1
|
Rare Gain-of-Function KCND3 Variant Associated with Cerebellar Ataxia, Parkinsonism, Cognitive Dysfunction, and Brain Iron Accumulation. Int J Mol Sci 2021; 22:ijms22158247. [PMID: 34361012 PMCID: PMC8347726 DOI: 10.3390/ijms22158247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function mutations in the KV4.3 channel-encoding KCND3 gene are linked to neurodegenerative cerebellar ataxia. Patients suffering from neurodegeneration associated with iron deposition may also present with cerebellar ataxia. The mechanism underlying brain iron accumulation remains unclear. Here, we aim to ascertain the potential pathogenic role of KCND3 variant in iron accumulation-related cerebellar ataxia. We presented a patient with slowly progressive cerebellar ataxia, parkinsonism, cognitive impairment, and iron accumulation in the basal ganglia and the cerebellum. Whole exome sequencing analyses identified in the patient a heterozygous KCND3 c.1256G>A (p.R419H) variant predicted to be disease-causing by multiple bioinformatic analyses. In vitro biochemical and immunofluorescence examinations revealed that, compared to the human KV4.3 wild-type channel, the p.R419H variant exhibited normal protein abundance and subcellular localization pattern. Electrophysiological investigation, however, demonstrated that the KV4.3 p.R419H variant was associated with a dominant increase in potassium current amplitudes, as well as notable changes in voltage-dependent gating properties leading to enhanced potassium window current. These observations indicate that, in direct contrast with the loss-of-function KCND3 mutations previously reported in cerebellar ataxia patients, we identified a rare gain-of-function KCND3 variant that may expand the clinical and molecular spectra of neurodegenerative cerebellar disorders associated with brain iron accumulation.
Collapse
|
2
|
Novel KCND3 Variant Underlying Nonprogressive Congenital Ataxia or SCA19/22 Disrupt K V4.3 Protein Expression and K+ Currents with Variable Effects on Channel Properties. Int J Mol Sci 2021; 22:ijms22094986. [PMID: 34067185 PMCID: PMC8125845 DOI: 10.3390/ijms22094986] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
KCND3 encodes the voltage-gated potassium channel KV4.3 that is highly expressed in the cerebellum, where it regulates dendritic excitability and calcium influx. Loss-of-function KV4.3 mutations have been associated with dominant spinocerebellar ataxia (SCA19/22). By targeted NGS sequencing, we identified two novel KCND3 missense variants of the KV4.3 channel: p.S347W identified in a patient with adult-onset pure cerebellar syndrome and p.W359G detected in a child with congenital nonprogressive ataxia. Neuroimaging showed mild cerebellar atrophy in both patients. We performed a two-electrode voltage-clamp recording of KV4.3 currents in Xenopus oocytes: both the p.G345V (previously reported in a SCA19/22 family) and p.S347W mutants exhibited reduced peak currents by 50%, while no K+ current was detectable for the p.W359G mutant. We assessed the effect of the mutations on channel gating by measuring steady-state voltage-dependent activation and inactivation properties: no significant alterations were detected in p.G345V and p.S347W disease-associated variants, compared to controls. KV4.3 expression studies in HEK293T cells showed 53% (p.G345V), 45% (p.S347W) and 75% (p.W359G) reductions in mutant protein levels compared with the wildtype. The present study broadens the spectrum of the known phenotypes and identifies additional variants for KCND3-related disorders, outlining the importance of SCA gene screening in early-onset and congenital ataxia.
Collapse
|
3
|
Hu JH, Malloy C, Tabor GT, Gutzmann JJ, Liu Y, Abebe D, Karlsson RM, Durell S, Cameron HA, Hoffman DA. Activity-dependent isomerization of Kv4.2 by Pin1 regulates cognitive flexibility. Nat Commun 2020; 11:1567. [PMID: 32218435 PMCID: PMC7099064 DOI: 10.1038/s41467-020-15390-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 02/29/2020] [Indexed: 11/28/2022] Open
Abstract
Voltage-gated K+ channels function in macromolecular complexes with accessory subunits to regulate brain function. Here, we describe a peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1)-dependent mechanism that regulates the association of the A-type K+ channel subunit Kv4.2 with its auxiliary subunit dipeptidyl peptidase 6 (DPP6), and thereby modulates neuronal excitability and cognitive flexibility. We show that activity-induced Kv4.2 phosphorylation triggers Pin1 binding to, and isomerization of, Kv4.2 at the pThr607-Pro motif, leading to the dissociation of the Kv4.2-DPP6 complex. We generated a novel mouse line harboring a knock-in Thr607 to Ala (Kv4.2TA) mutation that abolished dynamic Pin1 binding to Kv4.2. CA1 pyramidal neurons of the hippocampus from these mice exhibited altered Kv4.2-DPP6 interaction, increased A-type K+ current, and reduced neuronal excitability. Behaviorally, Kv4.2TA mice displayed normal initial learning but improved reversal learning in both Morris water maze and lever press paradigms. These findings reveal a Pin1-mediated mechanism regulating reversal learning and provide potential targets for the treatment of neuropsychiatric disorders characterized by cognitive inflexibility.
Collapse
Affiliation(s)
- Jia-Hua Hu
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Cole Malloy
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - G Travis Tabor
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
- Medical Scientist Training Program, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Jakob J Gutzmann
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Ying Liu
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Daniel Abebe
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Rose-Marie Karlsson
- Section on Neuroplasticity, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Stewart Durell
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Dax A Hoffman
- Section on Molecular Neurophysiology and Biophysics, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA.
| |
Collapse
|
4
|
Hsiao CT, Fu SJ, Liu YT, Lu YH, Zhong CY, Tang CY, Soong BW, Jeng CJ. Novel SCA19/22-associated KCND3 mutations disrupt human K V 4.3 protein biosynthesis and channel gating. Hum Mutat 2019; 40:2088-2107. [PMID: 31293010 DOI: 10.1002/humu.23865] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/05/2019] [Accepted: 07/07/2019] [Indexed: 11/07/2022]
Abstract
Mutations in the human voltage-gated K+ channel subunit KV 4.3-encoding KCND3 gene have been associated with the autosomal dominant neurodegenerative disorder spinocerebellar ataxia types 19 and 22 (SCA19/22). The precise pathophysiology underlying the dominant inheritance pattern of SCA19/22 remains elusive. Using cerebellar ataxia-specific targeted next-generation sequencing technology, we identified two novel KCND3 mutations, c.950 G>A (p.C317Y) and c.1123 C>T (p.P375S) from a cohort with inherited cerebellar ataxias in Taiwan. The patients manifested notable phenotypic heterogeneity that includes cognitive impairment. We employed in vitro heterologous expression systems to inspect the biophysical and biochemical properties of human KV 4.3 harboring the two novel mutations, as well as two previously reported but uncharacterized disease-related mutations, c.1013 T>A (p.V338E) and c.1130 C>T (p.T377M). Electrophysiological analyses revealed that all of these SCA19/22-associated KV 4.3 mutant channels manifested loss-of-function phenotypes. Protein chemistry and immunofluorescence analyses further demonstrated that these mutants displayed enhanced protein degradation and defective membrane trafficking. By coexpressing KV 4.3 wild-type with the disease-related mutants, we provided direct evidence showing that the mutants instigated anomalous protein biosynthesis and channel gating of KV 4.3. We propose that the dominant inheritance pattern of SCA19/22 may be explained by the dominant-negative effects of the mutants on protein biosynthesis and voltage-dependent gating of KV 4.3 wild-type channel.
Collapse
Affiliation(s)
- Cheng-Tsung Hsiao
- Department of Internal Medicine, Taipei Veterans General Hospital Taoyuan Branch, Taoyuan, Taiwan
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Neurology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ssu-Ju Fu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yo-Tsen Liu
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Neurology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Hsiang Lu
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ciao-Yu Zhong
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yung Tang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Wen Soong
- Department of Neurology, Taipei Veterans General Hospital, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
5
|
Cacace R, Heeman B, Van Mossevelde S, De Roeck A, Hoogmartens J, De Rijk P, Gossye H, De Vos K, De Coster W, Strazisar M, De Baets G, Schymkowitz J, Rousseau F, Geerts N, De Pooter T, Peeters K, Sieben A, Martin JJ, Engelborghs S, Salmon E, Santens P, Vandenberghe R, Cras P, P. De Deyn P, C. van Swieten J, M. van Duijn C, van der Zee J, Sleegers K, Van Broeckhoven C. Loss of DPP6 in neurodegenerative dementia: a genetic player in the dysfunction of neuronal excitability. Acta Neuropathol 2019; 137:901-918. [PMID: 30874922 PMCID: PMC6531610 DOI: 10.1007/s00401-019-01976-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Emerging evidence suggested a converging mechanism in neurodegenerative brain diseases (NBD) involving early neuronal network dysfunctions and alterations in the homeostasis of neuronal firing as culprits of neurodegeneration. In this study, we used paired-end short-read and direct long-read whole genome sequencing to investigate an unresolved autosomal dominant dementia family significantly linked to 7q36. We identified and validated a chromosomal inversion of ca. 4 Mb, segregating on the disease haplotype and disrupting the coding sequence of dipeptidyl-peptidase 6 gene (DPP6). DPP6 resequencing identified significantly more rare variants-nonsense, frameshift, and missense-in early-onset Alzheimer's disease (EOAD, p value = 0.03, OR = 2.21 95% CI 1.05-4.82) and frontotemporal dementia (FTD, p = 0.006, OR = 2.59, 95% CI 1.28-5.49) patient cohorts. DPP6 is a type II transmembrane protein with a highly structured extracellular domain and is mainly expressed in brain, where it binds to the potassium channel Kv4.2 enhancing its expression, regulating its gating properties and controlling the dendritic excitability of hippocampal neurons. Using in vitro modeling, we showed that the missense variants found in patients destabilize DPP6 and reduce its membrane expression (p < 0.001 and p < 0.0001) leading to a loss of protein. Reduced DPP6 and/or Kv4.2 expression was also detected in brain tissue of missense variant carriers. Loss of DPP6 is known to cause neuronal hyperexcitability and behavioral alterations in Dpp6-KO mice. Taken together, the results of our genomic, genetic, expression and modeling analyses, provided direct evidence supporting the involvement of DPP6 loss in dementia. We propose that loss of function variants have a higher penetrance and disease impact, whereas the missense variants have a variable risk contribution to disease that can vary from high to low penetrance. Our findings of DPP6, as novel gene in dementia, strengthen the involvement of neuronal hyperexcitability and alteration in the homeostasis of neuronal firing as a disease mechanism to further investigate.
Collapse
Affiliation(s)
- Rita Cacace
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Bavo Heeman
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Sara Van Mossevelde
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Arne De Roeck
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Julie Hoogmartens
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Peter De Rijk
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Helena Gossye
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Kristof De Vos
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Wouter De Coster
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Mojca Strazisar
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Greet De Baets
- Switch Laboratory, VIB-KU Leuven Centre for Brain and Disease Research, Louvain, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB-KU Leuven Centre for Brain and Disease Research, Louvain, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB-KU Leuven Centre for Brain and Disease Research, Louvain, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Nathalie Geerts
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Tim De Pooter
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Karin Peeters
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Anne Sieben
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology, University Hospital Ghent and University of Ghent, Ghent, Belgium
| | | | - Sebastiaan Engelborghs
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Eric Salmon
- Department of Neurology, Centre Hospitalier Universitaire de Liège and University of Liège, Liège, Belgium
| | - Patrick Santens
- Department of Neurology, University Hospital Ghent and University of Ghent, Ghent, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Faculty of Medicine, KU Leuven, Louvain, Belgium
- Laboratory of Cognitive Neurology, Department of Neurology, University Hospitals Leuven, Louvain, Belgium
| | - Patrick Cras
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Peter P. De Deyn
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - John C. van Swieten
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Julie van der Zee
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Kristel Sleegers
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, University of Antwerp, CDE, Universiteitsplein 1, 2610 Antwerp, Belgium
| |
Collapse
|
6
|
Lin L, Murphy JG, Karlsson RM, Petralia RS, Gutzmann JJ, Abebe D, Wang YX, Cameron HA, Hoffman DA. DPP6 Loss Impacts Hippocampal Synaptic Development and Induces Behavioral Impairments in Recognition, Learning and Memory. Front Cell Neurosci 2018; 12:84. [PMID: 29651237 PMCID: PMC5884885 DOI: 10.3389/fncel.2018.00084] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/08/2018] [Indexed: 11/13/2022] Open
Abstract
DPP6 is well known as an auxiliary subunit of Kv4-containing, A-type K+ channels which regulate dendritic excitability in hippocampal CA1 pyramidal neurons. We have recently reported, however, a novel role for DPP6 in regulating dendritic filopodia formation and stability, affecting synaptic development and function. These results are notable considering recent clinical findings associating DPP6 with neurodevelopmental and intellectual disorders. Here we assessed the behavioral consequences of DPP6 loss. We found that DPP6 knockout (DPP6-KO) mice are impaired in hippocampus-dependent learning and memory. Results from the Morris water maze and T-maze tasks showed that DPP6-KO mice exhibit slower learning and reduced memory performance. DPP6 mouse brain weight is reduced throughout development compared with WT, and in vitro imaging results indicated that DPP6 loss affects synaptic structure and motility. Taken together, these results show impaired synaptic development along with spatial learning and memory deficiencies in DPP6-KO mice.
Collapse
Affiliation(s)
- Lin Lin
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Jonathan G Murphy
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Rose-Marie Karlsson
- Section on Neuroplasticity, National Institute of Mental Health, Bethesda, MD, United States
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Jakob J Gutzmann
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Daniel Abebe
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, Bethesda, MD, United States
| | - Dax A Hoffman
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| |
Collapse
|
7
|
Abstract
The electrical output of neurons relies critically on voltage- and calcium-gated ion channels. The traditional view of ion channels is that they operate independently of each other in the plasma membrane in a manner that could be predicted according to biophysical characteristics of the isolated current. However, there is increasing evidence that channels interact with each other not just functionally but also physically. This is exemplified in the case of Cav3 T-type calcium channels, where new work indicates the ability to form signaling complexes with different types of calcium-gated and even voltage-gated potassium channels. The formation of a Cav3-K complex provides the calcium source required to activate KCa1.1 or KCa3.1 channels and, furthermore, to bestow a calcium-dependent regulation of Kv4 channels via associated KChIP proteins. Here, we review these interactions and discuss their significance in the context of neuronal firing properties.
Collapse
|
8
|
Boronat A, Gelfand JM, Gresa-Arribas N, Jeong HY, Walsh M, Roberts K, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R, Graus F, Rudy B, Dalmau J. Encephalitis and antibodies to dipeptidyl-peptidase-like protein-6, a subunit of Kv4.2 potassium channels. Ann Neurol 2012; 73:120-8. [PMID: 23225603 DOI: 10.1002/ana.23756] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/28/2012] [Accepted: 09/04/2012] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To report a novel cell surface autoantigen of encephalitis that is a critical regulatory subunit of the Kv4.2 potassium channels. METHODS Four patients with encephalitis of unclear etiology and antibodies with a similar pattern of neuropil brain immunostaining were selected for autoantigen characterization. Techniques included immunoprecipitation, mass spectrometry, cell-base experiments with Kv4.2 and several dipeptidyl-peptidase-like protein-6 (DPPX) plasmid constructs, and comparative brain immunostaining of wild-type and DPPX-null mice. RESULTS Immunoprecipitation studies identified DPPX as the target autoantigen. A cell-based assay confirmed that all 4 patients, but not 210 controls, had DPPX antibodies. Symptoms included agitation, confusion, myoclonus, tremor, and seizures (1 case with prominent startle response). All patients had pleocytosis, and 3 had severe prodromal diarrhea of unknown etiology. Given that DPPX tunes up the Kv4.2 potassium channels (involved in somatodendritic signal integration and attenuation of dendritic back-propagation of action potentials), we determined the epitope distribution in DPPX, DPP10 (a protein homologous to DPPX), and Kv4.2. Patients' antibodies were found to be specific for DPPX, without reacting with DPP10 or Kv4.2. The unexplained diarrhea led to a demonstration of a robust expression of DPPX in the myenteric plexus, which strongly reacted with patients' antibodies. The course of neuropsychiatric symptoms was prolonged and often associated with relapses during decreasing immunotherapy. Long-term follow-up showed substantial improvement in 3 patients (1 was lost to follow-up). INTERPRETATION Antibodies to DPPX are associated with a protracted encephalitis characterized by central nervous system hyperexcitability (agitation, myoclonus, tremor, seizures), pleocytosis, and frequent diarrhea at symptom onset. The disorder is potentially treatable with immunotherapy.
Collapse
Affiliation(s)
- Anna Boronat
- Institute of Biomedical Research August Pi i Sunyer and Service of Neurology, Hospital Clinic, University of Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Liang P, Chen H, Cui Y, Lei L, Wang K. Functional rescue of Kv4.3 channel tetramerization mutants by KChIP4a. Biophys J 2010; 98:2867-76. [PMID: 20550899 DOI: 10.1016/j.bpj.2010.03.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 03/12/2010] [Accepted: 03/15/2010] [Indexed: 11/16/2022] Open
Abstract
KChIP4a shows a high homology with other members of the family of Kv channel-interacting proteins (KChIPs) in the conserved C-terminal core region, but exhibits a unique modulation of Kv4 channel gating and surface expression. Unlike KChIP1, the KChIP4 splice variant KChIP4a has been shown to inhibit surface expression and function as a suppressor of channel inactivation of Kv4. In this study, we sought to determine whether the multitasking KChIP4a modulates Kv4 function in a clamping fashion similar to that shown by KChIP1. Injection of Kv4.3 T1 zinc mutants into Xenopus oocytes resulted in the nonfunctional expression of Kv4.3 channels. Coexpression of Kv4.3 zinc mutants with WT KChIP4a gave rise to the functional expression of Kv4.3 current. Oocyte surface labeling results confirm the correlation between functional rescue and enhanced surface expression of zinc mutant proteins. Chimeric mutations that replace the Kv4.3 N-terminus with N-terminal KChIP4a or N-terminal deletion of KChIP4a further demonstrate that the functional rescue of Kv4.3 channel tetramerization mutants depends on the KChIP4a core region, but not its N-terminus. Structure-guided mutation of two critical residues of core KChIP4a attenuated functional rescue and tetrameric assembly. Moreover, size exclusion chromatography combined with fast protein liquid chromatography showed that KChIP4a can drive zinc mutant monomers to assemble as tetramers. Taken together, our results show that KChIP4a can rescue the function of tetramerization-defective Kv4 monomers. Therefore, we propose that core KChIP4a functions to promote tetrameric assembly and enhance surface expression of Kv4 channels by a clamping action, whereas its N-terminus inhibits surface expression of Kv4 by a mechanism that remains elusive.
Collapse
Affiliation(s)
- Ping Liang
- Department of Neurobiology, Neuroscience Research Institute, Peking University Health Science Center, Beijing, China
| | | | | | | | | |
Collapse
|
10
|
Seikel E, Trimmer JS. Convergent modulation of Kv4.2 channel alpha subunits by structurally distinct DPPX and KChIP auxiliary subunits. Biochemistry 2009; 48:5721-30. [PMID: 19441798 PMCID: PMC2733230 DOI: 10.1021/bi802316m] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Kv4.2 is the major voltage-gated K(+) (Kv) channel alpha subunit responsible for the somatodendritic transient or A-type current I(SA) that activates at subthreshold membrane potentials. Stable association of Kv4.2 with diverse auxiliary subunits and reversible Kv4.2 phosphorylation regulate I(SA) function. Two classes of auxiliary subunits play distinct roles in modulating the biophysical properties of Kv4.2: dipeptidyl-peptidase-like type II transmembrane proteins typified by DPPX-S, and cytoplasmic Ca(2+) binding proteins known as K(+) channel interacting proteins (KChIPs). Here, we characterize the convergent roles that DPPX-S and KChIPs play as component subunits of Kv4.2 channel complexes. We coexpressed DPPX-S with Kv4.2 in heterologous cells and found a dramatic redistribution of Kv4.2, releasing it from intracellular retention and allowing plasma membrane expression, as well as altered Kv4.2 phosphorylation, detergent solubility, and stability. These changes are remarkably similar to those obtained upon coexpression of Kv4.2 with the structurally distinct KChIPs1-3 auxiliary subunits. KChIP4a, which negatively affects the impact of other KChIPs on Kv4.2, also inhibits the effects of DPPX-S, consistent with the formation of a ternary complex of Kv4.2, DPPX-S, and KChIPs early in channel biosynthesis. Tandem MS analyses reveal that coexpression with DPPX-S or KChIP2 leads to a pattern of Kv4.2 phosphorylation in heterologous cells similar to that observed in brain, but lacking in cells expressing Kv4.2 alone. In conclusion, transmembrane DPPX-S and cytoplasmic KChIPs, despite having distinct structures and binding sites on Kv4.2, exert similar effects on Kv4.2 trafficking, but distinct effects on Kv4.2 gating.
Collapse
Affiliation(s)
| | - James S. Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
11
|
Maffie J, Rudy B. Weighing the evidence for a ternary protein complex mediating A-type K+ currents in neurons. J Physiol 2008; 586:5609-23. [PMID: 18845608 DOI: 10.1113/jphysiol.2008.161620] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The subthreshold-operating A-type K(+) current in neurons (I(SA)) has important roles in the regulation of neuronal excitability, the timing of action potential firing and synaptic integration and plasticity. The channels mediating this current (Kv4 channels) have been implicated in epilepsy, the control of dopamine release, and the regulation of pain plasticity. It has been proposed that Kv4 channels in neurons are ternary complexes of three types of protein: pore forming subunits of the Kv4 subfamily and two types of auxiliary subunits, the Ca(2+) binding proteins KChIPs and the dipeptidyl peptidase-like proteins (DPPLs) DPP6 (also known as DPPX) and DPP10 (4 molecules of each per channel for a total of 12 proteins in the complex). Here we consider the evidence supporting this hypothesis. Kv4 channels in many neurons are likely to be ternary complexes of these three types of protein. KChIPs and DPPLs are required to efficiently traffic Kv4 channels to the plasma membrane and regulate the functional properties of the channels. These proteins may also be important in determining the localization of the channels to specific neuronal compartments, their dynamics, and their response to neuromodulators. A surprisingly large number of additional proteins have been shown to modify Kv4 channels in heterologous expression systems, but their association with native Kv4 channels in neurons has not been properly validated. A critical consideration of the evidence suggests that it is unlikely that association of Kv4 channels with these additional proteins is widespread in the CNS. However, we cannot exclude that some of these proteins may associate with the channels transiently or in specific neurons or neuronal compartments, or that they may associate with the channels in other tissues.
Collapse
Affiliation(s)
- Jonathon Maffie
- Smilow Neuroscience Program, Department of Physiology and Neuroscience, New York University School of Medicine, Smilow Research Center, 522 First Avenue, 6th Floor, New York, NY 10016, USA
| | | |
Collapse
|
12
|
Kim J, Nadal MS, Clemens AM, Baron M, Jung SC, Misumi Y, Rudy B, Hoffman DA. Kv4 accessory protein DPPX (DPP6) is a critical regulator of membrane excitability in hippocampal CA1 pyramidal neurons. J Neurophysiol 2008; 100:1835-47. [PMID: 18667548 PMCID: PMC2576216 DOI: 10.1152/jn.90261.2008] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Accepted: 07/23/2008] [Indexed: 11/22/2022] Open
Abstract
A-type K+ currents have unique kinetic and voltage-dependent properties that allow them to finely tune synaptic integration, action potential (AP) shape and firing patterns. In hippocampal CA1 pyramidal neurons, Kv4 channels make up the majority of the somatodendritic A-type current. Studies in heterologous expression systems have shown that Kv4 channels interact with transmembrane dipeptidyl-peptidase-like proteins (DPPLs) to regulate the surface trafficking and biophysical properties of Kv4 channels. To investigate the influence of DPPLs in a native system, we conducted voltage-clamp experiments in patches from CA1 pyramidal neurons expressing short-interfering RNA (siRNA) targeting the DPPL variant known to be expressed in hippocampal pyramidal neurons, DPPX (siDPPX). In accordance with heterologous studies, we found that DPPX downregulation in neurons resulted in depolarizing shifts of the steady-state inactivation and activation curves, a shallower conductance-voltage slope, slowed inactivation, and a delayed recovery from inactivation for A-type currents. We carried out current-clamp experiments to determine the physiological effect of the A-type current modifications by DPPX. Neurons expressing siDPPX exhibited a surprisingly large reduction in subthreshold excitability as measured by a decrease in input resistance, delayed time to AP onset, and an increased AP threshold. Suprathreshold DPPX downregulation resulted in slower AP rise and weaker repolarization. Computer simulations supported our experimental results and demonstrated how DPPX remodeling of A-channel properties can result in opposing sub- and suprathreshold effects on excitability. The Kv4 auxiliary subunit DPPX thus acts to increase neuronal responsiveness and enhance signal precision by advancing AP initiation and accelerating both the rise and repolarization of APs.
Collapse
Affiliation(s)
- Jinhyun Kim
- Molecular Neurophysiology and Biophysics Unit, Laboratory of Cellular and Synaptic Neurophysiology, National Institute of Child Health and Human Development, National Institutes of Health, 35 Lincoln Dr., Rm. 3C-905, Bethesda, MD 20892-3715, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Soh H, Goldstein SAN. I SA channel complexes include four subunits each of DPP6 and Kv4.2. J Biol Chem 2008; 283:15072-7. [PMID: 18364354 PMCID: PMC2397469 DOI: 10.1074/jbc.m706964200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Indexed: 11/06/2022] Open
Abstract
Kv4 potassium channels produce rapidly inactivating currents that regulate excitability of muscles and nerves. To reconstitute the neuronal A-type current I(SA), Kv4 subunits assemble with DPP6, a single transmembrane domain accessory subunit. DPP6 alters function-accelerating activation, inactivation, and recovery from inactivation-and increases surface expression. We sought here to determine the stoichiometry of Kv4 and DPP6 in complexes using functional and biochemical methods. First, wild type channels formed from subunit monomers were compared with channels carrying subunits linked in tandem to enforce 4:4 and 4:2 assemblies (Kv4.2-DPP6 and Kv4.2-Kv4.2-DPP6). Next, channels were overexpressed and purified so that the molar ratio of subunits in complexes could be assessed by direct amino acid analysis. Both biophysical and biochemical methods indicate that I(SA) channels carry four subunits each of Kv4.2 and DPP6.
Collapse
Affiliation(s)
| | - Steve A. N. Goldstein
- Department of Pediatrics and Institute for Molecular Pediatric Sciences, Pritzker School of Medicine, Biological Sciences Division, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
14
|
Cui YY, Liang P, Wang KW. Enhanced trafficking of tetrameric Kv4.3 channels by KChIP1 clamping. Neurochem Res 2008; 33:2078-84. [PMID: 18401705 DOI: 10.1007/s11064-008-9688-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Accepted: 03/26/2008] [Indexed: 11/26/2022]
Abstract
The cytoplamsic auxiliary KChIPs modulate surface expression and gating properties of Kv4 channels. Recent co-crystal structure of Kv4.3 N-terminus and KChIP1 reveals a clamping action of the complex in which a single KChIP1 molecule laterally binds two neighboring Kv4.3 N-termini at different locations, thus forming two contact interfaces involved in the protein-protein interaction. In the second interface, it functions to stabilize the tetrameric assembly, but the role it plays in channel trafficking remains elusive. In this study, we examined the effects of KChIP1 on Kv4 protein trafficking in COS-7 cells expressing EGFP-tagged Kv4.3 channels using confocal microscopy. Mutations either in KChIP1 (KChIP1 L39E-Y57A-K61A) or Kv4.3 (Kv4.3 E70A-F73E) that disrupt the protein-protein interaction within the second interface can reduce surface expression of Kv4 channel proteins. Kv4.3 C110A, the Zn2+ binding site mutation in T1 domain, that disrupts the tetrameric assembly of the channels can be rescued by WT KChIP1, but not the KChIP1 triple mutant. These results were further confirmed by whole cell current recordings in oocytes. Our findings show that key residues of second interface involved in stabilizing tetrameric assembly can regulate the channel trafficking, indicating an intrinsic link between tetrameric assembly and channel trafficking. The results also suggest that formation of octameric Kv4 and KChIP complex by KChIPs clamping takes place before their trafficking to final destination on the cell surface.
Collapse
Affiliation(s)
- Yuan Yuan Cui
- Neuroscience Research Institute and Department of Neurobiology, Key Laboratory for Neuroscience of the Ministry of Education, Center for Protein Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100083, China
| | | | | |
Collapse
|
15
|
Amarillo Y, De Santiago-Castillo JA, Dougherty K, Maffie J, Kwon E, Covarrubias M, Rudy B. Ternary Kv4.2 channels recapitulate voltage-dependent inactivation kinetics of A-type K+ channels in cerebellar granule neurons. J Physiol 2008; 586:2093-106. [PMID: 18276729 DOI: 10.1113/jphysiol.2007.150540] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Kv4 channels mediate most of the somatodendritic subthreshold operating A-type current (I(SA)) in neurons. This current plays essential roles in the regulation of spike timing, repetitive firing, dendritic integration and plasticity. Neuronal Kv4 channels are thought to be ternary complexes of Kv4 pore-forming subunits and two types of accessory proteins, Kv channel interacting proteins (KChIPs) and the dipeptidyl-peptidase-like proteins (DPPLs) DPPX (DPP6) and DPP10. In heterologous cells, ternary Kv4 channels exhibit inactivation that slows down with increasing depolarization. Here, we compared the voltage dependence of the inactivation rate of channels expressed in heterologous mammalian cells by Kv4.2 proteins with that of channels containing Kv4.2 and KChIP1, Kv4.2 and DPPX-S, or Kv4.2, KChIP1 and DPPX-S, and found that the relation between inactivation rate and membrane potential is distinct for these four conditions. Moreover, recordings from native neurons showed that the inactivation kinetics of the I(SA) in cerebellar granule neurons has voltage dependence that is remarkably similar to that of ternary Kv4 channels containing KChIP1 and DPPX-S proteins in heterologous cells. The fact that this complex and unique behaviour (among A-type K(+) currents) is observed in both the native current and the current expressed in heterologous cells by the ternary complex containing Kv4, DPPX and KChIP proteins supports the hypothesis that somatically recorded native Kv4 channels in neurons include both types of accessory protein. Furthermore, quantitative global kinetic modelling showed that preferential closed-state inactivation and a weakly voltage-dependent opening step can explain the slowing of the inactivation rate with increasing depolarization. Therefore, it is likely that preferential closed-state inactivation is the physiological mechanism that regulates the activity of both ternary Kv4 channel complexes and native I(SA)-mediating channels.
Collapse
Affiliation(s)
- Yimy Amarillo
- Smilow Neuroscience Program, Smilow Research Center, New York University School of Medicine, 522 First Avenue, 6th Floor, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Jerng HH, Lauver AD, Pfaffinger PJ. DPP10 splice variants are localized in distinct neuronal populations and act to differentially regulate the inactivation properties of Kv4-based ion channels. Mol Cell Neurosci 2007; 35:604-24. [PMID: 17475505 PMCID: PMC3674967 DOI: 10.1016/j.mcn.2007.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 12/20/2006] [Accepted: 03/16/2007] [Indexed: 10/23/2022] Open
Abstract
Dipeptidyl peptidase-like proteins (DPLs) and Kv-channel-interacting proteins (KChIPs) join Kv4 pore-forming subunits to form multi-protein complexes that underlie subthreshold A-type currents (I(SA)) in neuronal somatodendritic compartments. Here, we characterize the functional effects and brain distributions of N-terminal variants belonging to the DPL dipeptidyl peptidase 10 (DPP10). In the Kv4.2+KChIP3+DPP10 channel complex, all DPP10 variants accelerate channel gating kinetics; however, the splice variant DPP10a produces uniquely fast inactivation kinetics that accelerates with increasing depolarization. This DPP10a-specific inactivation dominates in co-expression studies with KChIP4a and other DPP10 isoforms. Real-time qRT-PCR and in situ hybridization analyses reveal differential expression of DPP10 variants in rat brain. DPP10a transcripts are prominently expressed in the cortex, whereas DPP10c and DPP10d mRNAs exhibit more diffuse distributions. Our results suggest that DPP10a underlies rapid inactivation of cortical I(SA), and the regulation of isoform expression may contribute to the variable inactivation properties of I(SA) across different brain regions.
Collapse
Affiliation(s)
- Henry H Jerng
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, S630 Houston, TX 77030, USA.
| | | | | |
Collapse
|
17
|
Patel SP, Campbell DL. Transient outward potassium current, 'Ito', phenotypes in the mammalian left ventricle: underlying molecular, cellular and biophysical mechanisms. J Physiol 2005; 569:7-39. [PMID: 15831535 PMCID: PMC1464208 DOI: 10.1113/jphysiol.2005.086223] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/07/2005] [Accepted: 04/13/2005] [Indexed: 11/08/2022] Open
Abstract
At least two functionally distinct transient outward K(+) current (I(to)) phenotypes can exist across the free wall of the left ventricle (LV). Based upon their voltage-dependent kinetics of recovery from inactivation, these two phenotypes are designated 'I(to,fast)' (recovery time constants on the order of tens of milliseconds) and 'I(to,slow)' (recovery time constants on the order of thousands of milliseconds). Depending upon species, either I(to,fast), I(to,slow) or both current phenotypes may be expressed in the LV free wall. The expression gradients of these two I(to) phenotypes across the LV free wall are typically heterogeneous and, depending upon species, may consist of functional phenotypic gradients of both I(to,fast) and I(to,slow) and/or density gradients of either phenotype. We review the present evidence (molecular, biophysical, electrophysiological and pharmacological) for Kv4.2/4.3 alpha subunits underlying LV I(to,fast) and Kv1.4 alpha subunits underlying LV I(to,slow) and speculate upon the potential roles of each of these currents in determining frequency-dependent action potential characteristics of LV subepicardial versus subendocardial myocytes in different species. We also review the possible functional implications of (i) ancillary subunits that regulate Kv1.4 and Kv4.2/4.3 (Kvbeta subunits, DPPs), (ii) KChIP2 isoforms, (iii) spider toxin-mediated block of Kv4.2/4.3 (Heteropoda toxins, phrixotoxins), and (iv) potential mechanisms of modulation of I(to,fast) and I(to,slow) by cellular redox state, [Ca(2)(+)](i) and kinase-mediated phosphorylation. I(to) phenotypic activation and state-dependent gating models and molecular structure-function relationships are also discussed.
Collapse
Affiliation(s)
- Sangita P Patel
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, NY 14214-3078, USA.
| | | |
Collapse
|
18
|
Ono K, Toyono T, Honda E, Inenaga K. Transient outward K+ currents in rat dissociated subfornical organ neurones and angiotensin II effects. J Physiol 2005; 568:979-91. [PMID: 16123110 PMCID: PMC1464187 DOI: 10.1113/jphysiol.2005.089508] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although angiotensin II inhibits transient outward K+ currents (I(A)s) in subfornical organ neurones, there is no evidence concerning which Kv channels are involved. We investigated I(A)-generating Kv channels in dissociated rat subfornical organ neurones, using molecular, electrophysiological and pharmacological techniques, and studied the effects of angiotensin II. Conventional RT-PCR showed the presence of mRNAs for channels of the Kv3.4, Kv1.4 and Kv4 families, which are capable of generating I(A)s. Tetraethylammonium at 1 mm, which blocks Kv3 channel-derived currents, and blood-depressing substance-I, a Kv3.4-specific blocker, at 2 microm suppressed the I(A)-like component of whole-cell outward currents in some neurones. 4-Aminopyridine at 5 mm inhibited I(A)s in the presence of tetraethylammonium at 1 mm. Cd2+ at 300 microm shifted the activation and inactivation curves of the 4-aminopyridine-sensitive and tetraethylammonium-resistant I(A)s positively. The tetraethylammonium-resistant I(A)s showed fast and slow components during the process of recovery from inactivation, but the slow component was not seen in all neurones. The time constant of the fast recovery component was less than 200 ms, while that of the slow recovery component was around 1 s. Using single-cell RT-PCR, mRNAs for Kv4.2 and Kv4.3L were detected frequently, but those for Kv1.4 and Kv3.4 were seen only rarely. Angiotensin II at 30 nm inhibited the fast recovery component of tetraethylammonium-resistant I(A)s in many neurones. These results suggest that the fast recovery component of the tetraethylammonium-resistant I(A) in subfornical organ neurones depends upon Kv4, and that it can be modulated by angiotensin II.
Collapse
Affiliation(s)
- Kentaro Ono
- Department of Biosciences, Kyushu Dental College, 2-6-1 Manazuru, Kokurakitaku, Kitakyushu, 803-8580, Japan
| | | | | | | |
Collapse
|
19
|
Rhodes KJ, Carroll KI, Sung MA, Doliveira LC, Monaghan MM, Burke SL, Strassle BW, Buchwalder L, Menegola M, Cao J, An WF, Trimmer JS. KChIPs and Kv4 alpha subunits as integral components of A-type potassium channels in mammalian brain. J Neurosci 2005; 24:7903-15. [PMID: 15356203 PMCID: PMC6729940 DOI: 10.1523/jneurosci.0776-04.2004] [Citation(s) in RCA: 216] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Voltage-gated potassium (Kv) channels from the Kv4, or Shal-related, gene family underlie a major component of the A-type potassium current in mammalian central neurons. We recently identified a family of calcium-binding proteins, termed KChIPs (Kv channel interacting proteins), that bind to the cytoplasmic N termini of Kv4 family alpha subunits and modulate their surface density, inactivation kinetics, and rate of recovery from inactivation (An et al., 2000). Here, we used single and double-label immunohistochemistry, together with circumscribed lesions and coimmunoprecipitation analyses, to examine the regional and subcellular distribution of KChIPs1-4 and Kv4 family alpha subunits in adult rat brain. Immunohistochemical staining using KChIP-specific monoclonal antibodies revealed that the KChIP polypeptides are concentrated in neuronal somata and dendrites where their cellular and subcellular distribution overlaps, in an isoform-specific manner, with that of Kv4.2 and Kv4.3. For example, immunoreactivity for KChIP1 and Kv4.3 is concentrated in the somata and dendrites of hippocampal, striatal, and neocortical interneurons. Immunoreactivity for KChIP2, KChIP4, and Kv4.2 is concentrated in the apical and basal dendrites of hippocampal and neocortical pyramidal cells. Double-label immunofluorescence labeling revealed that throughout the forebrain, KChIP2 and KChIP4 are frequently colocalized with Kv4.2, whereas in cortical, hippocampal, and striatal interneurons, KChIP1 is frequently colocalized with Kv4.3. Coimmunoprecipitation analyses confirmed that all KChIPs coassociate with Kv4 alpha subunits in brain membranes, indicating that KChIPs 1-4 are integral components of native A-type Kv channel complexes and are likely to play a major role as modulators of somatodendritic excitability.
Collapse
Affiliation(s)
- Kenneth J Rhodes
- Neuroscience, Wyeth Discovery Research, Princeton, New Jersey 08543, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kodirov SA, Zhuravlev VL, Pavlenko VK, Safonova TA, Brachmann J. K+ channels in cardiomyocytes of the pulmonate snail helix. J Membr Biol 2004; 197:145-54. [PMID: 15042346 DOI: 10.1007/s00232-004-0649-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Indexed: 11/24/2022]
Abstract
We used the patch-clamp technique to identify and characterize the electrophysiological, biophysical, and pharmacological properties of K(+) channels in enzymatically dissociated ventricular cells of the land pulmonate snail Helix. The family of outward K(+) currents started to activate at -30 mV and the activation was faster at more depolarized potentials (time constants: at 0 mV 17.4 +/- 1.2 ms vs. 2.5 +/- 0.1 ms at + 60 mV). The current waveforms were similar to those of the A-type family of voltage-dependent K(+) currents encoded by Kv4.2 in mammals. Inactivation of the current was relatively fast, i.e., 50.2 +/- 1.8% of current was inactivated within 250 ms at + 40 mV. The recovery of K(+) channels from inactivation was relatively slow with a mean time constant of 1.7 +/- 0.2 s. Closer examination of steady-state inactivation kinetics revealed that the voltage dependency of inactivation was U-shaped, exhibiting less inactivation at more depolarized membrane potentials. On the basis of this phenomenon, we suggest that a channel encoded by Kv2.1 similar to that in mammals does exist in land pulmonates of the Helix genus. Outward currents were sensitive to 4-aminopyridine and tetraethylammonium chloride. The last compound was most effective, with an IC(50) of 336 +/- 142 micro mol l(-1). Thus, using distinct pharmacological and biophysical tools we identified different types of voltage-gated K(+) channels.
Collapse
Affiliation(s)
- S A Kodirov
- Department of Cardiology, Medical University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
21
|
Patel SP, Parai R, Parai R, Campbell DL. Regulation of Kv4.3 voltage-dependent gating kinetics by KChIP2 isoforms. J Physiol 2004; 557:19-41. [PMID: 14724186 PMCID: PMC1665034 DOI: 10.1113/jphysiol.2003.058172] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We conducted a kinetic analysis of the voltage dependence of macroscopic inactivation (tau(fast), tau(slow)), closed-state inactivation (tau(closed,inact)), recovery (tau(rec)), activation (tau(act)), and deactivation (tau(deact)) of Kv4.3 channels expressed alone in Xenopus oocytes and in the presence of the calcium-binding ancillary subunits KChIP2b and KChIP2d. We demonstrate that for all expression conditions, tau(rec), tau(closed,inact) and tau(fast) are components of closed-state inactivation transitions. The values of tau(closed,inact) and tau(fast) monotonically merge from -30 to -20 mV while the values of tau(closed,inact) and tau(rec) approach each other from -60 to -50 mV. These data generate classic bell-shaped time-constant-potential curves. With the KChIPs, these curves are distinct from that of Kv4.3 expressed alone due to acceleration of tau(rec) and slowing of tau(closed,inact) and tau(fast). Only at depolarized potentials where channels open is tau(slow) detectable suggesting that it represents an open-state inactivation mechanism. With increasing depolarization, KChIPs favour this open-state inactivation mechanism, supported by the observation of larger transient reopening currents upon membrane hyperpolarization compared to Kv4.3 expressed alone. We propose a Kv4.3 gating model wherein KChIP2 isoforms accelerate recovery, slow closed-state inactivation, and promote open-state inactivation. This model supports the observations that with KChIPs, closed-state inactivation transitions are [Ca(2+)](i)-independent, while open-state inactivation is [Ca(2+)](i)-dependent. The selective KChIP- and Ca(2+)-dependent modulation of Kv4.3 inactivation mechanisms predicted by this model provides a basis for dynamic modulation of the native cardiac transient outward current by intracellular Ca(2+) fluxes during the action potential.
Collapse
Affiliation(s)
- Sangita P Patel
- Department of Physiology and Biophysics, University at Buffalo, State University of New York, 124 Sherman Hall, Buffalo, NY 14214, USA
| | | | | | | |
Collapse
|
22
|
Patel SP, Campbell DL, Morales MJ, Strauss HC. Heterogeneous expression of KChIP2 isoforms in the ferret heart. J Physiol 2002; 539:649-56. [PMID: 11897837 PMCID: PMC2290197 DOI: 10.1113/jphysiol.2001.015156] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Kv4 channels are believed to underlie the rapidly recovering cardiac transient outward current (I(to)) phenotype. However, heterologously expressed Kv4 channels fail to fully reconstitute the native current. Kv channel interacting proteins (KChIPs) have been shown to modulate Kv4 channel function. To determine the potential involvement of KChIPs in the rapidly recovering I(to), we cloned three KChIP2 isoforms (designated fKChIP2, 2a and 2b) from the ferret heart. Based upon immunoblot data suggesting the presence of a potential endogenous KChIP-like protein in HEK 293, CHO and COS cells but absence in Xenopus oocytes, we coexpressed Kv4.3 and the fKChIP2 isoforms in Xenopus oocytes. Functional analysis showed that while all fKChIP2 isoforms produced a fourfold acceleration of recovery kinetics compared to Kv4.3 expressed alone, only fKChIP2a produced large depolarizing shifts in the V(1/2) of steady-state activation and inactivation as seen for the native rapidly recovering I(to). Analysis of RNA and protein expression of the three fKChIP2 isoforms in ferret ventricles showed that fKChIP2b was most abundant and was expressed in a gradient paralleling the rapidly recovering I(to) distribution. Ferret KChIP2 and 2a were expressed at very low levels. The ventricular expression distribution suggests that fKChIP2 isoforms are involved in modulation of the rapidly recovering I(to); however, additional regulatory factors are also likely to be involved in generating the native current.
Collapse
Affiliation(s)
- Sangita P Patel
- Department of Physiology and Biophysics, University at Buffalo, SUNY, 124 Sherman Hall, 3435 Main Street, Buffalo, NY 14214, USA.
| | | | | | | |
Collapse
|