamp mode, but this potentiating effect was absent in the voltage-clamp recordings. The hyperexcitability induced by MBP was not prevented by the blockade of either transient receptor potential vanilloid type-1 receptor (TRPV1) selectively (inhibitor: AMG 9810; 1 microM, 2 min) or all TRPV1-4 channels (inhibitor: ruthenium red; 5 microM, 2 min). In addition, MBP also markedly potentiated the excitability of mouse pulmonary chemosensitive neurons, and no detectable difference was found between those isolated from wild-type and TRPV1 knockout mice. Furthermore, MBP pretreatment affected the decay time and recovery phase of the action potentials evoked by current injections and significantly inhibited both the sustained delayed-rectifier voltage-gated K(+) current (IK(dr)) and the A-type, fast-inactivating K(+) current (IK(a)) in these sensory neurons. In conclusion, our results indicate that the inhibition of IK(dr) and IK(a) should, at least in part, account for the hyperexcitability of pulmonary chemosensitive neurons induced by eosinophil-derived cationic proteins, whereas an interaction with TRPV1 channels does not seem to be required for the sensitizing effect of these proteins.
Collapse
Affiliation(s)
- Qihai Gu
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky 40536-0298, USA
| | | | | | | |
Collapse
|
24
|
Su T, Cong W, Long Y, Luo A, Sun W, Deng W, Liao W. Altered expression of voltage-gated potassium channel 4.2 and voltage-gated potassium channel 4-interacting protein, and changes in intracellular calcium levels following lithium-pilocarpine-induced status epilepticus. Neuroscience 2008; 157:566-76. [DOI: 10.1016/j.neuroscience.2008.09.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 09/16/2008] [Accepted: 09/19/2008] [Indexed: 11/16/2022]
|
25
|
Catacuzzeno L, Fioretti B, Pietrobon D, Franciolini F. The differential expression of low-threshold K+ currents generates distinct firing patterns in different subtypes of adult mouse trigeminal ganglion neurones. J Physiol 2008; 586:5101-18. [PMID: 18772201 PMCID: PMC2652152 DOI: 10.1113/jphysiol.2008.159384] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Accepted: 08/29/2008] [Indexed: 01/13/2023] Open
Abstract
In adult mouse trigeminal ganglion (TG) neurones we identified three neuronal subpopulations, defined in terms of their firing response to protracted depolarizations, namely MF neurones, characterized by a multiple tonic firing; DMF neurones, characterized by a delay before the beginning of repetitive firing; and SS neurones, characterized by a strongly adapting response. The three subpopulations also differed in several other properties important for defining their functional role in vivo, namely soma size, action potential (AP) shape and capsaicin sensitivity. MF neurones had small soma, markedly long AP and mostly responded to capsaicin, properties typical of a subgroup of C-type nociceptors. SS neurones had large soma, short AP duration and were mostly capsaicin insensitive, suggesting that most of them have functions other than nociception. DMF neurones were all capsaicin insensitive, had a small soma size and intermediate AP duration, making them functionally distinct from both MF and SS neurones. We investigated the ionic basis underlying the delay to the generation of the first AP of DMF neurones, and the strong adaptation of SS neurones. We found that the expression of a fast-inactivating, 4-AP- and CP-339,818-sensitive K+ current (I(A)) in DMF neurones plays a critical role in the generation of the delay, whereas a DTX-sensitive K+ current (I(DTX)) selectively expressed in SS neurones appeared to be determinant for their strong firing adaptation. A minimal theoretical model of TG neuronal excitability confirmed that I(A) and I(DTX) have properties congruent with their suggested role.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Dipartimento Biologia Cellulare e Ambientale, Universitá di Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
26
|
Khaliq ZM, Bean BP. Dynamic, nonlinear feedback regulation of slow pacemaking by A-type potassium current in ventral tegmental area neurons. J Neurosci 2008; 28:10905-17. [PMID: 18945898 PMCID: PMC2837275 DOI: 10.1523/jneurosci.2237-08.2008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2008] [Revised: 09/10/2008] [Accepted: 09/11/2008] [Indexed: 11/21/2022] Open
Abstract
We analyzed ionic currents that regulate pacemaking in dopaminergic neurons of the mouse ventral tegmental area by comparing voltage trajectories during spontaneous firing with ramp-evoked currents in voltage clamp. Most recordings were made in brain slice, with key experiments repeated using acutely dissociated neurons, which gave identical results. During spontaneous firing, net ionic current flowing between spikes was calculated from the time derivative of voltage multiplied by cell capacitance, signal-averaged over many firing cycles to enhance resolution. Net inward interspike current had a distinctive nonmonotonic shape, reaching a minimum (generally <1 pA) between -60 and -55 mV. Under voltage clamp, ramps over subthreshold voltages elicited a time- and voltage-dependent outward current that peaked near -55 mV. This current was undetectable with 5 mV/s ramps and increased steeply with depolarization rate over the range (10-50 mV/s) typical of natural pacemaking. Ramp-evoked subthreshold current was resistant to alpha-dendrotoxin, paxilline, apamin, and tetraethylammonium but sensitive to 4-aminopyridine and 0.5 mM Ba2+, consistent with A-type potassium current (I(A)). Same-cell comparison of currents elicited by various ramp speeds with natural spontaneous depolarization showed how the steep dependence of I(A) on depolarization rate results in small net inward currents during pacemaking. These results reveal a mechanism in which subthreshold I(A) is near zero at steady state, but is engaged at depolarization rates >10 mV/s to act as a powerful, supralinear feedback element. This feedback mechanism explains how net ionic current can be constrained to <1-2 pA but reliably inward, thus enabling slow, regular firing.
Collapse
Affiliation(s)
- Zayd M. Khaliq
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| | - Bruce P. Bean
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
27
|
Abstract
One of the major issues for modern neuroscience research concerns the molecular and cellular mechanisms that underlie the acquisition, storage, and recollection of memories by the brain. Regulation of the strength of individual synaptic inputs (synaptic plasticity) has, for decades, been the front-running candidate mechanism for cellular information storage, with some direct supporting evidence recently obtained. Research into the molecular mechanisms responsible for changing synaptic strength has, to date, primarily focused on trafficking and properties of the neurotransmitter receptors themselves (AMPARs and NMDARs). However, recent evidence indicates that, subsequent to receptor activation, synaptic inputs are subject to regulation by synaptically located K+ channels. It is therefore critical to understand the biophysical properties and subcellular localization (density and distribution) of these channels and how their properties are modulated. Here we will review recent findings showing that two different classes of K+ channels (A-type and small conductance, Ca2+ -activated K+ channels), beyond their traditional role in regulating action potential firing, contribute to the regulation of synaptic strength in the hippocampus. In addition, we discuss how modulation of these channels' properties and expression might contribute to synaptic plasticity.
Collapse
Affiliation(s)
- Jinhyun Kim
- Molecular Neurophysiology and Biophysics Unit, Laboratory of Cellular and Synaptic Neurophysiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
28
|
Jessica Chen M, Sepramaniam S, Armugam A, Shyan Choy M, Manikandan J, Melendez AJ, Jeyaseelan K, Sang Cheung N. Water and ion channels: crucial in the initiation and progression of apoptosis in central nervous system? Curr Neuropharmacol 2008; 6:102-16. [PMID: 19305791 PMCID: PMC2647147 DOI: 10.2174/157015908784533879] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 09/09/2007] [Accepted: 10/01/2007] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death (PCD), is a highly regulated and sophisticated cellular mechanism that commits cell to isolated death fate. PCD has been implicated in the pathogenesis of numerous neurodegenerative disorders. Countless molecular events underlie this phenomenon, with each playing a crucial role in death commitment. A precedent event, apoptotic volume decrease (AVD), is ubiquitously observed in various forms of PCD induced by different cellular insults. Under physiological conditions, cells when subjected to osmotic fluctuations will undergo regulatory volume increase/decrease (RVI/RVD) to achieve homeostatic balance with neurons in the brain being additionally protected by the blood-brain-barrier. However, during AVD following apoptotic trigger, cell undergoes anistonic shrinkage that involves the loss of water and ions, particularly monovalent ions e.g. K(+), Na(+) and Cl(-). It is worthwhile to concentrate on the molecular implications underlying the loss of these cellular components which posed to be significant and crucial in the successful propagation of the apoptotic signals. Microarray and real-time PCR analyses demonstrated several ion and water channel genes are regulated upon the onset of lactacystin (a proteosomal inhibitor)-mediated apoptosis. A time course study revealed that gene expressions of water and ion channels are being modulated just prior to apoptosis, some of which are aquaporin 4 and 9, potassium channels and chloride channels. In this review, we shall looked into the molecular protein machineries involved in the execution of AVD in the central nervous system (CNS), and focus on the significance of movements of each cellular component in affecting PCD commitment, thus provide some pharmacological advantages in the global apoptotic cell death.
Collapse
Affiliation(s)
- Minghui Jessica Chen
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Sugunavathi Sepramaniam
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Arunmozhiarasi Armugam
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Meng Shyan Choy
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jayapal Manikandan
- Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Alirio J Melendez
- Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Kandiah Jeyaseelan
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Nam Sang Cheung
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
29
|
Lee AW, Pfaff DW. Hormone effects on specific and global brain functions. J Physiol Sci 2008; 58:213-20. [PMID: 18505601 DOI: 10.2170/physiolsci.rv007008] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Accepted: 05/24/2008] [Indexed: 11/05/2022]
Abstract
The first demonstration of how biochemical changes in neurons in specific parts of the brain direct a complete mammalian behavior derived from the effects of estrogens in hypothalamic neurons that facilitate lordosis behavior, the primary reproductive behavior of female quadrupeds (Pfaff. Estrogens and Brain Function. 1980; Pfaff. Drive: Neurobiological and Molecular Mechanisms of Sexual Motivation. 1999). Sex behaviors depend on sexual arousal that in turn depends on a primitive function: generalized CNS arousal (Pfaff. Brain Arousal and Information Theory. 2006). Here we summarize one of the ways in which a generalized arousal transmitter, norepinephrine, can influence the electrical excitability of ventromedial hypothalamic cells in a way that will foster female sex behavior.
Collapse
Affiliation(s)
- A W Lee
- Laboratory of Neurobiology and Behavior, The Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
30
|
Estradiol modulation of phenylephrine-induced excitatory responses in ventromedial hypothalamic neurons of female rats. Proc Natl Acad Sci U S A 2008; 105:7333-8. [PMID: 18480251 DOI: 10.1073/pnas.0802760105] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Estrogens act within the ventromedial nucleus of the hypothalamus (VMN) to facilitate lordosis behavior. Estradiol treatment in vivo induces alpha(1b)-adrenoreceptor mRNA and increases the density of alpha(1B)-adrenoreceptor binding in the hypothalamus. Activation of hypothalamic alpha(1)-adrenoceptors also facilitates estrogen-dependent lordosis. To investigate the cellular mechanisms of adrenergic effects on VMN neurons, whole-cell patch-clamp recordings were carried out on hypothalamic slices from control and estradiol-treated female rats. In control slices, bath application of the alpha(1)-agonist phenylephrine (PHE; 10 microM) depolarized 10 of 25 neurons (40%), hyperpolarized three neurons (12%), and had no effect on 12 neurons (48%). The depolarization was associated with decreased membrane conductance, and this current had a reversal potential close to the K(+) equilibrium potential. The alpha(1b)-receptor antagonist chloroethylclonidine (10 microM) blocked the depolarization produced by PHE in all cells. From estradiol-treated rats, significantly more neurons in slices depolarized (71%) and fewer neurons showed no response (17%) to PHE. PHE-induced depolarizations were significantly attenuated with 4-aminopyridine (5 mM) but unaffected by tetraethylammonium chloride (20 mM) or blockers of Na(+) and Ca(2+) channels. These data indicate that alpha(1)-adrenoceptors depolarize VMN neurons by reducing membrane conductance for K(+). Estradiol amplifies alpha(1b)-adrenergic signaling by increasing the proportion of VMN neurons that respond to stimulation of alpha(1b)-adrenergic receptors, which is expected in turn to promote lordosis.
Collapse
|
31
|
Nerbonne JM, Gerber BR, Norris A, Burkhalter A. Electrical remodelling maintains firing properties in cortical pyramidal neurons lacking KCND2-encoded A-type K+ currents. J Physiol 2008; 586:1565-79. [PMID: 18187474 DOI: 10.1113/jphysiol.2007.146597] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Considerable experimental evidence has accumulated demonstrating a role for voltage-gated K(+) (Kv) channel pore-forming (alpha) subunits of the Kv4 subfamily in the generation of fast transient outward K(+), I(A), channels. Immunohistochemical data suggest that I(A) channels in hippocampal and cortical pyramidal neurons reflect the expression of homomeric Kv4.2 channels. The experiments here were designed to define directly the role of Kv4.2 in the generation of I(A) in cortical pyramidal neurons and to determine the functional consequences of the targeted deletion of Kv4.2 on the resting and active membrane properties of these cells. Whole-cell voltage-clamp recordings, obtained from visual cortical pyramidal neurons isolated from mice in which the KCND2 (Kv4.2) locus was disrupted (Kv4.2-/- mice), revealed that I(A) is indeed eliminated. In addition, the densities of other Kv current components, specifically I(K) and I(ss), are increased significantly (P < 0.001) in most ( approximately 80%) Kv4.2-/- cells. The deletion of KCND2 (Kv4.2) and the elimination of I(A) is also accompanied by the loss of the Kv4 channel accessory protein KChIP3, suggesting that in the absence of Kv4.2, the KChIP3 protein is targeted for degradation. The expression levels of several Kv alpha subunits (Kv4.3, Kv1.4, Kv2.1, Kv2.2), however, are not measurably altered in Kv4.2-/- cortices. Although I(A) is eliminated in Kv4.2-/- pyramidal neurons, the mean +/- s.e.m. current threshold for action potential generation and the waveforms of action potentials are indistinguishable from those recorded from wild-type cells. Repetitive firing is also maintained in Kv4.2-/- cortical pyramidal neurons, suggesting that the increased densities of I(K) and I(ss) compensate for the in vivo loss of I(A).
Collapse
Affiliation(s)
- Jeanne M Nerbonne
- Department of Molecular Biology and Pharmacology, Box 8103, Washington University Medical School, 660 South Euclid Avenue, St Louis, MO 63110-1093, USA.
| | | | | | | |
Collapse
|
32
|
Jackson AC, Bean BP. State-dependent enhancement of subthreshold A-type potassium current by 4-aminopyridine in tuberomammillary nucleus neurons. J Neurosci 2007; 27:10785-96. [PMID: 17913912 PMCID: PMC6672835 DOI: 10.1523/jneurosci.0935-07.2007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A-type potassium current (I(A)) both activates and inactivates at subthreshold voltages. We asked whether there is steady-state I(A) at subthreshold voltages, using dissociated mouse tuberomammillary nucleus neurons, pacemaking neurons with large I(A) currents in which subthreshold I(A) might regulate firing frequency. With slow depolarizing voltage ramps (20 mV/s), there was no discernible component of steady-state outward current in the range of -70 to -40 mV. However, faster ramps of 50-100 mV/s, similar to the rate of spontaneous depolarization during pacemaking, did evoke subthreshold outward currents. Ramp-evoked current at subthreshold voltages was unaffected by 10 mM tetraethylammonium and likely represents I(A), because its voltage dependence overlaps that of I(A) activation (midpoint near -44 mV) and inactivation (midpoint near -85 mV). However, although 4-aminopyridine (4-AP) inhibited peak I(A) activated by step depolarizations as expected (IC50, approximately 1 mM), ramp-evoked current was instead dramatically enhanced (current at -40 mV evoked by 50 mV/s ramp enhanced >15-fold by 10 mM 4-AP). In cell-attached recordings of spontaneous pacemaking, 10 mM 4-AP slowed rather than speeded firing, consistent with enhancement of subthreshold I(A). Also consistent with such enhancement, 4-AP also greatly increased the latency to first spike after long hyperpolarizations. The striking enhancement of I(A) during depolarizing ramps can be explained by a model in which 4-AP binds tightly to closed channels but must unbind before channels can inactivate. Thus, the state dependence of 4-AP binding to the channels underlying I(A) can result in effects on firing patterns opposite to those expected from simple block of I(A).
Collapse
Affiliation(s)
| | - Bruce P. Bean
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
33
|
Ellis LD, Krahe R, Bourque CW, Dunn RJ, Chacron MJ. Muscarinic receptors control frequency tuning through the downregulation of an A-type potassium current. J Neurophysiol 2007; 98:1526-37. [PMID: 17615127 PMCID: PMC5053812 DOI: 10.1152/jn.00564.2007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The functional role of cholinergic input in the modulation of sensory responses was studied using a combination of in vivo and in vitro electrophysiology supplemented by mathematical modeling. The electrosensory system of weakly electric fish recognizes different environmental stimuli by their unique alteration of a self-generated electric field. Variations in the patterns of stimuli are primarily distinguished based on their frequency. Pyramidal neurons in the electrosensory lateral line lobe (ELL) are often tuned to respond to specific input frequencies. Alterations in the tuning of the pyramidal neurons may allow weakly electric fish to preferentially select for certain stimuli. Here we show that muscarinic receptor activation in vivo enhances the excitability, burst firing, and subsequently the response of pyramidal cells to naturalistic sensory input. Through a combination of in vitro electrophysiology and mathematical modeling, we reveal that this enhanced excitability and bursting likely results from the down-regulation of an A-type potassium current. Further, we provide an explanation of the mechanism by which these currents can mediate frequency tuning.
Collapse
Affiliation(s)
- Lee D Ellis
- Center for Research in Neuroscience, McGill University, Montreal, Canada
| | | | | | | | | |
Collapse
|
34
|
Hammack SE, Mania I, Rainnie DG. Differential Expression of Intrinsic Membrane Currents in Defined Cell Types of the Anterolateral Bed Nucleus of the Stria Terminalis. J Neurophysiol 2007; 98:638-56. [PMID: 17537902 DOI: 10.1152/jn.00382.2007] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The anterolateral group of the bed nucleus of the stria terminalis (BNSTALG) plays a critical role in a diverse array of behaviors, although little is known of the physiological properties of neurons in this region. Using whole cell patch-clamp recordings from rat BNSTALG slices in vitro, we describe three distinct physiological cell types. Type I neurons were characterized by the presence of a depolarizing sag in response to hyperpolarizing current injection that resembled activation of the hyperpolarization-activated cation current Ih and a regular firing pattern in response to depolarizing current injection. Type II neurons exhibited the same depolarizing sag in response to hyperpolarizing current injection, but burst-fired in response to depolarizing current injection, which was indicative of the activation of the low-threshold calcium current IT. Type III neurons did not exhibit a depolarizing sag in response to hyperpolarizing current injection, but instead exhibited a fast time-independent rectification that became more pronounced with increased amplitude of hyperpolarizing current injection, and was indicative of activation of the inwardly rectifying potassium current IK(IR). Type III neurons also exhibited a regular firing pattern in response to depolarizing current. Using voltage-clamp analysis we further characterized the primary active currents that shaped the physiological properties of these distinct cell types, including Ih, IT, IK(IR), the voltage-dependent potassium current IA, and the persistent sodium current INaP. The functional relevance of each cell type is discussed in relation to prior anatomical studies, as well as how these currents may interact to modulate neuronal activity within the BNSTALG.
Collapse
Affiliation(s)
- Sayamwong E Hammack
- Department of Psychiatry and Behavioral Science, Center for Behavioral Neuroscience, Emory University, Atlanta, Georgia 30329, USA
| | | | | |
Collapse
|
35
|
Hess D, Nanou E, El Manira A. Characterization of Na+-Activated K+ Currents in Larval Lamprey Spinal Cord Neurons. J Neurophysiol 2007; 97:3484-93. [PMID: 17329626 DOI: 10.1152/jn.00742.2006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Potassium channels play an important role in controlling neuronal firing and synaptic interactions. Na+-activated K+ ( KNa) channels have been shown to exist in neurons in different regions of the CNS, but their physiological function has been difficult to assess. In this study, we have examined if neurons in the spinal cord possess KNa currents. We used whole cell recordings from isolated spinal cord neurons in lamprey. These neurons display two different KNa currents. The first was transient and activated by the Na+ influx during the action potentials, and it was abolished when Na+ channels were blocked by tetrodotoxin. The second KNa current was sustained and persisted in tetrodotoxin. Both KNa currents were abolished when Na+ was substituted with choline or N-methyl-d-glucamine, indicating that they are indeed dependent on Na+ influx into neurons. When Na+ was substituted with Li+, the amplitude of the inward current was unchanged, whereas the transient KNa current was reduced but not abolished. This suggests that the transient KNa current is partially activated by Li+. These two KNa currents have different roles in controlling the action potential waveform. The transient KNa appears to act as a negative feedback mechanism sensing the Na+ influx underlying the action potential and may thus be critical for setting the amplitude and duration of the action potential. The sustained KNa current has a slow kinetic of activation and may underlie the slow Ca2+-independent afterhyperpolarization mediated by repetitive firing in lamprey spinal cord neurons.
Collapse
Affiliation(s)
- Dietmar Hess
- Nobel Institute for Neurophysiology, Dept. of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | |
Collapse
|
36
|
Bourdeau ML, Morin F, Laurent CE, Azzi M, Lacaille JC. Kv4.3-mediated A-type K+ currents underlie rhythmic activity in hippocampal interneurons. J Neurosci 2007; 27:1942-53. [PMID: 17314290 PMCID: PMC6673559 DOI: 10.1523/jneurosci.3208-06.2007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 01/15/2007] [Accepted: 01/15/2007] [Indexed: 12/30/2022] Open
Abstract
Hippocampal-dependent learning and memory processes are associated with theta frequency rhythmic activity. Interneuron and pyramidal cell network interactions underlie this activity, but contributions of interneuron voltage-gated membrane conductances remain unclear. We show that interneurons at the CA1 lacunosum-moleculare (LM) and radiatum (RAD) junction (LM/RAD) display voltage-dependent subthreshold membrane potential oscillations (MPOs) generated by voltage-gated tetrodotoxin-sensitive Na+ and 4-aminopyridine (4-AP)-sensitive K+ currents. They also exhibit prominent 4-AP-sensitive A-type K+ currents, with gating properties showing activation at subthreshold membrane potentials. We found that LM/RAD cells are part of specific interneuron subpopulations expressing the K+ channel subunit Kv4.3 and their transfection with Kv4.3 small interfering RNA selectively impaired A-type K+ currents and MPOs. Thus, our findings reveal a novel function of Kv4.3-mediated A-type K+ currents in the generation of intrinsic MPOs in specific subpopulations of interneurons that may participate in hippocampal theta-related rhythmic activity.
Collapse
Affiliation(s)
- Mathieu L. Bourdeau
- Département de Physiologie, Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - France Morin
- Département de Physiologie, Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Charles E. Laurent
- Département de Physiologie, Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Mounia Azzi
- Département de Physiologie, Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | - Jean-Claude Lacaille
- Département de Physiologie, Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| |
Collapse
|
37
|
Kayssi A, Amadesi S, Bautista F, Bunnett NW, Vanner S. Mechanisms of protease-activated receptor 2-evoked hyperexcitability of nociceptive neurons innervating the mouse colon. J Physiol 2007; 580:977-91. [PMID: 17289784 PMCID: PMC2075455 DOI: 10.1113/jphysiol.2006.126599] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Agonists of protease-activated receptor 2 (PAR(2)) evoke hyperexcitability of dorsal root ganglia (DRG) neurons by unknown mechanisms. We examined the cellular mechanisms underlying PAR(2)-evoked hyperexcitability of mouse colonic DRG neurons to determine their potential role in pain syndromes such as visceral hyperalgesia. Colonic DRG neurons were identified by injecting Fast Blue and DiI retrograde tracers into the mouse colon. Using immunofluorescence, we found that DiI-labelled neurons contained PAR(2) immunoreactivity, confirming the presence of receptors on colonic neurons. Whole-cell current-clamp recordings of acutely dissociated neurons demonstrated that PAR(2) activation with a brief application (3 min) of PAR(2) agonists, SLIGRL-NH(2) and trypsin, evoked sustained depolarizations (up to 60 min) which were associated with increased input resistance and a marked reduction in rheobase (50% at 30 min). In voltage clamp, SLIGRL-NH(2) markedly suppressed delayed rectifier I(K) currents (55% at 10 min), but had no effect on the transient I(A) current or TTX-resistant Na(+) currents. In whole-cell current-clamp recordings, the sustained excitability evoked by PAR(2) activation was blocked by the PKC inhibitor, calphostin, and the ERK(1/2) inhibitor PD98059. Studies of ERK(1/2) phosphorylation using confocal microscopy demonstrated that SLIGRL-NH(2) increased levels of immunoreactive pERK(1/2) in DRG neurons, particularly in proximity to the plasma membrane. Thus, activation of PAR(2) receptors on colonic nociceptive neurons causes sustained hyperexcitability that is related, at least in part, to suppression of delayed rectifier I(K) currents. Both PKC and ERK(1/2) mediate the PAR(2)-induced hyperexcitability. These studies describe a novel mechanism of sensitization of colonic nociceptive neurons that may be implicated in conditions of visceral hyperalgesia such as irritable bowel syndrome.
Collapse
Affiliation(s)
- Ahmed Kayssi
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
38
|
Yu HB, Li ZB, Zhang HX, Wang XL. Role of potassium channels in Abeta(1-40)-activated apoptotic pathway in cultured cortical neurons. J Neurosci Res 2007; 84:1475-84. [PMID: 17022037 DOI: 10.1002/jnr.21054] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Potassium channel dysfunction has been implicated in Alzheimer's disease (AD). In the present study, by using potassium channel blocker tetraethylammonium (TEA), we investigated the relationship between the enhancement of potassium currents and the alteration of apoptotic cascade in the neuronal apoptotic model induced by beta-amyloid peptide 1-40(Abeta(1-40)). Cortical neurons exposed to Abeta(1-40) 5 muM developed a specific increase in the delayed rectifier potassium current (I(K)), but not the transient outward potassium currents (I(A)), before the appearance of neuronal apoptosis. Abeta(1-40) induced various apoptotic features such as chromatin condensation, a decrease in the amount of Bcl-2 protein, an increase in the amount of Bax protein, cytochrome c release from mitochondria, and caspase-3 activation. Potassium channel blocker 5 mM TEA attenuated Abeta(1-40)-induced neuronal death and prevented the alterations of all above mentioned apoptotic indicators. The study indicates that I(K) enhancement might play an important role in certain form of programmed cell death induced by beta-amyloid peptide (Abeta). Increased potassium channel activity might trigger the activation of apoptosis cascade in Abeta(1-40)-treated rat cortical neurons.
Collapse
Affiliation(s)
- Hai-Bo Yu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
39
|
Vacher H, Diochot S, Bougis PE, Martin-Eauclaire MF, Mourre C. Kv4 channels sensitive to BmTX3 in rat nervous system: autoradiographic analysis of their distribution during brain ontogenesis. Eur J Neurosci 2006; 24:1325-40. [PMID: 16987219 DOI: 10.1111/j.1460-9568.2006.05020.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The binding site distribution of sBmTX3, a chemically synthesized toxin originally purified from the venom of the scorpion Buthus martensi, was investigated in adult and developing rat brain, using patch-clamp experiments and quantitative autoradiography. The molecular basis of these sBmTX3 sites was analysed by electrophysiology on transient Kv currents recorded in mammalian transfected cells. The rapidly activating and inactivating Kv4.1 current was inhibited by sBmTX3 (IC50, 105 nM). The inhibition was less effective on Kv4.2 and Kv4.3 channels and the toxin did not affect other transient currents such as Kv1.4 and Kv3.4. The distribution of the 125I-sBmTX3 binding sites was heterogeneous, with a 113-fold difference between the highest and the lowest densities in adult rat brain. The site density was particularly important in the caudate-putamen and accumbens nucleus, thalamus, hippocampal formation and cerebellum. The affinity of sBmTX3 remained constant during brain ontogenesis. The level of sBmTX3 binding sites was very low in prenatal and postnatal stages to postnatal day (P)12 but drastically increased from P15 in the major part of the studied structures except in the CA3 hippocampal field where the density was very high from P6. Thus, the distribution of sBmTX3 binding sites in rat brain and its electrophysiological characteristics suggest that sBmTX3 specifically binds to the Kv4 subfamily of K channels.
Collapse
Affiliation(s)
- Hélène Vacher
- CNRS, UMR 6149, Neurobiologie des Processus Mnésiques, Université de Provence, Pôle 3C, Centre St Charles, 3 Place Victor Hugo, 13331 Marseille cedex 03, France
| | | | | | | | | |
Collapse
|
40
|
Reyes-Haro D, Miledi R, García-Colunga J. Potassium currents in primary cultured astrocytes from the rat corpus callosum. ACTA ACUST UNITED AC 2006; 34:411-20. [PMID: 16902762 DOI: 10.1007/s11068-006-8727-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Revised: 02/23/2005] [Accepted: 04/08/2005] [Indexed: 11/25/2022]
Abstract
The corpus callosum (CC) is the main white matter tract in the brain and is involved in interhemispheric communication. Using the whole-cell voltage-clamp technique, a study was made of K(+)-currents in primary cultured astrocytes from the CC of newborn rats. These cells were positive to glial fibrillary acidic protein after culturing in Dulbecco's Modified Eagle Medium (> 95% of cells) or in serum-free neurobasal medium with G5 supplement (> 99% of cells). Astrocytes cultured in either medium displayed similar voltage-activated ion currents. In 81% of astrocytes, the current had a transient component and a sustained component, which were blocked by 4-aminopyridine and tetraethylammonium, respectively; and both had a reversal potential of -66 mV, indicating that they were carried by K(+) ions. Based on the Ba(2+)-sensitivity and activation kinetics of the K(+)-current, two groups of astrocytes were discerned. One group (55% of cells) displayed a strong Ba(2+) blockade of the K(+)-current whose activation kinetics, time course of decay, and the current-voltage relationship were modified by Ba(2+). This current was greatly blocked (52%) by Ba(2+) in a voltage-dependent way. Another group (45% of cells) presented weak Ba(2+)-blockade, which was only blocked 24% by Ba(2+). The activation kinetics and time course of decay of this current component were unaffected by Ba(2+). These results may help to understand better the roles of voltage-activated K(+)-currents in astrocytes from the rat CC in particular and glial cells in general.
Collapse
Affiliation(s)
- Daniel Reyes-Haro
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, 76230, México
| | | | | |
Collapse
|
41
|
Moulder KL, Meeks JP, Mennerick S. Homeostatic regulation of glutamate release in response to depolarization. Mol Neurobiol 2006; 33:133-53. [PMID: 16603793 DOI: 10.1385/mn:33:2:133] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Revised: 11/30/1999] [Accepted: 08/04/2005] [Indexed: 11/11/2022]
Abstract
Proper nervous system function requires a balance between excitation and inhibition. Systems of homeostasis may have evolved in neurons to help maintain or restore balance between excitation and inhibition, presumably because excessive excitation can cause dysfunction and cell death. This article reviews evidence for homeostatic mechanisms within the hippocampus that lead to differential regulation of glutamate and gamma-aminobutyric acid release in response to conditions of excess depolarization. We recently found differential effects on glutamate release at the level of action potential coupling to transmitter release, vesicular release probability, and vesicle availability. Such differential regulation may help to prevent excitotoxicity and runaway excitation.
Collapse
Affiliation(s)
- Krista L Moulder
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
42
|
Hu HJ, Carrasquillo Y, Karim F, Jung WE, Nerbonne JM, Schwarz TL, Gereau RW. The kv4.2 potassium channel subunit is required for pain plasticity. Neuron 2006; 50:89-100. [PMID: 16600858 DOI: 10.1016/j.neuron.2006.03.010] [Citation(s) in RCA: 212] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Revised: 08/10/2005] [Accepted: 03/03/2006] [Indexed: 01/22/2023]
Abstract
A-type potassium currents are important determinants of neuronal excitability. In spinal cord dorsal horn neurons, A-type currents are modulated by extracellular signal-regulated kinases (ERKs), which mediate central sensitization during inflammatory pain. Here, we report that Kv4.2 mediates the majority of A-type current in dorsal horn neurons and is a critical site for modulation of neuronal excitability and nociceptive behaviors. Genetic elimination of Kv4.2 reduces A-type currents and increases excitability of dorsal horn neurons, resulting in enhanced sensitivity to tactile and thermal stimuli. Furthermore, ERK-mediated modulation of excitability in dorsal horn neurons and ERK-dependent forms of pain hypersensitivity are absent in Kv4.2(-/-) mice compared to wild-type littermates. Finally, mutational analysis of Kv4.2 indicates that S616 is the functionally relevant ERK phosphorylation site for modulation of Kv4.2-mediated currents in neurons. These results show that Kv4.2 is a downstream target of ERK in spinal cord and plays a crucial role in pain plasticity.
Collapse
Affiliation(s)
- Hui-Juan Hu
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Casassus G, Blanchet C, Mulle C. Short-term regulation of information processing at the corticoaccumbens synapse. J Neurosci 2006; 25:11504-12. [PMID: 16354908 PMCID: PMC6726026 DOI: 10.1523/jneurosci.2466-05.2005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In relation to expectation and delivery of reward, pyramidal neurons of the prefrontal cortex either switch from a single spiking mode to transient phasic bursting, or gradually increase their sustained tonic activity. Here, we examined how switching between firing modes affects information processing at the corticoaccumbens synapse. We report that increasing presynaptic firing frequency in a tonic manner either depresses or facilitates synaptic transmission, depending on initial probability of release. In contrast, repeated bursts of stimulation of cortical afferents trigger a new form of short-term potentiation of synaptic transmission (RB-STP) in the nucleus accumbens (NAc). RB-STP involves the regulation of axonal excitability mediated by 4-AP-sensitive potassium channels in afferent cortical neurons. Thus, in a tonic mode, information flow is tightly controlled by regulatory mechanisms at the level of presynaptic terminals, whereas switching to a bursting mode reliably enhances efficacy of information processing for all cortical afferents to NAc neurons.
Collapse
Affiliation(s)
- Guillaume Casassus
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5091, Institut François Magendie, Université Victor Segalen-Bordeaux II, 33077 Bordeaux, France
| | | | | |
Collapse
|
44
|
Kourrich S, Manrique C, Salin P, Mourre C. Transient hippocampal down-regulation of Kv1.1 subunit mRNA during associative learning in rats. Learn Mem 2006; 12:511-9. [PMID: 16204203 PMCID: PMC1240063 DOI: 10.1101/lm.86305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Voltage-gated potassium channels (Kv) are critically involved in learning and memory processes. It is not known, however, whether the expression of the Kv1.1 subunit, constituting Kv1 channels, can be specifically regulated in brain areas important for learning and memory processing. Radioactive in situ hybridization was used to evaluate the content of Kv1.1 alpha-subunit mRNA in the olfactory bulb, ventral, and dorsal hippocampus at different stages of an odor-discrimination associative task in rats. Naive, conditioned, and pseudoconditioned animals were sacrificed at different times either prior to a two-odor significance learning or after odor discrimination was established. Important decreases of Kv1.1 mRNA levels were transiently observed in the ventral hippocampus before successful learning when compared with the pseudoconditioned group. Moreover, temporal group analysis showed significant labeling alterations in the hippocampus of conditioned and pseudoconditioned groups throughout the training. Finally, Kv1.1 mRNA levels in the hippocampus were positively correlated with odor-reward association learning in rats that were beginning to discriminate between odors. These findings indicate that the Kv1.1 subunit is transiently down-regulated in the early stages of learning and suggest that Kv1 channel expression regulation is critical for the modification of neuronal substrates underlying new information acquisition.
Collapse
Affiliation(s)
- Saïd Kourrich
- Laboratoire de Neurobiologie des Processus Mnésiques, UMR 6149 CNRS-Université de Provence, Marseille, France
| | | | | | | |
Collapse
|
45
|
Han P, Lucero MT. Pituitary adenylate cyclase activating polypeptide reduces expression of Kv1.4 and Kv4.2 subunits underlying A-type K(+) current in adult mouse olfactory neuroepithelia. Neuroscience 2006; 138:411-9. [PMID: 16426762 DOI: 10.1016/j.neuroscience.2005.11.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2005] [Revised: 11/09/2005] [Accepted: 11/20/2005] [Indexed: 10/25/2022]
Abstract
A-type K(+) currents (I(A)) in olfactory receptor neurons have been characterized electrophysiologically but the molecular identities of the underlying channel subunits have not been determined. Using RT-PCR, immunoblot and immunohistochemistry, we found that the two candidate channel families underlying I(A), shaker and shal, are expressed in olfactory epithelia of Swiss Webster mice. Specifically, Kv1.4, the only I(A) candidate from the shaker family, and Kv4.2 and Kv4.3 from the shal family were expressed, but Kv4.1 mRNA was not amplified from the olfactory epithelia. Immunoblot and immunohistochemical studies confirmed the existence of Kv1.4 and Kv4.2/3 subunits. Furthermore, quantitative RT-PCR showed that pituitary adenylate cyclase activating polypeptide (PACAP) reduced the expression of Kv1.4 and Kv4.2 but did not reduce the already low expression of Kv4.3. The PACAP-induced reduction of Kv4.1 and Kv4.2 expression was completely blocked by inhibiting the phospholipase C (PLC) pathway but was still significantly downregulated by PACAP when the cyclic AMP pathway was inhibited. In addition, downstream of the PLC pathway, calcium mediated the reduction of both Kv1.4 and Kv4.2 expression and I(A) current density. Phosphokinase C (PKC) activation did not affect Kv1.4 and Kv4.2 mRNA expression, even though PKC reduced I(A) current density. Together with our previous studies, our data suggest that A-type K(+) currents in olfactory receptor neurons are composed of multiple K(+) channel subunits, among which Kv1.4 and Kv4.2 are subject to transcriptional modulation by PACAP. We also found that PACAP predominately uses a PLC-calcium pathway to modulate Kv4.1 and Kv4.2 expression. Modulation of A-type K(+) current expression may contribute to the previously observed neuroprotective effects of PACAP on olfactory receptor neurons.
Collapse
Affiliation(s)
- P Han
- Interdepartmental Neuroscience Program, University of Utah, 20 North 1900 East, Salt Lake City, UT 84132-3401, USA
| | | |
Collapse
|
46
|
Toledo-Rodriguez M, El Manira A, Wallén P, Svirskis G, Hounsgaard J. Cellular signalling properties in microcircuits. Trends Neurosci 2005; 28:534-40. [PMID: 16112756 DOI: 10.1016/j.tins.2005.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 06/29/2005] [Accepted: 08/10/2005] [Indexed: 10/25/2022]
Abstract
Molecules and cells are the signalling elements in microcircuits. Recent studies have uncovered bewildering diversity in postsynaptic signalling properties in all areas of the vertebrate nervous system. Major effort is now being invested in establishing the specialized signalling properties at the cellular and molecular levels in microcircuits in specific brain regions. This review is part of the TINS Microcircuits Special Feature.
Collapse
Affiliation(s)
- Maria Toledo-Rodriguez
- Laboratory for Neural Microcircuitry, Brain Mind Institute, EPFL, Lausanne CH-1015, Switzerland
| | | | | | | | | |
Collapse
|
47
|
Yuan W, Burkhalter A, Nerbonne JM. Functional role of the fast transient outward K+ current IA in pyramidal neurons in (rat) primary visual cortex. J Neurosci 2005; 25:9185-94. [PMID: 16207878 PMCID: PMC6725755 DOI: 10.1523/jneurosci.2858-05.2005] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A molecular genetic approach was exploited to directly test the hypothesis that voltage-gated K+ (Kv) channel pore-forming (alpha) subunits of the Kv4 subfamily encode the fast transient outward K+ current (IA) in cortical pyramidal neurons and to explore the functional role of IA in shaping action potential waveforms and in controlling repetitive firing in these cells. Using the biolistic gene gun, cDNAs encoding a mutant Kv4.2 alpha subunit (Kv4.2W362F), which functions as a dominant negative (Kv4.2DN), and enhanced green fluorescent protein (EGFP) were introduced in vitro into neurons isolated from postnatal rat primary visual cortex. Whole-cell voltage-clamp recordings obtained from EGFP-positive pyramidal neurons revealed that IA is selectively eliminated in cells expressing Kv4.2DN. The densities and properties of the other Kv currents are unaffected. In neurons expressing Kv4.2DN, input resistances are increased and the (current) thresholds for action potential generation are decreased. In addition, action potential durations are prolonged, the amplitudes of afterhyperpolarizations are reduced, and the responses to prolonged depolarizing inputs are altered markedly in cells expressing Kv 4.2DN. At low stimulus intensities, firing rates are increased in Kv4.2DN-expressing cells, whereas at high stimulus intensities, Kv4.2DN-expressing cells adapt strongly. Together, these results demonstrate that Kv4alpha subunits encode IA channels and that IA plays a pivotal role in shaping the waveforms of individual action potentials and in controlling repetitive firing in visual cortical pyramidal neurons.
Collapse
Affiliation(s)
- Weilong Yuan
- Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
48
|
Han P, Lucero MT. Pituitary adenylate cyclase activating polypeptide reduces A-type K+ currents and caspase activity in cultured adult mouse olfactory neurons. Neuroscience 2005; 134:745-56. [PMID: 16019148 DOI: 10.1016/j.neuroscience.2005.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Revised: 03/24/2005] [Accepted: 05/07/2005] [Indexed: 11/20/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide has been shown to reduce apoptosis in neonatal cerebellar and olfactory receptor neurons, however the underlying mechanisms have not been elucidated. In addition, the neuroprotective effects of pituitary adenylate cyclase activating polypeptide have not been examined in adult tissues. To study the effects of pituitary adenylate cyclase activating polypeptide on neurons in apoptosis, we measured caspase activation in adult olfactory receptor neurons in vitro. Interestingly, we found that the protective effects of pituitary adenylate cyclase activating polypeptide were related to the absence of a 4-aminopyridine (IC50=144 microM) sensitive rapidly inactivating potassium current often referred to as A-type current. In the presence of 40 nM pituitary adenylate cyclase activating polypeptide 38, both A-type current and activated caspases were significantly reduced. A-type current reduction by pituitary adenylate cyclase activating polypeptide was blocked by inhibiting the phospholipase C pathway, but not the adenylyl cyclase pathway. Our observation that 5 mM 4-aminopyridine mimicked the caspase inhibiting effects of pituitary adenylate cyclase activating polypeptide indicates that A-type current is involved in apoptosis. This work contributes to our growing understanding that potassium currents are involved with the activation of caspases to affect the balance between cell life and death.
Collapse
Affiliation(s)
- P Han
- Interdepartmental Neuroscience Program, University of Utah, Salt Lake City, UT 84108, USA
| | | |
Collapse
|
49
|
Wagatsuma A, Sadamoto H, Kitahashi T, Lukowiak K, Urano A, Ito E. Determination of the exact copy numbers of particular mRNAs in a single cell by quantitative real-time RT-PCR. ACTA ACUST UNITED AC 2005; 208:2389-98. [PMID: 15939778 DOI: 10.1242/jeb.01625] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Gene expression is differently regulated in every cell even though the cells are included in the same tissue. For this reason, we need to measure the amount of mRNAs in a single cell to understand transcription mechanism better. However, there are no accurate, rapid and appropriate methods to determine the exact copy numbers of particular mRNAs in a single cell. We therefore developed a procedure for isolating a single, identifiable cell and determining the exact copy numbers of mRNAs within it. We first isolated the cerebral giant cell of the pond snail Lymnaea stagnalis as this neuron plays a key role in the process of memory consolidation of a learned behavior brought about by associative learning of feeding behavior. We then determined the copy numbers of mRNAs for the cyclic AMP-responsive element binding proteins (CREBs). These transcription factors play an important role in memory formation across animal species. The protocol uses two techniques in concert with each other: a technique for isolating a single neuron with newly developed micromanipulators coupled to an assay of mRNAs by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). The molecular assay determined the mRNA copy numbers, each of which was compared with a standard curve prepared from cDNA solutions corresponding to the serially diluted solutions of Lymnaea CREB mRNA. The standard curves were linear within a range of 10 to 10(5) copies, and the intra-assay variation was within 15%. Each neuron removed from the ganglia was punctured to extract the total RNA directly and was used for the assay without further purification. Using this two-step procedure, we found that the mRNA copy number of CREB repressor (CREB2) was 30-240 in a single cerebral giant cell, whereas that of CREB activator (CREB1) was below the detection limits of the assay (< 25). These results suggest that the CREB cascade is regulated by an excess amount of CREB2 in the cerebral giant cells. Our procedure is the only quantitative analysis for elucidation of the dynamics of gene transcription in a single cell.
Collapse
Affiliation(s)
- Akiko Wagatsuma
- Division of Biological Sciences, Graduate School of Science, Hokkaido University, North 10, West 8, Kita-ku, Sapporo 060-0810, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Christophe E, Doerflinger N, Lavery DJ, Molnár Z, Charpak S, Audinat E. Two populations of layer v pyramidal cells of the mouse neocortex: development and sensitivity to anesthetics. J Neurophysiol 2005; 94:3357-67. [PMID: 16000529 DOI: 10.1152/jn.00076.2005] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous studies have shown that layer V pyramidal neurons projecting either to subcortical structures or the contralateral cortex undergo different morphological and electrophysiological patterns of development during the first three postnatal weeks. To isolate the determinants of this differential maturation, we analyzed the gene expression and intrinsic membrane properties of layer V pyramidal neurons projecting either to the superior colliculus (SC cells) or the contralateral cortex (CC cells) by combining whole cell recordings and single-cell RT-PCR in acute slices prepared from postnatal day (P) 5-7 or P21-30 old mice. Among the 24 genes tested, the calcium channel subunits alpha1B and alpha1C, the protease Nexin 1, and the calcium-binding protein calbindin were differentially expressed in adult SC and CC cells and the potassium channel subunit Kv4.3 was expressed preferentially in CC cells at both stages of development. Intrinsic membrane properties, including input resistance, amplitude of the hyperpolarization-activated current, and action potential threshold, differed quantitatively between the two populations as early as from the first postnatal week and persisted throughout adulthood. However, the two cell types had similar regular action potential firing behaviors at all developmental stages. Surprisingly, when we increased the duration of anesthesia with ketamine-xylazine or pentobarbital before decapitation, a proportion of mature SC cells, but not CC cells, fired bursts of action potentials. Together these results indicate that the two populations of layer V pyramidal neurons already start to differ during the first postnatal week and exhibit different firing capabilities after anesthesia.
Collapse
Affiliation(s)
- Elodie Christophe
- Neurophysiology and New Microscopies Laboratory, INSERM U603, CNRS FRE 2500, Université Paris Descartes, 45 Rue des StPères, 75006 Paris, France
| | | | | | | | | | | |
Collapse
|