1
|
Sun H, Patil MJ, Ru F, Meeker S, Undem BJ. K
V
1/D‐type potassium channels inhibit the excitability of bronchopulmonary vagal afferent nerves. J Physiol 2022; 600:2953-2971. [PMID: 35430729 PMCID: PMC9203938 DOI: 10.1113/jp282803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract The KV1/D‐type potassium current (ID) is an important determinant of neuronal excitability. This study explored whether and how ID channels regulate the activation of bronchopulmonary vagal afferent nerves. The single‐neuron RT‐PCR assay revealed that nearly all mouse bronchopulmonary nodose neurons expressed the transcripts of α‐dendrotoxin (α‐DTX)‐sensitive, ID channel‐forming KV1.1, KV1.2 and/or KV1.6 α‐subunits, with the expression of KV1.6 being most prevalent. Patch‐clamp recordings showed that ID, defined as the α‐DTX‐sensitive K+ current, activated at voltages slightly more negative than the resting membrane potential in lung‐specific nodose neurons and displayed little inactivation at subthreshold voltages. Inhibition of ID channels by α‐DTX depolarized the lung‐specific nodose neurons and caused an increase in input resistance, decrease in rheobase, as well as increase in action potential number and firing frequency in response to suprathreshold current steps. Application of α‐DTX to the lungs via trachea in the mouse ex vivo vagally innervated trachea–lungs preparation led to action potential discharges in nearly half of bronchopulmonary nodose afferent nerve fibres, including nodose C‐fibres, as detected by the two‐photon microscopic Ca2+ imaging technique and extracellular electrophysiological recordings. In conclusion, ID channels act as a critical brake on the activation of bronchopulmonary vagal afferent nerves by stabilizing the membrane potential, counterbalancing the subthreshold depolarization and promoting the adaptation of action potential firings. Down‐regulation of ID channels, as occurs in various inflammatory diseases, may contribute to the enhanced C‐fibre activity in airway diseases that are associated with excessive coughing, dyspnoea, and reflex bronchospasm and secretions. Key points The α‐dendrotoxin (α‐DTX)‐sensitive D‐type K+ current (ID) is an important determinant of neuronal excitability. Nearly all bronchopulmonary nodose afferent neurons in the mouse express ID and the transcripts of α‐DTX‐sensitive, ID channel‐forming KV1.1, KV1.2 and/or KV1.6 α‐subunits. Inhibition of ID channels by α‐DTX depolarizes the bronchopulmonary nodose neurons, reduces the minimal depolarizing current needed to evoke an action potential (AP) and increases AP number and AP firing frequency in response to suprathreshold stimulations. Application of α‐DTX to the lungs ex vivo elicits AP discharges in about half of bronchopulmonary nodose C‐fibre terminals.
Our novel finding that ID channels act as a critical brake on the activation of bronchopulmonary vagal afferent nerves suggests that their down‐regulation, as occurs in various inflammatory diseases, may contribute to the enhanced C‐fibre activity in airway inflammation associated with excessive respiratory symptoms.
Collapse
Affiliation(s)
- Hui Sun
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Mayur J. Patil
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Fei Ru
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Sonya Meeker
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| | - Bradley J. Undem
- Division of Allergy and Clinical Immunology Department of Medicine Johns Hopkins University School of Medicine 5501 Hopkins Bayview Circle Baltimore 21224
| |
Collapse
|
2
|
Abstract
Transient outward potassium currents were first described nearly 60 years ago, since then major strides have been made in understanding their molecular basis and physiological roles. From the large family of voltage-gated potassium channels members of 3 subfamilies can produce such fast-inactivating A-type potassium currents. Each subfamily gives rise to currents with distinct biophysical properties and pharmacological profiles and a simple workflow is provided to aid the identification of channels mediating A-type currents in native cells. Their unique properties and regulation enable A-type K+ channels to perform varied roles in excitable cells including repolarisation of the cardiac action potential, controlling spike and synaptic timing, regulating dendritic integration and long-term potentiation as well as being a locus of neural plasticity.
Collapse
Affiliation(s)
- Jamie Johnston
- Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
3
|
Sun H. Different sensitivity of action potential generation to the rate of depolarization in vagal afferent A-fiber versus C-fiber neurons. J Neurophysiol 2021; 125:2000-2012. [PMID: 33881911 DOI: 10.1152/jn.00722.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This study demonstrates that the action potential discharge in vagal afferent A-fiber neurons is about 20 times more sensitive to the rate of membrane depolarization compared to C-fiber neurons. The sensitivity of action potential generation to the depolarization rate in vagal sensory neurons is independent of the intensity of current stimuli but nearly abrogated by inhibiting the D-type potassium channel. These findings help better understand the mechanisms that control the activation of vagal afferent nerves.
Collapse
Affiliation(s)
- Hui Sun
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Zhang L, Peng Z, Bian W, Zhu P, Tang B, Liao WP, Su T. Functional Differences Between Two Kv1.1 RNA Editing Isoforms: a Comparative Study on Neuronal Overexpression in Mouse Prefrontal Cortex. Mol Neurobiol 2021; 58:2046-2060. [PMID: 33411244 DOI: 10.1007/s12035-020-02229-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
The Shaker-related potassium channel Kv1.1 subunit has important implications for controlling neuronal excitabilities. A particular recoding by A-to-I RNA editing at I400 of Kv1.1 mRNA is an underestimated mechanism for fine-tuning the properties of Kv1.1-containing channels. Knowledge about functional differences between edited (I400V) and non-edited Kv1.1 isoforms is insufficient, especially in neurons. To understand their different roles, the two Kv1.1 isoforms were overexpressed in the prefrontal cortex via local adeno-associated virus-mediated gene delivery. The I400V isoform showed a higher competitiveness in membrane translocalization, but failed to reduce current-evoked discharges and showed weaker impact on spiking-frequency adaptation in the transduced neurons. The non-edited Kv1.1 overexpression led to slight elevations in both fast- and non-inactivating current components of macroscopic potassium current. By contrast, the I400V overexpression did not impact the fast-inactivating current component. Further isolation of Kv1.1-specific current by its specific blocker dendrotoxin-κ showed that both isoforms did result in significant increases in current amplitude, whereas the I400V was less efficient in contributing the fast-inactivating current component. Voltage-dependent properties of the fast-inactivating current component did not alter for both isoforms. For recovery kinetics, the I400V showed a significant acceleration of recovery from fast inactivation. The gene delivery of the I400V rather than the wild type exhibited anxiolytic activities, which was assessed by an open field test. These results suggest that the Kv1.1 RNA editing isoforms have different properties and outcomes, reflecting the functional and phenotypic significance of the Kv1.1 RNA editing in neurons.
Collapse
Affiliation(s)
- Liting Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Zetong Peng
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wenjun Bian
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Pingping Zhu
- Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Bin Tang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wei-Ping Liao
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China.,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Tao Su
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Chang-gang-dong Road 250, Guangzhou, 510260, China. .,Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China.
| |
Collapse
|
5
|
Assembly and Function of the Juxtaparanodal Kv1 Complex in Health and Disease. Life (Basel) 2020; 11:life11010008. [PMID: 33374190 PMCID: PMC7824554 DOI: 10.3390/life11010008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The precise axonal distribution of specific potassium channels is known to secure the shape and frequency of action potentials in myelinated fibers. The low-threshold voltage-gated Kv1 channels located at the axon initial segment have a significant influence on spike initiation and waveform. Their role remains partially understood at the juxtaparanodes where they are trapped under the compact myelin bordering the nodes of Ranvier in physiological conditions. However, the exposure of Kv1 channels in de- or dys-myelinating neuropathy results in alteration of saltatory conduction. Moreover, cell adhesion molecules associated with the Kv1 complex, including Caspr2, Contactin2, and LGI1, are target antigens in autoimmune diseases associated with hyperexcitability such as encephalitis, neuromyotonia, or neuropathic pain. The clustering of Kv1.1/Kv1.2 channels at the axon initial segment and juxtaparanodes is based on interactions with cell adhesion molecules and cytoskeletal linkers. This review will focus on the trafficking and assembly of the axonal Kv1 complex in the peripheral and central nervous system (PNS and CNS), during development, and in health and disease.
Collapse
|
6
|
Abstract
Voltage-gated Kv1.1 potassium channel α-subunits are broadly expressed in the nervous system where they act as critical regulators of neuronal excitability. Mutations in the KCNA1 gene, which encodes Kv1.1, are associated with the neurological diseases episodic ataxia and epilepsy. Studies in mouse models have shown that Kv1.1 is important for neural control of the heart and that Kcna1 deletion leads to cardiac dysfunction that appears to be brain-driven. Traditionally, KCNA1 was not believed to be expressed in the heart. However, recent studies have revealed that Kv1.1 subunits are not only present in cardiomyocytes, but that they also make an important heart-intrinsic functional contribution to outward K+ currents and action potential repolarization. This review recounts the winding history of discovery of KCNA1 gene expression and neurocardiac function from fruit flies to mammals and from brain to heart and looks at some of the salient questions that remain to be answered regarding emerging cardiac roles of Kv1.1.
Collapse
Affiliation(s)
- Edward Glasscock
- a Department of Biological Sciences , Southern Methodist University , Dallas , TX , USA
| |
Collapse
|
7
|
Al-Sabi A, Daly D, Rooney M, Hughes C, Kinsella GK, Kaza SK, Nolan K, Oliver Dolly J. Development of a selective inhibitor for Kv1.1 channels prevalent in demyelinated nerves. Bioorg Chem 2020; 100:103918. [PMID: 32428746 DOI: 10.1016/j.bioorg.2020.103918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 03/23/2020] [Accepted: 05/05/2020] [Indexed: 11/19/2022]
Abstract
Members of the voltage-gated K+ channel subfamily (Kv1), involved in regulating transmission between neurons or to muscles, are associated with human diseases and, thus, putative targets for neurotherapeutics. This applies especially to those containing Kv1.1 α subunits which become prevalent in murine demyelinated axons and appear abnormally at inter-nodes, underlying the perturbed propagation of nerve signals. To overcome this dysfunction, akin to the consequential debilitation in multiple sclerosis (MS), small inhibitors were sought that are selective for the culpable hyper-polarising K+ currents. Herein, we report a new semi-podand - compound 3 - that was designed based on the modelling of its interactions with the extracellular pore region in a deduced Kv1.1 channel structure. After synthesis, purification, and structural characterisation, compound 3 was found to potently (IC50 = 8 µM) and selectively block Kv1.1 and 1.6 channels. The tested compound showed no apparent effect on native Nav and Cav channels expressed in F-11 cells. Compound 3 also extensively and selectively inhibited MS-related Kv1.1 homomer but not the brain native Kv1.1- or 1.6-containing channels. These collective findings highlight the therapeutic potential of compound 3 to block currents mediated by Kv1.1 channels enriched in demyelinated central neurons.
Collapse
Affiliation(s)
- Ahmed Al-Sabi
- College of Engineering and Technology, American University of the Middle East, Kuwait; International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Declan Daly
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Myles Rooney
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Cian Hughes
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Gemma K Kinsella
- School of Food Science and Environmental Health, College of Sciences and Health, Technological University Dublin, Cathal Brugha Street, Dublin 1, Ireland
| | - Seshu K Kaza
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Kieran Nolan
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - J Oliver Dolly
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
8
|
Si M, Trosclair K, Hamilton KA, Glasscock E. Genetic ablation or pharmacological inhibition of Kv1.1 potassium channel subunits impairs atrial repolarization in mice. Am J Physiol Cell Physiol 2019; 316:C154-C161. [PMID: 30427720 PMCID: PMC6397341 DOI: 10.1152/ajpcell.00335.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 12/14/2022]
Abstract
Voltage-gated Kv1.1 potassium channel α-subunits, encoded by the Kcna1 gene, have traditionally been regarded as neural-specific with no expression or function in the heart. However, recent data revealed that Kv1.1 subunits are expressed in atria where they may have an overlooked role in controlling repolarization and arrhythmia susceptibility independent of the nervous system. To explore this concept in more detail and to identify functional and molecular effects of Kv1.1 channel impairment in the heart, atrial cardiomyocyte patch-clamp electrophysiology and gene expression analyses were performed using Kcna1 knockout ( Kcna1-/-) mice. Specifically, we hypothesized that Kv1.1 subunits contribute to outward repolarizing K+ currents in mouse atria and that their absence prolongs cardiac action potentials. In voltage-clamp experiments, dendrotoxin-K (DTX-K), a Kv1.1-specific inhibitor, significantly reduced peak outward K+ currents in wild-type (WT) atrial cells but not Kcna1-/- cells, demonstrating an important contribution by Kv1.1-containing channels to mouse atrial repolarizing currents. In current-clamp recordings, Kcna1-/- atrial myocytes exhibited significant action potential prolongation which was exacerbated in right atria, effects that were partially recapitulated in WT cells by application of DTX-K. Quantitative RT-PCR measurements showed mRNA expression remodeling in Kcna1-/- atria for several ion channel genes that contribute to the atrial action potential including the Kcna5, Kcnh2, and Kcnj2 potassium channel genes and the Scn5a sodium channel gene. This study demonstrates a previously undescribed heart-intrinsic role for Kv1.1 subunits in mediating atrial repolarization, thereby adding a new member to the already diverse collection of known K+ channels in the heart.
Collapse
Affiliation(s)
- Man Si
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| | - Krystle Trosclair
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| | - Kathryn A Hamilton
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| | - Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center , Shreveport, Louisiana
| |
Collapse
|
9
|
Neurobiology and therapeutic applications of neurotoxins targeting transmitter release. Pharmacol Ther 2019; 193:135-155. [DOI: 10.1016/j.pharmthera.2018.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
10
|
Zhu P, Li J, Zhang L, Liang Z, Tang B, Liao WP, Yi YH, Su T. Development-related aberrations in Kv1.1 α-subunit exert disruptive effects on bioelectrical activities of neurons in a mouse model of fragile X syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:140-151. [PMID: 29481897 DOI: 10.1016/j.pnpbp.2018.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 02/22/2018] [Accepted: 02/22/2018] [Indexed: 10/18/2022]
Abstract
Kv1.1, a Shaker homologue potassium channel, plays a critical role in homeostatic regulation of neuronal excitability. Aberrations in the functional properties of Kv1.1 have been implicated in several neurological disorders featured by neuronal hyperexcitability. Fragile X syndrome (FXS), the most common form of inherited mental retardation, is characterized by hyperexcitability in neural network and intrinsic membrane properties. The Kv1.1 channel provides an intriguing mechanistic candidate for FXS. We investigated the development-related expression pattern of the Kv1.1 α-subunit by using a Fmr1 knockout (KO) mouse model of FXS. Markedly decreased protein expression of Kv1.1 was found in neonatal and adult stages when compared to age-matched wild-type (WT) mice. Immunohistochemical investigations supported the delayed development-related increases in Kv1.1 expression, especially in CA3 pyramidal neurons. By applying a Kv1.1-specific blocker, dendrotoxin-κ (DTX-κ), we isolated the Kv1.1-mediated currents in the CA3 pyramidal neurons. The isolated DTX-κ-sensitive current of neurons from KO mice exhibited decreased amplitude, lower threshold of activation, and faster recovery from inactivation. The equivalent reduction in potassium current in the WT neurons following application of the appropriate amount of DTX-κ reproduced the enhanced firing abilities of KO neurons, suggesting the Kv1.1 channel as a critical contributor to the hyperexcitability of KO neurons. The role of Kv1.1 in controlling neuronal discharges was further supported by the parallel developmental trajectories of Kv1.1 expression, current amplitude, and discharge impacts, with a significant correlation between the amplitude of Kv1.1-mediated currents and Kv1.1-blocking-induced firing enhancement. These data suggest that the expression of the Kv1.1 α-subunit has a profound pathological relevance to hyperexcitability in FXS, as well as implications for normal development, maintenance, and control of neuronal activities.
Collapse
Affiliation(s)
- Pingping Zhu
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China; Department of Neurology, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Jialing Li
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Liting Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Zhanrong Liang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Bin Tang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wei-Ping Liao
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Yong-Hong Yi
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Tao Su
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China.
| |
Collapse
|
11
|
Guan D, Pathak D, Foehring RC. Functional roles of Kv1-mediated currents in genetically identified subtypes of pyramidal neurons in layer 5 of mouse somatosensory cortex. J Neurophysiol 2018; 120:394-408. [PMID: 29641306 DOI: 10.1152/jn.00691.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We used voltage-clamp recordings from somatic outside-out macropatches to determine the amplitude and biophysical properties of putative Kv1-mediated currents in layer 5 pyramidal neurons (PNs) from mice expressing EGFP under the control of promoters for etv1 or glt. We then used whole cell current-clamp recordings and Kv1-specific peptide blockers to test the hypothesis that Kv1 channels differentially regulate action potential (AP) voltage threshold, repolarization rate, and width as well as rheobase and repetitive firing in these two PN types. We found that Kv1-mediated currents make up a similar percentage of whole cell K+ current in both cell types, and only minor biophysical differences were observed between PN types or between currents sensitive to different Kv1 blockers. Putative Kv1 currents contributed to AP voltage threshold in both PN types, but AP width and rate of repolarization were only affected in etv1 PNs. Kv1 currents regulate rheobase, delay to the first AP, and firing rate similarly in both cell types, but the frequency-current slope was much more sensitive to Kv1 block in etv1 PNs. In both cell types, Kv1 block shifted the current required to elicit an onset doublet of action potentials to lower currents. Spike frequency adaptation was also affected differently by Kv1 block in the two PN types. Thus, despite similar expression levels and minimal differences in biophysical properties, Kv1 channels differentially regulate APs and repetitive firing in etv1 and glt PNs. This may reflect differences in subcellular localization of channel subtypes or differences in the other K+ channels expressed. NEW & NOTEWORTHY In two types of genetically identified layer 5 pyramidal neurons, α-dendrotoxin blocked approximately all of the putative Kv1 current (on average). We used outside-out macropatches and whole cell recordings at 33°C to show that despite similar expression levels and minimal differences in biophysical properties, Kv1 channels differentially regulate action potentials and repetitive firing in etv1 and glt pyramidal neurons. This may reflect differences in subcellular localization of channel subtypes or differences in the other K+ channels expressed.
Collapse
Affiliation(s)
- Dongxu Guan
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Dhruba Pathak
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Robert C Foehring
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
12
|
Cao XJ, Oertel D. Genetic perturbations suggest a role of the resting potential in regulating the expression of the ion channels of the KCNA and HCN families in octopus cells of the ventral cochlear nucleus. Hear Res 2017; 345:57-68. [PMID: 28065805 DOI: 10.1016/j.heares.2017.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 10/20/2022]
Abstract
Low-voltage-activated K+ (gKL) and hyperpolarization-activated mixed cation conductances (gh) mediate currents, IKL and Ih, through channels of the Kv1 (KCNA) and HCN families respectively and give auditory neurons the temporal precision required for signaling information about the onset, fine structure, and time of arrival of sounds. Being partially activated at rest, gKL and gh contribute to the resting potential and shape responses to even small subthreshold synaptic currents. Resting gKL and gh also affect the coupling of somatic depolarization with the generation of action potentials. To learn how these important conductances are regulated we have investigated how genetic perturbations affect their expression in octopus cells of the ventral cochlear nucleus (VCN). We report five new findings: First, the magnitude of gh and gKL varied over more than two-fold between wild type strains of mice. Second, average resting potentials are not different in different strains of mice even in the face of large differences in average gKL and gh. Third, IKL has two components, one being α-dendrotoxin (α-DTX)-sensitive and partially inactivating and the other being α-DTX-insensitive, tetraethylammonium (TEA)-sensitive, and non-inactivating. Fourth, the loss of Kv1.1 results in diminution of the α-DTX-sensitive IKL, and compensatory increased expression of an α-DTX-insensitive, tetraethylammonium (TEA)-sensitive IKL. Fifth, Ih and IKL are balanced at the resting potential in all wild type and mutant octopus cells even when resting potentials vary in individual cells over nearly 10 mV, indicating that the resting potential influences the expression of gh and gKL. The independence of resting potentials on gKL and gh shows that gKL and gh do not, over days or weeks, determine the resting potential but rather that the resting potential plays a role in regulating the magnitude of either or both gKL and gh.
Collapse
Affiliation(s)
- Xiao-Jie Cao
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Donata Oertel
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
13
|
KV1 and KV3 Potassium Channels Identified at Presynaptic Terminals of the Corticostriatal Synapses in Rat. Neural Plast 2016; 2016:8782518. [PMID: 27379187 PMCID: PMC4917754 DOI: 10.1155/2016/8782518] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/12/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022] Open
Abstract
In the last years it has been increasingly clear that KV-channel activity modulates neurotransmitter release. The subcellular localization and composition of potassium channels are crucial to understanding its influence on neurotransmitter release. To investigate the role of KV in corticostriatal synapses modulation, we combined extracellular recording of population-spike and pharmacological blockage with specific and nonspecific blockers to identify several families of KV channels. We induced paired-pulse facilitation (PPF) and studied the changes in paired-pulse ratio (PPR) before and after the addition of specific KV blockers to determine whether particular KV subtypes were located pre- or postsynaptically. Initially, the presence of KV channels was tested by exposing brain slices to tetraethylammonium or 4-aminopyridine; in both cases we observed a decrease in PPR that was dose dependent. Further experiments with tityustoxin, margatoxin, hongotoxin, agitoxin, dendrotoxin, and BDS-I toxins all rendered a reduction in PPR. In contrast heteropodatoxin and phrixotoxin had no effect. Our results reveal that corticostriatal presynaptic KV channels have a complex stoichiometry, including heterologous combinations KV1.1, KV1.2, KV1.3, and KV1.6 isoforms, as well as KV3.4, but not KV4 channels. The variety of KV channels offers a wide spectrum of possibilities to regulate neurotransmitter release, providing fine-tuning mechanisms to modulate synaptic strength.
Collapse
|
14
|
Rash JE, Vanderpool KG, Yasumura T, Hickman J, Beatty JT, Nagy JI. KV1 channels identified in rodent myelinated axons, linked to Cx29 in innermost myelin: support for electrically active myelin in mammalian saltatory conduction. J Neurophysiol 2016; 115:1836-59. [PMID: 26763782 PMCID: PMC4869480 DOI: 10.1152/jn.01077.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 11/22/2022] Open
Abstract
Saltatory conduction in mammalian myelinated axons was thought to be well understood before recent discoveries revealed unexpected subcellular distributions and molecular identities of the K(+)-conductance pathways that provide for rapid axonal repolarization. In this study, we visualize, identify, localize, quantify, and ultrastructurally characterize axonal KV1.1/KV1.2 channels in sciatic nerves of rodents. With the use of light microscopic immunocytochemistry and freeze-fracture replica immunogold labeling electron microscopy, KV1.1/KV1.2 channels are localized to three anatomically and compositionally distinct domains in the internodal axolemmas of large myelinated axons, where they form densely packed "rosettes" of 9-nm intramembrane particles. These axolemmal KV1.1/KV1.2 rosettes are precisely aligned with and ultrastructurally coupled to connexin29 (Cx29) channels, also in matching rosettes, in the surrounding juxtaparanodal myelin collars and along the inner mesaxon. As >98% of transmembrane proteins large enough to represent ion channels in these specialized domains, ∼500,000 KV1.1/KV1.2 channels define the paired juxtaparanodal regions as exclusive membrane domains for the voltage-gated K(+)conductance that underlies rapid axonal repolarization in mammals. The 1:1 molecular linkage of KV1 channels to Cx29 channels in the apposed juxtaparanodal collars, plus their linkage to an additional 250,000-400,000 Cx29 channels along each inner mesaxon in every large-diameter myelinated axon examined, supports previously proposed K(+)conductance directly from juxtaparanodal axoplasm into juxtaparanodal myeloplasm in mammalian axons. With neither Cx29 protein nor myelin rosettes detectable in frog myelinated axons, these data showing axon-to-myelin linkage by abundant KV1/Cx29 channels in rodent axons support renewed consideration of an electrically active role for myelin in increasing both saltatory conduction velocity and maximum propagation frequency in mammalian myelinated axons.
Collapse
Affiliation(s)
- John E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado; Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, Colorado; and
| | - Kimberly G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas Yasumura
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jordan Hickman
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jonathan T Beatty
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - James I Nagy
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
15
|
Abstract
UNLABELLED The role of interneurons in cortical microcircuits is strongly influenced by their passive and active electrical properties. Although different types of interneurons exhibit unique electrophysiological properties recorded at the soma, it is not yet clear whether these differences are also manifested in other neuronal compartments. To address this question, we have used voltage-sensitive dye to image the propagation of action potentials into the fine collaterals of axons and dendrites in two of the largest cortical interneuron subtypes in the mouse: fast-spiking interneurons, which are typically basket or chandelier neurons; and somatostatin containing interneurons, which are typically regular spiking Martinotti cells. We found that fast-spiking and somatostatin-expressing interneurons differed in their electrophysiological characteristics along their entire dendrosomatoaxonal extent. The action potentials generated in the somata and axons, including axon collaterals, of somatostatin-expressing interneurons are significantly broader than those generated in the same compartments of fast-spiking inhibitory interneurons. In addition, action potentials back-propagated into the dendrites of somatostatin-expressing interneurons much more readily than fast-spiking interneurons. Pharmacological investigations suggested that axonal action potential repolarization in both cell types depends critically upon Kv1 channels, whereas the axonal and somatic action potentials of somatostatin-expressing interneurons also depend on BK Ca(2+)-activated K(+) channels. These results indicate that the two broad classes of interneurons studied here have expressly different subcellular physiological properties, allowing them to perform unique computational roles in cortical circuit operations. SIGNIFICANCE STATEMENT Neurons in the cerebral cortex are of two major types: excitatory and inhibitory. The proper balance of excitation and inhibition in the brain is critical for its operation. Neurons contain three main compartments: dendritic, somatic, and axonal. How the neurons receive information, process it, and pass on new information depends upon how these three compartments operate. While it has long been assumed that axons are simply for conducting information from the cell body to the synapses, here we demonstrate that the axons of different types of interneurons, the inhibitory cells, possess differing electrophysiological properties. This result implies that differing types of interneurons perform different tasks in the cortex, not only through their anatomical connections, but also through how their axons operate.
Collapse
|
16
|
Ovsepian SV, LeBerre M, Steuber V, O'Leary VB, Leibold C, Oliver Dolly J. Distinctive role of KV1.1 subunit in the biology and functions of low threshold K+ channels with implications for neurological disease. Pharmacol Ther 2016; 159:93-101. [DOI: 10.1016/j.pharmthera.2016.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
17
|
Glasscock E, Voigt N, McCauley MD, Sun Q, Li N, Chiang DY, Zhou XB, Molina CE, Thomas D, Schmidt C, Skapura DG, Noebels JL, Dobrev D, Wehrens XHT. Expression and function of Kv1.1 potassium channels in human atria from patients with atrial fibrillation. Basic Res Cardiol 2015; 110:505. [PMID: 26162324 DOI: 10.1007/s00395-015-0505-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 07/02/2015] [Accepted: 07/03/2015] [Indexed: 11/24/2022]
Abstract
Voltage-gated Kv1.1 channels encoded by the Kcna1 gene are traditionally regarded as being neural-specific with no known expression or intrinsic functional role in the heart. However, recent studies in mice reveal low-level Kv1.1 expression in heart and cardiac abnormalities associated with Kv1.1-deficiency suggesting that the channel may have a previously unrecognized cardiac role. Therefore, this study tests the hypothesis that Kv1.1 channels are associated with arrhythmogenesis and contribute to intrinsic cardiac function. In intra-atrial burst pacing experiments, Kcna1-null mice exhibited increased susceptibility to atrial fibrillation (AF). The atria of Kcna1-null mice showed minimal Kv1 family ion channel remodeling and fibrosis as measured by qRT-PCR and Masson's trichrome histology, respectively. Using RT-PCR, immunocytochemistry, and immunoblotting, KCNA1 mRNA and protein were detected in isolated mouse cardiomyocytes and human atria for the first time. Patients with chronic AF (cAF) showed no changes in KCNA1 mRNA levels relative to controls; however, they exhibited increases in atrial Kv1.1 protein levels, not seen in paroxysmal AF patients. Patch-clamp recordings of isolated human atrial myocytes revealed significant dendrotoxin-K (DTX-K)-sensitive outward current components that were significantly increased in cAF patients, reflecting a contribution by Kv1.1 channels. The concomitant increases in Kv1.1 protein and DTX-K-sensitive currents in atria of cAF patients suggest that the channel contributes to the pathological mechanisms of persistent AF. These findings provide evidence of an intrinsic cardiac role of Kv1.1 channels and indicate that they may contribute to atrial repolarization and AF susceptibility.
Collapse
Affiliation(s)
- Edward Glasscock
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Highway, P.O. Box 33932, Shreveport, LA, 71130-393, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Daly D, Al-Sabi A, Kinsella GK, Nolan K, Dolly JO. Porphyrin derivatives as potent and selective blockers of neuronal Kv1 channels. Chem Commun (Camb) 2015; 51:1066-9. [DOI: 10.1039/c4cc05639f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Docking model of compound12into the homology model of Kv1.1.
Collapse
Affiliation(s)
- D. Daly
- School of Chemical Sciences
- Dublin City University
- Dublin 9
- Ireland
| | - A. Al-Sabi
- International Centre for Neurotherapeutics
- Dublin City University
- Dublin 9
- Ireland
| | - G. K. Kinsella
- Biology Department
- National University of Ireland Maynooth
- Co. Kildare
- Ireland
| | - K. Nolan
- School of Chemical Sciences
- Dublin City University
- Dublin 9
- Ireland
| | - J. O. Dolly
- International Centre for Neurotherapeutics
- Dublin City University
- Dublin 9
- Ireland
| |
Collapse
|
19
|
Bagchi B, Al-Sabi A, Kaza S, Scholz D, O'Leary VB, Dolly JO, Ovsepian SV. Disruption of myelin leads to ectopic expression of K(V)1.1 channels with abnormal conductivity of optic nerve axons in a cuprizone-induced model of demyelination. PLoS One 2014; 9:e87736. [PMID: 24498366 PMCID: PMC3912067 DOI: 10.1371/journal.pone.0087736] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 12/30/2013] [Indexed: 11/19/2022] Open
Abstract
The molecular determinants of abnormal propagation of action potentials along axons and ectopic conductance in demyelinating diseases of the central nervous system, like multiple sclerosis (MS), are poorly defined. Widespread interruption of myelin occurs in several mouse models of demyelination, rendering them useful for research. Herein, considerable myelin loss is shown in the optic nerves of cuprizone-treated demyelinating mice. Immuno-fluorescence confocal analysis of the expression and distribution of voltage-activated K⁺ channels (K(V)1.1 and 1.2 α subunits) revealed their spread from typical juxta-paranodal (JXP) sites to nodes in demyelinated axons, albeit with a disproportionate increase in the level of K(V)1.1 subunit. Functionally, in contrast to monophasic compound action potentials (CAPs) recorded in controls, responses derived from optic nerves of cuprizone-treated mice displayed initial synchronous waveform followed by a dispersed component. Partial restoration of CAPs by broad spectrum (4-aminopyridine) or K(V)1.1-subunit selective (dendrotoxin K) blockers of K⁺ currents suggest enhanced K(V)1.1-mediated conductance in the demyelinated optic nerve. Biophysical profiling of K⁺ currents mediated by recombinant channels comprised of different K(V)1.1 and 1.2 stoichiometries revealed that the enrichment of K(V)1 channels K(V)1.1 subunit endows a decrease in the voltage threshold and accelerates the activation kinetics. Together with the morphometric data, these findings provide important clues to a molecular basis for temporal dispersion of CAPs and reduced excitability of demyelinated optic nerves, which could be of potential relevance to the patho-physiology of MS and related disorders.
Collapse
Affiliation(s)
- Bandita Bagchi
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin, Republic of Ireland
| | - Ahmed Al-Sabi
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin, Republic of Ireland
| | - Seshu Kaza
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin, Republic of Ireland
| | - Dimitri Scholz
- Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Valerie B. O'Leary
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin, Republic of Ireland
| | - J. Oliver Dolly
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin, Republic of Ireland
- * E-mail: (SVO); (JOD)
| | - Saak V. Ovsepian
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin, Republic of Ireland
- Department of Biotechnology, Dublin City University, Glasnevin, Dublin, Republic of Ireland
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ludwig-Maximilians-Universität München, Zentrum für Neuropathologie, Feodor-Lynen-Str. 23, Munich, Germany
- * E-mail: (SVO); (JOD)
| |
Collapse
|
20
|
Kirchheim F, Tinnes S, Haas CA, Stegen M, Wolfart J. Regulation of action potential delays via voltage-gated potassium Kv1.1 channels in dentate granule cells during hippocampal epilepsy. Front Cell Neurosci 2013; 7:248. [PMID: 24367293 PMCID: PMC3852106 DOI: 10.3389/fncel.2013.00248] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 11/20/2013] [Indexed: 11/13/2022] Open
Abstract
Action potential (AP) responses of dentate gyrus granule (DG) cells have to be tightly regulated to maintain hippocampal function. However, which ion channels control the response delay of DG cells is not known. In some neuron types, spike latency is influenced by a dendrotoxin (DTX)-sensitive delay current (ID) mediated by unidentified combinations of voltage-gated K(+) (Kv) channels of the Kv1 family Kv1.1-6. In DG cells, the ID has not been characterized and its molecular basis is unknown. The response phenotype of mature DG cells is usually considered homogenous but intrinsic plasticity likely occurs in particular in conditions of hyperexcitability, for example during temporal lobe epilepsy (TLE). In this study, we examined response delays of DG cells and underlying ion channel molecules by employing a combination of gramicidin-perforated patch-clamp recordings in acute brain slices and single-cell reverse transcriptase quantitative polymerase chain reaction (SC RT-qPCR) experiments. An in vivo mouse model of TLE consisting of intrahippocampal kainate (KA) injection was used to examine epilepsy-related plasticity. Response delays of DG cells were DTX-sensitive and strongly increased in KA-injected hippocampi; Kv1.1 mRNA was elevated 10-fold, and the response delays correlated with Kv1.1 mRNA abundance on the single cell level. Other Kv1 subunits did not show overt changes in mRNA levels. Kv1.1 immunolabeling was enhanced in KA DG cells. The biophysical properties of ID and a delay heterogeneity within the DG cell population was characterized. Using organotypic hippocampal slice cultures (OHCs), where KA incubation also induced ID upregulation, the homeostatic reversibility and neuroprotective potential for DG cells were tested. In summary, the AP timing of DG cells is effectively controlled via scaling of Kv1.1 subunit transcription. With this antiepileptic mechanism, DG cells delay their responses during hyperexcitation.
Collapse
Affiliation(s)
- Florian Kirchheim
- Cellular Neurophysiology, Department of Neurosurgery, University Medical Center Freiburg Freiburg, Germany ; Faculty of Biology, University of Freiburg Freiburg, Germany
| | - Stefanie Tinnes
- Experimental Epilepsy Research, Department of Neurosurgery, University Medical Center Freiburg Freiburg, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, University Medical Center Freiburg Freiburg, Germany
| | - Michael Stegen
- Cellular Neurophysiology, Department of Neurosurgery, University Medical Center Freiburg Freiburg, Germany ; Department of Biomedicine, Institute of Physiology, University of Basel Basel, Switzerland
| | - Jakob Wolfart
- Cellular Neurophysiology, Department of Neurosurgery, University Medical Center Freiburg Freiburg, Germany ; Oscar Langendorff Institute of Physiology, University of Rostock Rostock, Germany
| |
Collapse
|
21
|
Hao J, Padilla F, Dandonneau M, Lavebratt C, Lesage F, Noël J, Delmas P. Kv1.1 Channels Act as Mechanical Brake in the Senses of Touch and Pain. Neuron 2013; 77:899-914. [DOI: 10.1016/j.neuron.2012.12.035] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
|
22
|
Bildl W, Haupt A, Müller CS, Biniossek ML, Thumfart JO, Hüber B, Fakler B, Schulte U. Extending the dynamic range of label-free mass spectrometric quantification of affinity purifications. Mol Cell Proteomics 2011; 11:M111.007955. [PMID: 22067099 DOI: 10.1074/mcp.m111.007955] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Affinity purification (AP) of protein complexes combined with LC-MS/MS analysis is the current method of choice for identification of protein-protein interactions. Their interpretation with respect to significance, specificity, and selectivity requires quantification methods coping with enrichment factors of more than 1000-fold, variable amounts of total protein, and low abundant, unlabeled samples. We used standardized samples (0.1-1000 fmol) measured on high resolution hybrid linear ion trap instruments (LTQ-FT/Orbitrap) to characterize and improve linearity and dynamic range of label-free approaches. Quantification based on spectral counts was limited by saturation and ion suppression effects with samples exceeding 100 ng of protein, depending on the instrument setup. In contrast, signal intensities of peptides (peak volumes) selected by a novel correlation-based method (TopCorr-PV) were linear over at least 4 orders of magnitude and allowed for accurate relative quantification of standard proteins spiked into a complex protein background. Application of this procedure to APs of the voltage-gated potassium channel Kv1.1 as a model membrane protein complex unambiguously identified the whole set of known interaction partners together with novel candidates. In addition to discriminating these proteins from background, we could determine efficiency, cross-reactivities, and selection biases of the used purification antibodies. The enhanced dynamic range of the developed quantification procedure appears well suited for sensitive identification of specific protein-protein interactions, detection of antibody-related artifacts, and optimization of AP conditions.
Collapse
Affiliation(s)
- Wolfgang Bildl
- Institute of Physiology, University of Freiburg, 79104 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Baker MD, Chen YC, Shah SU, Okuse K. In vitro and intrathecal siRNA mediated K(V)1.1 knock-down in primary sensory neurons. Mol Cell Neurosci 2011; 48:258-65. [PMID: 21903165 PMCID: PMC3240745 DOI: 10.1016/j.mcn.2011.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 08/16/2011] [Accepted: 08/19/2011] [Indexed: 11/28/2022] Open
Abstract
K(V)1.1 is a Shaker homologue K(+) channel that contributes to the juxta-paranodal membrane conductance in myelinated axons, and is blocked by fampridine (4-aminopyridine), used to treat the symptoms of multiple sclerosis. The present experiments investigate K(V)1.1 function in primary sensory neurons and A-fibres, and help define its characteristics as a drug-target using sequence specific small-interfering RNAs (siRNAs). siRNA (71nM) was used to knock-down functional expression of K(V)1.1 in sensory neurons (>25μm in apparent diameter) in culture, and was also delivered intrathecally in vivo (9.3μg). K(+) channel knock-down in sensory neurons was found to make the voltage-threshold for action potential generation significantly more negative than in control (p=0.02), led to the breakdown of accommodation and promoted spontaneous action potential firing. Exposure to dendrotoxin-K (DTX-K, 10-100nM) also selectively abolished K(+) currents at negative potentials and made voltage-threshold more negative, consistent with K(V)1.1 controlling excitability close to the nominal resting potential of the neuron cell body, near -60mV. Introduction of one working siRNA sequence into the intrathecal space in vivo was associated with a small increase in the amplitude of the depolarising after-potential in sacral spinal roots (p<0.02), suggesting a reduction in the number of working K(+) channels in internodal axon membrane. Our study provides evidence that K(V)1.1 contributes to the control of peripheral sensory nerve excitability, and suggests that its characteristics as a putative drug target can be assessed by siRNA transfection in primary sensory neurons in vitro and in vivo.
Collapse
Affiliation(s)
- Mark D Baker
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Neuroscience and Trauma Centre, Blizard Institute of Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK.
| | | | | | | |
Collapse
|
24
|
Cao XJ, Oertel D. The magnitudes of hyperpolarization-activated and low-voltage-activated potassium currents co-vary in neurons of the ventral cochlear nucleus. J Neurophysiol 2011; 106:630-40. [PMID: 21562186 DOI: 10.1152/jn.00015.2010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the ventral cochlear nucleus (VCN), neurons have hyperpolarization-activated conductances, which in some cells are enormous, that contribute to the ability of neurons to convey acoustic information in the timing of their firing by decreasing the input resistance and speeding-up voltage changes. Comparisons of the electrophysiological properties of neurons in the VCN of mutant mice that lack the hyperpolarization-activated cyclic nucleotide-gated channel α subunit 1 (HCN1(-/-)) (Nolan et al. 2003) with wild-type controls (HCN1(+/+)) and with outbred ICR mice reveal that octopus, T stellate, and bushy cells maintain their electrophysiological distinctions in all strains. Hyperpolarization-activated (I(h)) currents were smaller and slower, input resistances were higher, and membrane time constants were longer in HCN1(-/-) than in HCN1(+/+) in octopus, bushy, and T stellate cells. There were significant differences in the average magnitudes of I(h), input resistances, and time constants between HCN1(+/+) and ICR mice, but the resting potentials did not differ between strains. I(h) is opposed by a low-voltage-activated potassium (I(KL)) current in bushy and octopus cells, whose magnitudes varied widely between neuronal types and between strains. The magnitudes of I(h) and I(KL) were correlated across neuronal types and across mouse strains. Furthermore, these currents balanced one another at the resting potential in individual cells. The magnitude of I(h) and I(KL) is linked in bushy and octopus cells and varies not only between HCN1(-/-) and HCN1(+/+) but also between "wild-type" strains of mice, raising the question to what extent the wild-type strains reflect normal mice.
Collapse
Affiliation(s)
- Xiao-Jie Cao
- Department of Neuroscience, School of Neuroscience Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
| | | |
Collapse
|
25
|
Gazula VR, Strumbos JG, Mei X, Chen H, Rahner C, Kaczmarek LK. Localization of Kv1.3 channels in presynaptic terminals of brainstem auditory neurons. J Comp Neurol 2010; 518:3205-20. [PMID: 20575068 DOI: 10.1002/cne.22393] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Elimination of the Kv1.3 voltage-dependent potassium channel gene produces striking changes in the function of the olfactory bulb, raising the possibility that this channel also influences other sensory systems. We have examined the cellular and subcellular localization of Kv1.3 in the medial nucleus of the trapezoid body (MNTB) in the auditory brainstem, a nucleus in which neurons fire at high rates with high temporal precision. A clear gradient of Kv1.3 immunostaining along the lateral to medial tonotopic axis of the MNTB was detected. Highest levels were found in the lateral region of the MNTB, which corresponds to neurons that respond selectively to low-frequency auditory stimuli. Previous studies have demonstrated that MNTB neurons and their afferent inputs from the cochlear nucleus express three other members of the Kv1 family, Kv1.1, Kv1.2, and Kv1.6. Nevertheless, confocal microscopy of MNTB sections coimmunostained for Kv1.3 with these subunits revealed that the distribution of Kv1.3 differed significantly from other Kv1 family subunits. In particular, no axonal staining of Kv1.3 was detected, and most prominent labeling was in structures surrounding the somata of the principal neurons, suggesting specific localization to the large calyx of Held presynaptic endings that envelop the principal cells. The presence of Kv1.3 in presynaptic terminals was confirmed by coimmunolocalization with the synaptic markers synaptophysin, syntaxin, and synaptotagmin and by immunogold electron microscopy. Kv1.3 immunogold particles in the terminals were arrayed along the plasma membrane and on internal vesicular structures. To confirm these patterns of staining, we carried out immunolabeling on sections from Kv1.3(-/-) mice. No immunoreactivity could be detected in Kv1.3(-/-) mice either at the light level or in immunogold experiments. The finding of a tonotopic gradient in presynaptic terminals suggests that Kv1.3 may regulate neurotransmitter release differentially in neurons that respond to different frequencies of sound.
Collapse
Affiliation(s)
- Valeswara-Rao Gazula
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | | | |
Collapse
|
26
|
Castle NA. Pharmacological modulation of voltage-gated potassium channels as a therapeutic strategy. Expert Opin Ther Pat 2010; 20:1471-503. [PMID: 20726689 DOI: 10.1517/13543776.2010.513384] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE OF THE FIELD The human genome encodes at least 40 distinct voltage-gated potassium channel subtypes, which vary in regional expression, pharmacological and biophysical properties. Voltage-dependent potassium (Kv) channels help orchestrate many of the physiological and pathophysiological processes that promote and sometimes hinder the healthy functioning of our bodies. AREAS COVERED IN THIS REVIEW This review summarizes patent and scientific literature reports from the past decade highlighting the opportunities that Kv channels offer for the development of new therapeutic interventions for a wide variety of disorders. WHAT THE READER WILL GAIN The reader will gain an insight from an analysis of the associations of different Kv family members with disease processes, summary and evaluation of the development of therapeutically relevant pharmacological modulators of these channels, particularly focusing on proprietary agents being developed. TAKE HOME MESSAGE Development of new drugs that target Kv channels continue to be of great interest but is proving to be challenging. Nevertheless, opportunities for Kv channel modulators to have an impact on a wide range of disorders in the future remain an exciting prospect.
Collapse
|
27
|
Heeroma JH, Henneberger C, Rajakulendran S, Hanna MG, Schorge S, Kullmann DM. Episodic ataxia type 1 mutations differentially affect neuronal excitability and transmitter release. Dis Model Mech 2009; 2:612-9. [PMID: 19779067 DOI: 10.1242/dmm.003582] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Heterozygous mutations of KCNA1, the gene encoding potassium channel Kv1.1 subunits, cause episodic ataxia type 1 (EA1), which is characterized by paroxysmal cerebellar incoordination and interictal myokymia. Some mutations are also associated with epilepsy. Although Kv1.1-containing potassium channels play important roles in neuronal excitability and neurotransmitter release, it is not known how mutations associated with different clinical features affect the input-output relationships of individual neurons. We transduced rat hippocampal neurons, which were cultured on glial micro-islands, with lentiviruses expressing wild-type or mutant human KCNA1, and injected either depolarizing currents to evoke action potentials or depolarizing voltage commands to evoke autaptic currents. alpha-Dendrotoxin and tetraethylammonium allowed a pharmacological dissection of potassium currents underlying excitability and neurotransmission. Overexpression of wild-type Kv1.1 decreased both neuronal excitability and neurotransmitter release. By contrast, the C-terminus-truncated R417stop mutant, which is associated with severe drug-resistant EA1, had the opposite effect: increased excitability and release probability. Another mutant, T226R, which is associated with EA1 that is complicated by contractures and epilepsy, had no detectable effect on neuronal excitability; however, in common with R417stop, it markedly enhanced neurotransmitter release. The results provide direct evidence that EA1 mutations increase neurotransmitter release, and provide an insight into mechanisms underlying the phenotypic differences that are associated with different mutations.
Collapse
Affiliation(s)
- Joost H Heeroma
- Department of Clinical and Experimental Epilepsy, Department of Molecular Neuroscience and MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, UK
| | | | | | | | | | | |
Collapse
|
28
|
Sack JT, Shamotienko O, Dolly JO. How to validate a heteromeric ion channel drug target: assessing proper expression of concatenated subunits. ACTA ACUST UNITED AC 2008; 131:415-20. [PMID: 18411330 PMCID: PMC2346572 DOI: 10.1085/jgp.200709939] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Jon T Sack
- ternational Centre for Neurotherapeutics, Dublin City University, Dublin 9, Ireland.
| | | | | |
Collapse
|
29
|
Ionic channel function in action potential generation: current perspective. Mol Neurobiol 2008; 35:129-50. [PMID: 17917103 DOI: 10.1007/s12035-007-8001-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 11/30/1999] [Accepted: 11/10/2006] [Indexed: 10/23/2022]
Abstract
Over 50 years ago, Hodgkin and Huxley laid down the foundations of our current understanding of ionic channels. An impressive progress has been made during the following years that culminated in the revelation of the details of potassium channel structure. Nevertheless, even today, we cannot separate well currents recorded in central mammalian neurons. Many modern concepts about the function of sodium and potassium currents are based on experiments performed in nonmammalian cells. The recent recognition of the fast delayed rectifier current indicates that we need to reevaluate the biophysical role of sodium and potassium currents. This review will consider high quality voltage clamp data obtained from the soma of central mammalian neurons in the view of our current knowledge about proteins forming ionic channels. Fast sodium currents and three types of outward potassium currents, the delayed rectifier, the subthreshold A-type, and the D-type potassium currents, are discussed here. An updated current classification with biophysical role of each current subtype is provided. This review shows that details of kinetics of both sodium and outward potassium currents differ significantly from the classical descriptions and these differences may be of functional significance.
Collapse
|
30
|
Rampino T, Gregorini M, Guidetti C, Broggini M, Marchini S, Bonomi R, Maggio M, Roscini E, Soccio G, Tiboldo R, Dal Canton A. KCNA1 and TRPC6 ion channels and NHE1 exchanger operate the biological outcome of HGF/scatter factor in renal tubular cells. Growth Factors 2007; 25:382-91. [PMID: 18365869 DOI: 10.1080/08977190801892184] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Hepatocyte growth factor (HGF) is a glycoprotein that induces in vitro epithelial tubular cell growth, motility, scattering and branching morphogenesis. The cell machineries that account for HGF biological effects are still unclear. In previous study, we found that HGF upregulated in epithelial tubular cell line (HK2) 3 genes: potassium channel KCNA1, calcium channel (transient receptor potential channel, subfamily C, member 6, TRPC6) and Na(+)/H(+) exchanger-1 (NHE1). In this study, we validated these results with reverse transcription PCR and WB analysis. To investigate whether KCNA1, TRPC6, NHE1 mediate the changes induced by HGF in HK2, we studied the effects of their inhibitors: 4-aminopyridine, charybdotoxin, dendrotoxin K inhibitors of KCNA1, lanthanum, N-(p-amylcinnamoyl) anthranilic acid inhibitors of TRPC6, 5-(N-ethyl-N-isopropyl)amiloride, cariporide inhibitors of NHE1. The inhibitors prevented HGF-induced growth, migration, cytoskeletal reorganization and tubulogenesis in HK2. These results indicate that KCNA1, TRPC6 and NHE1 are cell machineries that are exploited by HGF to effect its biological outcome in renal tubular cells.
Collapse
Affiliation(s)
- Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chi XX, Nicol GD. Manipulation of the potassium channel Kv1.1 and its effect on neuronal excitability in rat sensory neurons. J Neurophysiol 2007; 98:2683-92. [PMID: 17855588 DOI: 10.1152/jn.00437.2007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Potassium channels play a critical role in regulating many aspects of action potential (AP) firing. To establish the contribution of the voltage-dependent potassium channel Kv1.1 in regulating excitability, we used the selective blocker dendrotoxin-K (DTX-K) and small interfering RNA (siRNA) targeted to Kv1.1 to determine their effects on AP firing in small-diameter capsaicin-sensitive sensory neurons. A 5-min exposure to 10 nM DTX-K suppressed the total potassium current (I(K)) measured at +40 mV by about 33%. DTX-K produced a twofold increase in the number of APs evoked by a ramp of depolarizing current. Associated with increased firing was a decrease in firing threshold and rheobase. DTX-K did not alter the resting membrane potential or the AP duration. A 48-h treatment with siRNA targeted to Kv1.1 reduced the expression of this channel protein by about 60% as measured in Western blots. After treatment with siRNA, I(K) was no longer sensitive to DTX-K, indicating a loss of functional protein. Similarly, after siRNA treatment exposure to DTX-K had no effect on the number of evoked APs, firing threshold, or rheobase. However, after siRNA treatment, the firing threshold had values similar to those obtained after acute exposure to DTX-K, suggesting that the loss of Kv1.1 plays a critical role in setting this parameter of excitability. These results demonstrate that Kv1.1 plays an important role in limiting AP firing and that siRNA may be a useful approach to establish the role of specific ion channels in the absence of selective antagonists.
Collapse
Affiliation(s)
- Xian Xuan Chi
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
32
|
Brew HM, Gittelman JX, Silverstein RS, Hanks TD, Demas VP, Robinson LC, Robbins CA, McKee-Johnson J, Chiu SY, Messing A, Tempel BL. Seizures and reduced life span in mice lacking the potassium channel subunit Kv1.2, but hypoexcitability and enlarged Kv1 currents in auditory neurons. J Neurophysiol 2007; 98:1501-25. [PMID: 17634333 DOI: 10.1152/jn.00640.2006] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genes Kcna1 and Kcna2 code for the voltage-dependent potassium channel subunits Kv1.1 and Kv1.2, which are coexpressed in large axons and commonly present within the same tetramers. Both contribute to the low-voltage-activated potassium current I Kv1, which powerfully limits excitability and facilitates temporally precise transmission of information, e.g., in auditory neurons of the medial nucleus of the trapezoid body (MNTB). Kcna1-null mice lacking Kv1.1 exhibited seizure susceptibility and hyperexcitability in axons and MNTB neurons, which also had reduced I Kv1. To explore whether a lack of Kv1.2 would cause a similar phenotype, we created and characterized Kcna2-null mice (-/-). The -/- mice exhibited increased seizure susceptibility compared with their +/+ and +/- littermates, as early as P14. The mRNA for Kv1.1 and Kv1.2 increased strongly in +/+ brain stems between P7 and P14, suggesting the increasing importance of these subunits for limiting excitability. Surprisingly, MNTB neurons in brain stem slices from -/- and +/- mice were hypoexcitable despite their Kcna2 deficit, and voltage-clamped -/- MNTB neurons had enlarged I Kv1. This contrasts strikingly with the Kcna1-null MNTB phenotype. Toxin block experiments on MNTB neurons suggested Kv1.2 was present in every +/+ Kv1 channel, about 60% of +/- Kv1 channels, and no -/- Kv1 channels. Kv1 channels lacking Kv1.2 activated at abnormally negative potentials, which may explain why MNTB neurons with larger proportions of such channels had larger I Kv1. If channel voltage dependence is determined by how many Kv1.2 subunits each contains, neurons might be able to fine-tune their excitability by adjusting the Kv1.1:Kv1.2 balance rather than altering Kv1 channel density.
Collapse
Affiliation(s)
- Helen M Brew
- Virginia Merrill Bloedel Hearing Research Center, Box 357923, University of Washington, Seattle, WA 98195-7923, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Shu Y, Yu Y, Yang J, McCormick DA. Selective control of cortical axonal spikes by a slowly inactivating K+ current. Proc Natl Acad Sci U S A 2007; 104:11453-8. [PMID: 17581873 PMCID: PMC2040919 DOI: 10.1073/pnas.0702041104] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurons are flexible electrophysiological entities in which the distribution and properties of ionic channels control their behaviors. Through simultaneous somatic and axonal whole-cell recording of layer 5 pyramidal cells, we demonstrate a remarkable differential expression of slowly inactivating K(+) currents. Depolarizing the axon, but not the soma, rapidly activated a low-threshold, slowly inactivating, outward current that was potently blocked by low doses of 4-aminopyridine, alpha-dendrotoxin, and rTityustoxin-K alpha. Block of this slowly inactivating current caused a large increase in spike duration in the axon but only a small increase in the soma and could result in distal axons generating repetitive discharge in response to local current injection. Importantly, this current was also responsible for slow changes in the axonal spike duration that are observed after somatic membrane potential change. These data indicate that low-threshold, slowly inactivating K(+) currents, containing Kv1.2 alpha subunits, play a key role in the flexible properties of intracortical axons and may contribute significantly to intracortical processing.
Collapse
Affiliation(s)
- Yousheng Shu
- *Institute of Neuroscience and Key Laboratory of Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Yuguo Yu
- Department of Neurobiology, Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510
| | - Jing Yang
- *Institute of Neuroscience and Key Laboratory of Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - David A. McCormick
- Department of Neurobiology, Kavli Institute for Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
34
|
Sokolov MV, Shamotienko O, Dhochartaigh SN, Sack JT, Dolly JO. Concatemers of brain Kv1 channel alpha subunits that give similar K+ currents yield pharmacologically distinguishable heteromers. Neuropharmacology 2007; 53:272-82. [PMID: 17637465 DOI: 10.1016/j.neuropharm.2007.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 04/16/2007] [Accepted: 05/15/2007] [Indexed: 01/06/2023]
Abstract
At least five subtypes of voltage-gated (Kv1) channels occur in neurons as tetrameric combinations of different alpha subunits. Their involvement in controlling cell excitability and synaptic transmission make them potential targets for neurotherapeutics. As a prerequisite for this, we established herein how the characteristics of hetero-oligomeric K(+) channels can be influenced by alpha subunit composition. Since the three most prevalent Kv1 subunits in brain are Kv1.2, 1.1 and 1.6, new Kv1.6-1.2 and Kv1.1-1.2 concatenated constructs in pIRES-EGFP were stably expressed in HEK cells and the biophysical plus pharmacological properties of their K(+) currents determined relative to those for the requisite homo-tetramers. These heteromers yielded delayed-rectifier type K(+) currents whose activation, deactivation and inactivation parameters are fairly similar although substituting Kv1.1 with Kv1.6 led to a small negative shift in the conductance-voltage relationship, a direction unexpected from the characteristics of the parental homo-tetramers. Changes resulting from swapping Kv1.6 for Kv1.1 in the concatemers were clearly discerned with two pharmacological agents, as measured by inhibition of the K(+) currents and Rb(+) efflux. alphaDendrotoxin and 4-aminopyridine gave a similar blockade of both hetero-tetramers, as expected. Most important for pharmacological dissection of channel subtypes, dendrotoxin(k) and tetraethylammonium readily distinguished the susceptible Kv1.1-1.2 containing oligomers from the resistant Kv1.6-1.2 channels. Moreover, the discriminating ability of dendrotoxin(k) was further confirmed by its far greater ability to displace (125)I-labelled alphadendrotoxin binding to Kv1.1-1.2 than Kv1.6-1.2 channels. Thus, due to the profiles of these two channel subtypes being found to differ, it seems that only multimers corresponding to those present in the nervous system provide meaningful targets for drug development.
Collapse
Affiliation(s)
- Maxim V Sokolov
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | | | | | | | | |
Collapse
|
35
|
Cao XJ, Shatadal S, Oertel D. Voltage-sensitive conductances of bushy cells of the Mammalian ventral cochlear nucleus. J Neurophysiol 2007; 97:3961-75. [PMID: 17428908 DOI: 10.1152/jn.00052.2007] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Bushy cells in the ventral cochlear nucleus convey firing of auditory nerve fibers to neurons in the superior olivary complex that compare the timing and intensity of sounds at the two ears and enable animals to localize sound sources in the horizontal plane. Three voltage-sensitive conductances allow bushy cells to convey acoustic information with submillisecond temporal precision. All bushy cells have a low-voltage-activated, alpha-dendrotoxin (alpha-DTX)-sensitive K(+) conductance (g(KL)) that was activated by depolarization past -70 mV, was half-activated at -39.0 +/- 1.7 (SE) mV, and inactivated approximately 60% over 5 s. Maximal g(KL) varied between 40 and 150 nS (mean: 80.8 +/- 16.7 nS). An alpha-DTX-insensitive, tetraethylammonium (TEA)-sensitive, K(+) conductance (g(KH)) was activated at voltages positive to -40 mV, was half-activated at -18.1 +/- 3.8 mV, and inactivated by 90% over 5 s. Maximal g(KH) varied between 35 and 80 nS (mean: 58.2 +/- 6.5 nS). A ZD7288-sensitive, mixed cation conductance (g(h)) was activated by hyperpolarization greater than -60 mV and half-activated at -83.1 +/- 1.1 mV. Maximum g(h) ranged between 14.5 and 56.6 nS (mean: 30.0 +/- 5.5 nS). 8-Br-cAMP shifted the voltage sensitivity of g(h) positively. Changes in temperature stably altered the steady-state magnitude of I(h). Both g(KL) and g(KH) contribute to repolarizing action potentials and to sharpening synaptic potentials. Those cells with the largest g(h) and the largest g(KL) fired least at the onset of a depolarization, required the fastest depolarizations to fire, and tended to be located nearest the nerve root.
Collapse
Affiliation(s)
- Xiao-Jie Cao
- Dept. of Physiology, University of Wisconsin School of Medicine, 1300 University Ave., Madison, WI 53706, USA
| | | | | |
Collapse
|
36
|
Vacher H, Mohapatra DP, Misonou H, Trimmer JS. Regulation of Kv1 channel trafficking by the mamba snake neurotoxin dendrotoxin K. FASEB J 2007; 21:906-14. [PMID: 17185748 PMCID: PMC2737685 DOI: 10.1096/fj.06-7229com] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Modulation of voltage-gated potassium (Kv) channel surface expression can profoundly affect neuronal excitability. Some, but not all, mammalian Shaker or Kv1 alpha subunits contain a dominant endoplasmic reticulum (ER) retention signal in their pore region, preventing surface expression of Kv1.1 homotetrameric channels and of heteromeric Kv1 channels containing more than one Kv1.1 subunit. The critical amino acid residues within this ER pore-region retention signal are also critical for high-affinity binding of snake dendrotoxins (DTX). This suggests that ER retention may be mediated by an ER protein with a domain structurally similar to that of DTX. One facet of such a model is that expression of soluble DTX in the ER lumen should compete for binding to the retention protein and allow for surface expression of retained Kv1.1. Here, we show that luminal DTX expression dramatically increased both the level of cell surface Kv1.1 immunofluorescence staining and the proportion of Kv1.1 with processed N-linked oligosaccharides. Electrophysiological analyses showed that luminal DTX expression led to significant increases in Kv1.1 currents. Together, these data showed that luminal DTX expression increases surface expression of functional Kv1.1 homotetrameric channels and support a model whereby a DTX-like ER protein regulates abundance of cell surface Kv1 channels.
Collapse
Affiliation(s)
- Helene Vacher
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | - Durga P. Mohapatra
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| | | | - James S. Trimmer
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
37
|
Vicente R, Escalada A, Villalonga N, Texidó L, Roura-Ferrer M, Martín-Satué M, López-Iglesias C, Soler C, Solsona C, Tamkun MM, Felipe A. Association of Kv1.5 and Kv1.3 contributes to the major voltage-dependent K+ channel in macrophages. J Biol Chem 2006; 281:37675-85. [PMID: 17038323 DOI: 10.1074/jbc.m605617200] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Voltage-dependent K(+) (Kv) currents in macrophages are mainly mediated by Kv1.3, but biophysical properties indicate that the channel composition could be different from that of T-lymphocytes. K(+) currents in mouse bone marrow-derived and Raw-264.7 macrophages are sensitive to Kv1.3 blockers, but unlike T-cells, macrophages express Kv1.5. Because Shaker subunits (Kv1) may form heterotetrameric complexes, we investigated whether Kv1.5 has a function in Kv currents in macrophages. Kv1.3 and Kv1.5 co-localize at the membrane, and half-activation voltages and pharmacology indicate that K(+) currents may be accounted for by various Kv complexes in macrophages. Co-expression of Kv1.3 and Kv1.5 in human embryonic kidney 293 cells showed that the presence of Kv1.5 leads to a positive shift in K(+) current half-activation voltages and that, like Kv1.3, Kv1.3/Kv1.5 heteromers are sensitive to r-margatoxin. In addition, both proteins co-immunoprecipitate and co-localize. Fluorescence resonance energy transfer studies further demonstrated that Kv1.5 and Kv1.3 form heterotetramers. Electrophysiological and pharmacological studies of different ratios of Kv1.3 and Kv1.5 co-expressed in Xenopus oocytes suggest that various hybrids might be responsible for K(+) currents in macrophages. Tumor necrosis factor-alpha-induced activation of macrophages increased Kv1.3 with no changes in Kv.1.5, which is consistent with a hyperpolarized shift in half-activation voltage and a lower IC(50) for margatoxin. Taken together, our results demonstrate that Kv1.5 co-associates with Kv1.3, generating functional heterotetramers in macrophages. Changes in the oligomeric composition of functional Kv channels would give rise to different biophysical and pharmacological properties, which could determine specific cellular responses.
Collapse
Affiliation(s)
- Rubén Vicente
- Molecular Physiology Laboratory, Departament de Bioquiámica i Biologia Molecular, Departament de Fisiologia, and Unitat de Reconeixement Molecular in situ, Serveis Cientificotècnics Universitat de Barcelona, E-08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Schulte U, Thumfart JO, Klöcker N, Sailer CA, Bildl W, Biniossek M, Dehn D, Deller T, Eble S, Abbass K, Wangler T, Knaus HG, Fakler B. The epilepsy-linked Lgi1 protein assembles into presynaptic Kv1 channels and inhibits inactivation by Kvbeta1. Neuron 2006; 49:697-706. [PMID: 16504945 DOI: 10.1016/j.neuron.2006.01.033] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Revised: 01/25/2006] [Accepted: 01/30/2006] [Indexed: 11/18/2022]
Abstract
The voltage-gated potassium (Kv) channel subunit Kv1.1 is a major constituent of presynaptic A-type channels that modulate synaptic transmission in CNS neurons. Here, we show that Kv1.1-containing channels are complexed with Lgi1, the functionally unassigned product of the leucine-rich glioma inactivated gene 1 (LGI1), which is causative for an autosomal dominant form of lateral temporal lobe epilepsy (ADLTE). In the hippocampal formation, both Kv1.1 and Lgi1 are coassembled with Kv1.4 and Kvbeta1 in axonal terminals. In A-type channels composed of these subunits, Lgi1 selectively prevents N-type inactivation mediated by the Kvbeta1 subunit. In contrast, defective Lgi1 molecules identified in ADLTE patients fail to exert this effect resulting in channels with rapid inactivation kinetics. The results establish Lgi1 as a novel subunit of Kv1.1-associated protein complexes and suggest that changes in inactivation gating of presynaptic A-type channels may promote epileptic activity.
Collapse
Affiliation(s)
- Uwe Schulte
- Logopharm GmbH, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
McGinley MJ, Oertel D. Rate thresholds determine the precision of temporal integration in principal cells of the ventral cochlear nucleus. Hear Res 2006; 216-217:52-63. [PMID: 16647828 DOI: 10.1016/j.heares.2006.02.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Revised: 02/15/2006] [Accepted: 02/16/2006] [Indexed: 11/25/2022]
Abstract
The three types of principal cells of the ventral cochlear nucleus (VCN), bushy, octopus, and T stellate, differ in the detection of coincidence among synaptic inputs. To explore the role of the action-potential-generation mechanism in the detection of coincident inputs, we examined responses to depolarizing currents that increased at varying rates. To fire an action potential, bushy cells, likely of the globular subtype, had to be depolarized faster than 4.8+/-2.8 mV/ms, octopus cells faster than 9.5+/-3.6 mV/ms, and T stellate cells fired irrespective of the rate of depolarization. The threshold rate of depolarization permitted definition of a time window over which depolarization could contribute to generating action potentials. This integration window differed between cell types. It was 5.3+/-1.8 ms for bushy cells and 1.4+/-0.3 ms for octopus cells. T Stellate cells fired action potentials in response to even slow depolarizations, showing that their integration window was unlimited so that temporal summation in these cells is limited by the time course of synaptic potentials. The rate of depolarization threshold in octopus and bushy cells was decreased by alpha-dendrotoxin while T stellate cells were largely insensitive to alpha-dendrotoxin indicating that low-voltage-activated K+ conductances (gKL) are important determinants of the integration window.
Collapse
Affiliation(s)
- Matthew J McGinley
- Department of Physiology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | | |
Collapse
|
40
|
Guan D, Lee JCF, Tkatch T, Surmeier DJ, Armstrong WE, Foehring RC. Expression and biophysical properties of Kv1 channels in supragranular neocortical pyramidal neurones. J Physiol 2005; 571:371-89. [PMID: 16373387 PMCID: PMC1796796 DOI: 10.1113/jphysiol.2005.097006] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Potassium channels are extremely diverse regulators of neuronal excitability. As part of an investigation into how this molecular diversity is utilized by neurones, we examined the expression and biophysical properties of native Kv1 channels in layer II/III pyramidal neurones from somatosensory and motor cortex. Single-cell RT-PCR, immunocytochemistry, and whole cell recordings with specific peptide toxins revealed that individual pyramidal cells express multiple Kv1 alpha-subunits. The most abundant subunit mRNAs were Kv1.1 > 1.2 > 1.4 > 1.3. All of these subunits were localized to somatodendritic as well as axonal cell compartments. These data suggest variability in the subunit complexion of Kv1 channels in these cells. The alpha-dendrotoxin (alpha-DTX)-sensitive current activated more rapidly and at more negative potentials than the alpha-DTX-insensitive current, was first observed at voltages near action potential threshold, and was relatively insensitive to holding potential. The alpha-DTX-sensitive current comprised about 10% of outward current at steady-state, in response to steps from -70 mV. From -50 mV, this percentage increased to approximately 20%. All cells expressed an alpha-DTX-sensitive current with slow inactivation kinetics. In some cells a transient component was also present. Deactivation kinetics were voltage dependent, such that deactivation was slow at potentials traversed by interspike intervals during repetitive firing. Because of its kinetics and voltage dependence, the alpha-DTX-sensitive current should be most important at physiological resting potentials and in response to brief stimuli. Kv1 channels should also be important at voltages near threshold and corresponding to interspike intervals.
Collapse
Affiliation(s)
- D Guan
- Department of Anatomy and Neurobiology, University of Tennessee, 855 Monroe Avenue, Memphis, TN 38163, USA
| | | | | | | | | | | |
Collapse
|
41
|
Persson AS, Klement G, Almgren M, Sahlholm K, Nilsson J, Petersson S, Århem P, Schalling M, Lavebratt C. A truncated Kv1.1 protein in the brain of the megencephaly mouse: expression and interaction. BMC Neurosci 2005; 6:65. [PMID: 16305740 PMCID: PMC1322225 DOI: 10.1186/1471-2202-6-65] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Accepted: 11/23/2005] [Indexed: 11/10/2022] Open
Abstract
Background The megencephaly mouse, mceph/mceph, is epileptic and displays a dramatically increased brain volume and neuronal count. The responsible mutation was recently revealed to be an eleven base pair deletion, leading to a frame shift, in the gene encoding the potassium channel Kv1.1. The predicted MCEPH protein is truncated at amino acid 230 out of 495. Truncated proteins are usually not expressed since nonsense mRNAs are most often degraded. However, high Kv1.1 mRNA levels in mceph/mceph brain indicated that it escaped this control mechanism. Therefore, we hypothesized that the truncated Kv1.1 would be expressed and dysregulate other Kv1 subunits in the mceph/mceph mice. Results We found that the MCEPH protein is expressed in the brain of mceph/mceph mice. MCEPH was found to lack mature (Golgi) glycosylation, but to be core glycosylated and trapped in the endoplasmic reticulum (ER). Interactions between MCEPH and other Kv1 subunits were studied in cell culture, Xenopus oocytes and the brain. MCEPH can form tetramers with Kv1.1 in cell culture and has a dominant negative effect on Kv1.2 and Kv1.3 currents in oocytes. However, it does not retain Kv1.2 in the ER of neurons. Conclusion The megencephaly mice express a truncated Kv1.1 in the brain, and constitute a unique tool to study Kv1.1 trafficking relevant for understanding epilepsy, ataxia and pathologic brain overgrowth.
Collapse
Affiliation(s)
- Ann-Sophie Persson
- Neurogenetic Unit, Department of Molecular Medicine and Surgery, CMM, Karolinska Hospital, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Göran Klement
- Nobel Institute for Neurophysiology, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Malin Almgren
- Neurogenetic Unit, Department of Molecular Medicine and Surgery, CMM, Karolinska Hospital, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Kristoffer Sahlholm
- Nobel Institute for Neurophysiology, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Nilsson
- Nobel Institute for Neurophysiology, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Petersson
- Neurogenetic Unit, Department of Molecular Medicine and Surgery, CMM, Karolinska Hospital, Karolinska Institutet, 171 76 Stockholm, Sweden
- The Ludwig Institute for Cancer Research, Stockholm Branch, Stockholm, Sweden
| | - Peter Århem
- Nobel Institute for Neurophysiology, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Schalling
- Neurogenetic Unit, Department of Molecular Medicine and Surgery, CMM, Karolinska Hospital, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Catharina Lavebratt
- Neurogenetic Unit, Department of Molecular Medicine and Surgery, CMM, Karolinska Hospital, Karolinska Institutet, 171 76 Stockholm, Sweden
| |
Collapse
|
42
|
Khavandgar S, Walter JT, Sageser K, Khodakhah K. Kv1 channels selectively prevent dendritic hyperexcitability in rat Purkinje cells. J Physiol 2005; 569:545-57. [PMID: 16210348 PMCID: PMC1464225 DOI: 10.1113/jphysiol.2005.098053] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Purkinje cells, the sole output of the cerebellar cortex, encode the timing signals required for motor coordination in their firing rate and activity pattern. Dendrites of Purkinje cells express a high density of P/Q-type voltage-gated calcium channels and fire dendritic calcium spikes. Here we show that dendritic subthreshold Kv1.2 subunit-containing Kv1 potassium channels prevent generation of random spontaneous calcium spikes. With Kv1 channels blocked, dendritic calcium spikes drive bursts of somatic sodium spikes and prevent the cell from faithfully encoding motor timing signals. The selective dendritic function of Kv1 channels in Purkinje cells allows them to effectively suppress dendritic hyperexcitability without hindering the generation of somatic action potentials. Further, we show that Kv1 channels also contribute to dendritic integration of parallel fibre synaptic input. Kv1 channels are often targeted to soma and axon and the data presented support a major dendritic function for these channels.
Collapse
Affiliation(s)
- Simin Khavandgar
- Department of Neuroscience, Albert Einstein College of Medicine, 506 Kennedy Center, 1410 Pelham Parkway South, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
43
|
Dolly JO. Molecular definition of neuronal targets for novel neurotherapeutics: SNAREs and Kv1 channels. Neurotoxicology 2005; 26:753-60. [PMID: 16125246 DOI: 10.1016/j.neuro.2005.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Accepted: 05/25/2005] [Indexed: 01/16/2023]
Affiliation(s)
- J Oliver Dolly
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
44
|
Bessone R, Martin-Eauclaire MF, Crest M, Mourre C. Heterogeneous competition of Kv1 channel toxins with kaliotoxin for binding in rat brain: autoradiographic analysis. Neurochem Int 2004; 45:1039-47. [PMID: 15337303 DOI: 10.1016/j.neuint.2004.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2003] [Accepted: 05/12/2004] [Indexed: 11/24/2022]
Abstract
The alpha-subunits of Kv1 channels display characteristic distributions and restricted co-assembly in mammalian brain. The heterogeneous composition of Kv1 channels has made it difficult to use specific toxins to label brain structures. We used autoradiography to analyse the competitive behaviour of three Kv1 channel toxins--alpha-dendrotoxin, kaliotoxin, and mast cell degranulating peptide--for binding to kaliotoxin binding sites in various brain structures. IC(50) varied considerably between brain regions (by up to three orders of magnitude) for each ligand. alpha-dendrotoxin and kaliotoxin competed equally in some regions and to different extents in others, identifying two types of structure. Mast cell degranulating peptide competed with (125)I-kaliotoxin less efficiently than alpha-dendrotoxin and kaliotoxin, in all regions. Thus, differences in the capacity of these three toxins to bind to kaliotoxin binding sites provide evidence of major differences in the composition of the Kv1 channels constituting the kaliotoxin binding sites.
Collapse
Affiliation(s)
- Richard Bessone
- ITIS, CNRS-UMR 6150, IFR Jean Roche, Faculté de Médecine de Marseille, Université de la Méditerranée, Boulevard Pierre Dramard 13916 Marseille, France
| | | | | | | |
Collapse
|
45
|
Dodson PD, Forsythe ID. Presynaptic K+ channels: electrifying regulators of synaptic terminal excitability. Trends Neurosci 2004; 27:210-7. [PMID: 15046880 DOI: 10.1016/j.tins.2004.02.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Potassium channels are crucial regulators of neuronal excitability, setting resting membrane potentials and firing thresholds, repolarizing action potentials and limiting excitability. Although most of our understanding of K+ channels is based on somatic recordings, there is good evidence that these channels are present in synaptic terminals. In recent years the improved access to presynaptic compartments afforded by direct recording techniques has indicated diverse roles for native K+ channels, from suppression of aberrant firing to action potential repolarization and activity-dependent modulation of synaptic activity. This article reviews the growing evidence for multiple roles and discrete localization of distinct K+ channels at presynaptic terminals.
Collapse
Affiliation(s)
- Paul D Dodson
- Department of Cell Physiology and Pharmacology, University of Leicester, PO Box 138, Leicester LE1 9HN, UK
| | | |
Collapse
|
46
|
Abstract
The intrinsic electrical properties of neurons are shaped in large part by the action of voltage-gated ion channels. Molecular cloning studies have revealed a large family of ion channel genes, many of which are expressed in mammalian brain. Much recent effort has focused on determining the contribution of the protein products of these genes to neuronal function. This requires knowledge of the abundance and distribution of the constituent subunits of the channels in specific mammalian central neurons. Here we review progress made in recent studies aimed at localizing specific ion channel subunits using in situ hybridization and immunohistochemistry. We then discuss the implications of these results in terms of neuronal physiology and neuronal mechanisms underlying the observed patterns of expression.
Collapse
Affiliation(s)
- James S Trimmer
- Department of Pharmacology, School of Medicine, University of California, Davis, California 95616-8635, USA.
| | | |
Collapse
|
47
|
Petersson S, Persson AS, Johansen JE, Ingvar M, Nilsson J, Klement G, Arhem P, Schalling M, Lavebratt C. Truncation of the Shaker-like voltage-gated potassium channel, Kv1.1, causes megencephaly. Eur J Neurosci 2003; 18:3231-40. [PMID: 14686897 DOI: 10.1111/j.1460-9568.2003.03044.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The megencephaly mouse, mceph/mceph, displays dramatically increased brain volume and hypertrophic brain cells. Despite overall enlargement, the mceph/mceph brain appears structurally normal, without oedema, hydrocephaly or leukodystrophy, and with only minor astrocytosis. Furthermore, it presents striking disturbances in expression of trophic and neuromodulating factors within the hippocampus and cortex. Using a positional cloning approach we have identified the mceph mutation. We show that mceph/mceph mice carry an 11-base-pair deletion in the gene encoding the Shaker-like voltage-gated potassium channel subtype 1, Kcna1. The mutation leads to a frame shift and the predicted MCEPH protein is truncated at amino acid 230 (out of 495), terminating with six aberrant amino acids. The expression of Kcna1 mRNA is increased in the mceph/mceph brain. However, the C-terminal domains of the corresponding Kv1.1 protein are absent. The putative MCEPH protein retains only the N-terminal domains for channel assembly and may congregate nonfunctional complexes of multiple Shaker-like subunits. Indeed, whereas Kcna2 and Kcna3 mRNA expression is normal, the mceph/mceph hippocampus displays decreased amounts of Kv1.2 and Kv1.3 proteins, suggesting interactions at the protein level. We show that mceph/mceph mice have disturbed brain electrophysiology and experience recurrent behavioural seizures, in agreement with the abnormal electrical brain activity found in Shaker mutants. However, in contrast to the commonly demonstrated epilepsy-induced neurodegeneration, we find that the mceph mutation leads to seizures with a concomitant increase in brain size, without overt neural atrophy.
Collapse
Affiliation(s)
- Susanna Petersson
- Neurogenetic Unit, Department of Molecular Medicine, CMM, L8:00, Karolinska Institutet, 171 76 Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Shen W, Hernandez-Lopez S, Tkatch T, Held JE, Surmeier DJ. Kv1.2-containing K+ channels regulate subthreshold excitability of striatal medium spiny neurons. J Neurophysiol 2003; 91:1337-49. [PMID: 13679409 DOI: 10.1152/jn.00414.2003] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A slowly inactivating, low-threshold K(+) current has been implicated in the regulation of state transitions and repetitive activity in striatal medium spiny neurons. However, the molecular identity of the channels underlying this current and their biophysical properties remain to be clearly determined. Because previous work had suggested this current arose from Kv1 family channels, high-affinity toxins for this family were tested for their ability to block whole cell K(+) currents activated by depolarization of acutely isolated neurons. alpha-Dendrotoxin, which blocks channels containing Kv1.1, Kv1.2, or Kv1.6 subunits, decreased currents evoked by depolarization. Three other Kv1 family toxins that lack a high affinity for Kv1.2 subunits, r-agitoxin-2, dendrotoxin-K, and r-margatoxin, failed to significantly reduce currents, implicating channels with Kv1.2 subunits. RT-PCR results confirmed the expression of Kv1.2 mRNA in identified medium spiny neurons. Currents attributable to Kv1.2 channels activated rapidly, inactivated slowly, and recovered from inactivation slowly. In the subthreshold range (ca. -60 mV), these currents accounted for as much as 50% of the depolarization-activated K(+) current. Moreover, their rapid activation and relatively slow deactivation suggested that they contribute to spike afterpotentials regulating repetitive discharge. This inference was confirmed in current-clamp recordings from medium spiny neurons in the slice preparation where Kv1.2 blockade reduced first-spike latency and increased discharge frequency evoked from hyperpolarized membrane potentials resembling the "down-state" found in vivo. These studies establish a clear functional role for somato-dendritic Kv1.2 channels in the regulation of state transitions and repetitive discharge in striatal medium spiny neurons.
Collapse
Affiliation(s)
- Weixing Shen
- Department of Physiology and Institute for Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | |
Collapse
|
49
|
Albarwani S, Nemetz LT, Madden JA, Tobin AA, England SK, Pratt PF, Rusch NJ. Voltage-gated K+ channels in rat small cerebral arteries: molecular identity of the functional channels. J Physiol 2003; 551:751-63. [PMID: 12815189 PMCID: PMC2343285 DOI: 10.1113/jphysiol.2003.040014] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Voltage-gated potassium (KV) channels represent an important dilator influence in the cerebral circulation, but the composition of these tetrameric ion channels remains unclear. The goals of the present study were to evaluate the contribution of KV1 family channels to the resting membrane potential and diameter of small rat cerebral arteries, and to identify the alpha-subunit composition of these channels using patch-clamp, molecular and immunological techniques. Initial studies indicated that 1 micromol l(-1) correolide (COR), a specific antagonist of KV1 channels, depolarized vascular smooth muscle cells (VSMCs) in pressurized (60 mmHg) cerebral arteries from -55 +/- 1 mV to -34 +/- 1 mV, and reduced the resting diameter from 152 +/- 15 microm to 103 +/- 20 microm. In patch clamped VSMCs from these arteries, COR-sensitive KV1 current accounted for 65 % of total outward KV current and was observed at physiological membrane potentials. RT-PCR identified mRNA encoding each of the six classical KV1 alpha-subunits, KV1.1-1.6, in rat cerebral arteries. However, only the KV1.2 and 1.5 proteins were detected by Western blot. The expression of these proteins in VSMCs was confirmed by immunocytochemistry and co-immunoprecipitation of KV1.2 and 1.5 from VSMC membranes suggested KV1.2/1.5 channel assembly. Subsequently, the pharmacological and voltage-sensitive properties of KV1 current in VSMCs were found to be consistent with a predominant expression of KV1.2/1.5 heterotetrameric channels. The findings of this study suggest that KV1.2/1.5 heterotetramers are preferentially expressed in rat cerebral VSMCs, and that these channels contribute to the resting membrane potential and diameter of rat small cerebral arteries.
Collapse
Affiliation(s)
- Sulayma Albarwani
- Department of Physiology, College of Medicine, Sultan Qaboos University, Al-khod, Sultanate of Oman
| | | | | | | | | | | | | |
Collapse
|
50
|
Brew HM, Hallows JL, Tempel BL. Hyperexcitability and reduced low threshold potassium currents in auditory neurons of mice lacking the channel subunit Kv1.1. J Physiol 2003; 548:1-20. [PMID: 12611922 PMCID: PMC2342794 DOI: 10.1113/jphysiol.2002.035568] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
A low voltage-activated potassium current, IKL, is found in auditory neuron types that have low excitability and precisely preserve the temporal pattern of activity present in their presynaptic inputs. The gene Kcna1 codes for Kv1.1 potassium channel subunits, which combine in expression systems to produce channel tetramers with properties similar to those of IKL, including sensitivity to dendrotoxin (DTX). Kv1.1 is strongly expressed in neurons with IKL, including auditory neurons of the medial nucleus of the trapezoid body (MNTB). We therefore decided to investigate how the absence of Kv1.1 affected channel properties and function in MNTB neurons from mice lacking Kcna1. We used the whole cell version of the patch clamp technique to record from MNTB neurons in brainstem slices from Kcna1-null (-/-) mice and their wild-type (+/+) and heterozygous (+/-) littermates. There was an IKL in voltage-clamped -/- MNTB neurons, but it was about half the amplitude of the IKL in +/+ neurons, with otherwise similar properties. Consistent with this, -/- MNTB neurons were more excitable than their +/+ counterparts; they fired more than twice as many action potentials (APs) during current steps, and the threshold current amplitude required to generate an AP was roughly halved. +/- MNTB neurons had excitability and IKL amplitudes identical to the +/+ neurons. The IKL remaining in -/- neurons was blocked by DTX, suggesting the underlying channels contained subunits Kv1.2 and/or Kv1.6 (also DTX-sensitive). DTX increased excitability further in the already hyperexcitable -/- MNTB neurons, suggesting that -/- IKL limited excitability despite its reduced amplitude in the absence of Kv1.1 subunits.
Collapse
Affiliation(s)
- Helen M Brew
- The Virginia Merrill Bloedel Hearing Research Center and the Department of Otolaryngology - Head and Neck Surgery, Box 357923, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | | | | |
Collapse
|