Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 599(7883): 158-164, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34552243

RESUMO

Modulation of voltage-gated potassium (Kv) channels by auxiliary subunits is central to the physiological function of channels in the brain and heart1,2. Native Kv4 tetrameric channels form macromolecular ternary complexes with two auxiliary ß-subunits-intracellular Kv channel-interacting proteins (KChIPs) and transmembrane dipeptidyl peptidase-related proteins (DPPs)-to evoke rapidly activating and inactivating A-type currents, which prevent the backpropagation of action potentials1-5. However, the modulatory mechanisms of Kv4 channel complexes remain largely unknown. Here we report cryo-electron microscopy structures of the Kv4.2-DPP6S-KChIP1 dodecamer complex, the Kv4.2-KChIP1 and Kv4.2-DPP6S octamer complexes, and Kv4.2 alone. The structure of the Kv4.2-KChIP1 complex reveals that the intracellular N terminus of Kv4.2 interacts with its C terminus that extends from the S6 gating helix of the neighbouring Kv4.2 subunit. KChIP1 captures both the N and the C terminus of Kv4.2. In consequence, KChIP1 would prevent N-type inactivation and stabilize the S6 conformation to modulate gating of the S6 helices within the tetramer. By contrast, unlike the reported auxiliary subunits of voltage-gated channel complexes, DPP6S interacts with the S1 and S2 helices of the Kv4.2 voltage-sensing domain, which suggests that DPP6S stabilizes the conformation of the S1-S2 helices. DPP6S may therefore accelerate the voltage-dependent movement of the S4 helices. KChIP1 and DPP6S do not directly interact with each other in the Kv4.2-KChIP1-DPP6S ternary complex. Thus, our data suggest that two distinct modes of modulation contribute in an additive manner to evoke A-type currents from the native Kv4 macromolecular complex.


Assuntos
Microscopia Crioeletrônica , Ativação do Canal Iônico , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Canais de Potássio Shal/química , Canais de Potássio Shal/metabolismo , Animais , Dipeptidil Peptidases e Tripeptidil Peptidases/química , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Feminino , Humanos , Proteínas Interatuantes com Canais de Kv/química , Proteínas Interatuantes com Canais de Kv/metabolismo , Modelos Moleculares , Complexos Multiproteicos/genética , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Oócitos/metabolismo , Canais de Potássio/química , Canais de Potássio/metabolismo , Ligação Proteica , Canais de Potássio Shal/genética , Xenopus laevis
2.
Int J Mol Sci ; 22(15)2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34361012

RESUMO

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.


Assuntos
Disfunção Cognitiva/genética , Mutação com Ganho de Função , Ferro/metabolismo , Transtornos Parkinsonianos/genética , Canais de Potássio Shal/genética , Ataxias Espinocerebelares/genética , Potenciais de Ação , Idoso , Encéfalo/metabolismo , Disfunção Cognitiva/patologia , Células HEK293 , Humanos , Masculino , Transtornos Parkinsonianos/patologia , Domínios Proteicos , Canais de Potássio Shal/química , Canais de Potássio Shal/metabolismo , Ataxias Espinocerebelares/patologia
3.
Hum Mol Genet ; 30(23): 2300-2314, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34245260

RESUMO

Here, we report on six unrelated individuals, all presenting with early-onset global developmental delay, associated with impaired motor, speech and cognitive development, partly with developmental epileptic encephalopathy and physical dysmorphisms. All individuals carry heterozygous missense variants of KCND2, which encodes the voltage-gated potassium (Kv) channel α-subunit Kv4.2. The amino acid substitutions associated with the variants, p.(Glu323Lys) (E323K), p.(Pro403Ala) (P403A), p.(Val404Leu) (V404L) and p.(Val404Met) (V404M), affect sites known to be critical for channel gating. To unravel their likely pathogenicity, recombinant mutant channels were studied in the absence and presence of auxiliary ß-subunits under two-electrode voltage clamp in Xenopus oocytes. All channel mutants exhibited slowed and incomplete macroscopic inactivation, and the P403A variant in addition slowed activation. Co-expression of KChIP2 or DPP6 augmented the functional expression of both wild-type and mutant channels; however, the auxiliary ß-subunit-mediated gating modifications differed from wild type and among mutants. To simulate the putative setting in the affected individuals, heteromeric Kv4.2 channels (wild type + mutant) were studied as ternary complexes (containing both KChIP2 and DPP6). In the heteromeric ternary configuration, the E323K variant exhibited only marginal functional alterations compared to homomeric wild-type ternary, compatible with mild loss-of-function. By contrast, the P403A, V404L and V404M variants displayed strong gating impairment in the heteromeric ternary configuration, compatible with loss-of-function or gain-of-function. Our results support the etiological involvement of Kv4.2 channel gating impairment in early-onset monogenic global developmental delay. In addition, they suggest that gain-of-function mechanisms associated with a substitution of V404 increase epileptic seizure susceptibility.


Assuntos
Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/metabolismo , Variação Genética , Ativação do Canal Iônico , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo , Alelos , Substituição de Aminoácidos , Biomarcadores , Deficiências do Desenvolvimento/diagnóstico , Suscetibilidade a Doenças , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Fenótipo , Subunidades Proteicas , Canais de Potássio Shal/química
4.
Int J Mol Sci ; 21(16)2020 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-32824677

RESUMO

The subthreshold, transient A-type K+ current is a vital regulator of the excitability of neurons throughout the brain. In mammalian hippocampal pyramidal neurons, this current is carried primarily by ion channels comprising Kv4.2 α-subunits. These channels occupy the somatodendritic domains of these principle excitatory neurons and thus regulate membrane voltage relevant to the input-output efficacy of these cells. Owing to their robust control of membrane excitability and ubiquitous expression in the hippocampus, their dysfunction can alter network stability in a manner that manifests in recurrent seizures. Indeed, growing evidence implicates these channels in intractable epilepsies of the temporal lobe, which underscores the importance of determining the molecular mechanisms underlying their regulation and contribution to pathologies. Here, we describe the role of p38 kinase phosphorylation of a C-terminal motif in Kv4.2 in modulating hippocampal neuronal excitability and behavioral seizure strength. Using a combination of biochemical, single-cell electrophysiology, and in vivo seizure techniques, we show that kainic acid-induced seizure induces p38-mediated phosphorylation of Thr607 in Kv4.2 in a time-dependent manner. The pharmacological and genetic disruption of this process reduces neuronal excitability and dampens seizure intensity, illuminating a cellular cascade that may be targeted for therapeutic intervention to mitigate seizure intensity and progression.


Assuntos
Convulsões/metabolismo , Canais de Potássio Shal/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Potenciais de Ação , Motivos de Aminoácidos , Animais , Ondas Encefálicas , Feminino , Células HEK293 , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/fisiologia , Fosforilação , Convulsões/etiologia , Convulsões/fisiopatologia , Canais de Potássio Shal/química
5.
Elife ; 92020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32490811

RESUMO

CCK-expressing interneurons (CCK+INs) are crucial for controlling hippocampal activity. We found two firing phenotypes of CCK+INs in rat hippocampal CA3 area; either possessing a previously undetected membrane potential-dependent firing or regular firing phenotype, due to different low-voltage-activated potassium currents. These different excitability properties destine the two types for distinct functions, because the former is essentially silenced during realistic 8-15 Hz oscillations. By contrast, the general intrinsic excitability, morphology and gene-profiles of the two types were surprisingly similar. Even the expression of Kv4.3 channels were comparable, despite evidences showing that Kv4.3-mediated currents underlie the distinct firing properties. Instead, the firing phenotypes were correlated with the presence of distinct isoforms of Kv4 auxiliary subunits (KChIP1 vs. KChIP4e and DPP6S). Our results reveal the underlying mechanisms of two previously unknown types of CCK+INs and demonstrate that alternative splicing of few genes, which may be viewed as a minor change in the cells' whole transcriptome, can determine cell-type identity.


Assuntos
Região CA3 Hipocampal/citologia , Colecistocinina/metabolismo , Interneurônios , Canais de Potássio Shal , Animais , Células Cultivadas , Interneurônios/química , Interneurônios/classificação , Interneurônios/metabolismo , Potenciais da Membrana/fisiologia , Fenótipo , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Ratos Wistar , Canais de Potássio Shal/química , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo , Transcriptoma/genética
6.
Nat Commun ; 11(1): 1567, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-32218435

RESUMO

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.


Assuntos
Cognição , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Canais de Potássio Shal/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Células HEK293 , Humanos , Imidazóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Isomerismo , Aprendizagem , Camundongos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotreonina/metabolismo , Ligação Proteica , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Piridinas/farmacologia , Convulsões/metabolismo , Convulsões/patologia , Canais de Potássio Shal/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Phys Chem Chem Phys ; 21(45): 25290-25301, 2019 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-31701097

RESUMO

The voltage-gated potassium channel Kv4.3 plays a vital role in shaping the timing, frequency, and backpropagation of electrical signals in the brain and heart by generating fast transient currents at subthreshold membrane potentials in repetitive firing neurons. To achieve its physiological function, Kv4.3 is assisted by auxiliary ß-subunits that become integral parts of the native A-type potassium channels, among which there are the Kv channel-interacting proteins (KChIPs). KChIPs are a family of cytosolic proteins that, when coexpressed with Kv4, lead to higher current density, modulation of channel inactivation and faster recovery from inactivation, while the loss of KChIP function may lead to severe pathological states. Recently, the structural basis of the KChIP1-Kv4.3 interaction was reported by using two similar X-ray crystallographic structures, which supported a crucial role for KChIP1 in enhancing the stability of the Kv4.3 tetrameric assembly, thus helping the trafficking of the channel to the plasma membrane. Here, we investigate through fully atomistic simulations the structure and stability of the human Kv4.3 tetramerization (T1) domain in complex with KChIP1 upon specific mutations located in the first and second interfaces of the complex, as compared to the wild-type (WT). Our results nicely complement the available structural and biophysical information collected so far on these complex variants. In particular, the degree of structural deviations and energetic instability, from small to substantial, observed in these variants with respect to the WT model seems to parallel well the level of channel dysfunction known from electrophysiology data. Our simulations provide an octameric structure of the WT KChIP1-Kv4.3 assembly very similar to the known crystal structures, and, at the same time, highlight the importance of a previously overlooked site of interaction between KChIP1 and the Kv4.3 T1 domain.


Assuntos
Simulação por Computador , Proteínas Interatuantes com Canais de Kv/química , Canais de Potássio Shal/química , Cristalografia por Raios X , Humanos , Proteínas Interatuantes com Canais de Kv/genética , Modelos Moleculares , Mutação , Canais de Potássio Shal/genética
8.
Hum Mutat ; 40(11): 2088-2107, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31293010

RESUMO

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.


Assuntos
Ativação do Canal Iônico , Mutação , Biossíntese de Proteínas , Canais de Potássio Shal/metabolismo , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/metabolismo , Adulto , Idoso , Alelos , Sequência de Aminoácidos , Animais , Linhagem Celular , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Fenótipo , Domínios Proteicos , Canais de Potássio Shal/química , Canais de Potássio Shal/genética , Degenerações Espinocerebelares/diagnóstico , Relação Estrutura-Atividade , Adulto Jovem
9.
Mol Cell Neurosci ; 98: 121-130, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31212013

RESUMO

Kv4.2 voltage-gated K+ channel subunits, the primary source of the somatodendritic A-type K+ current in CA1 pyramidal neurons of the hippocampus, play important roles in regulating dendritic excitability and plasticity. To better study the trafficking and subcellular distribution of Kv4.2, we created and characterized a novel Kv4.2 construct encoding a bungarotoxin binding site in the extracellular S3-S4 linker region of the α-subunit. When expressed, this construct can be visualized in living cells after staining with rhodamine-conjugated bungarotoxin. We validated the utility of this construct by visualizing the spontaneous internalization and insertion of Kv4.2 in HEK 293T cells. We further report that Kv4.2 colocalized with several endosome markers in HEK 293T cells. In addition, Kv4.2 internalization is significantly impaired by mitogen-activated protein kinase (MAPK) inhibitors in transfected primary hippocampal neurons. Therefore, this newly developed BBS-Kv4.2 construct provides a novel and powerful tool for studying surface Kv4.2 channel localization and trafficking.


Assuntos
Bungarotoxinas/farmacologia , Canais de Potássio Shal/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Células HEK293 , Hipocampo/citologia , Humanos , Proteínas Interatuantes com Canais de Kv/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Ratos , Canais de Potássio Shal/química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
10.
PLoS One ; 13(1): e0191911, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29385176

RESUMO

Kv4.2 channels mediate a subthreshold-activating somatodendritic A-type current (ISA) in hippocampal neurons. We examined the role of accessory Kv channel interacting protein (KChIP) binding in somatodendritic surface expression and activity-dependent decrease in the availability of Kv4.2 channels. For this purpose we transfected cultured hippocampal neurons with cDNA coding for Kv4.2 wild-type (wt) or KChIP binding-deficient Kv4.2 mutants. All channels were equipped with an externally accessible hemagglutinin (HA)-tag and an EGFP-tag, which was attached to the C-terminal end. Combined analyses of EGFP self-fluorescence, surface HA immunostaining and patch-clamp recordings demonstrated similar dendritic trafficking and functional surface expression for Kv4.2[wt]HA,EGFP and the KChIP binding-deficient Kv4.2[A14K]HA,EGFP. Coexpression of exogenous KChIP2 augmented the surface expression of Kv4.2[wt]HA,EGFP but not Kv4.2[A14K]HA,EGFP. Notably, activity-dependent decrease in availability was more pronounced in Kv4.2[wt]HA,EGFP + KChIP2 coexpressing than in Kv4.2[A14K]HA,EGFP + KChIP2 coexpressing neurons. Our results do not support the notion that accessory KChIP binding is a prerequisite for dendritic trafficking and functional surface expression of Kv4.2 channels, however, accessory KChIP binding may play a potential role in Kv4.2 modulation during intrinsic plasticity processes.


Assuntos
Hipocampo/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Canais de Potássio Shal/metabolismo , Potenciais de Ação , Animais , Células Cultivadas , Dendritos/metabolismo , Epitopos/genética , Epitopos/metabolismo , Hipocampo/citologia , Humanos , Imuno-Histoquímica , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ligação Proteica , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Canais de Potássio Shal/química , Canais de Potássio Shal/genética , Transfecção
11.
Sci Rep ; 6: 31131, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27502553

RESUMO

Voltage-gated K(+) (Kv) channel activation depends on interactions between voltage sensors and an intracellular activation gate that controls access to a central pore cavity. Here, we hypothesize that this gate is additionally responsible for closed-state inactivation (CSI) in Kv4.x channels. These Kv channels undergo CSI by a mechanism that is still poorly understood. To test the hypothesis, we deduced the state of the Kv4.1 channel intracellular gate by exploiting the trap-door paradigm of pore blockade by internally applied quaternary ammonium (QA) ions exhibiting slow blocking kinetics and high-affinity for a blocking site. We found that inactivation gating seemingly traps benzyl-tributylammonium (bTBuA) when it enters the central pore cavity in the open state. However, bTBuA fails to block inactivated Kv4.1 channels, suggesting gated access involving an internal gate. In contrast, bTBuA blockade of a Shaker Kv channel that undergoes open-state P/C-type inactivation exhibits fast onset and recovery inconsistent with bTBuA trapping. Furthermore, the inactivated Shaker Kv channel is readily blocked by bTBuA. We conclude that Kv4.1 closed-state inactivation modulates pore blockade by QA ions in a manner that depends on the state of the internal activation gate.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Compostos de Amônio Quaternário/química , Compostos de Amônio Quaternário/farmacologia , Canais de Potássio Shal/química , Canais de Potássio Shal/metabolismo , Animais , Drosophila melanogaster , Células HEK293 , Humanos , Ratos , Canais de Potássio Shal/genética , Xenopus laevis
12.
Biophys J ; 110(1): 157-75, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26745419

RESUMO

We studied the kinetics and structural determinants of closed-state inactivation (CSI) in Kv4.2 channels, considering a multistep process and the possibility that both intra- and intersubunit dynamic binding (i.e., loss and restoration of physical contact) may occur between the S4-S5 linker, including the initial S5 segment (S4S5), and the S6 gate. We expressed Kv4.2 channels in Xenopus oocytes and measured the onset of low-voltage inactivation under two-electrode voltage clamp. Indicative of a transitory state, the onset kinetics were best described by a double-exponential function. To examine the involvement of individual S4S5 and S6 amino acid residues in dynamic binding, we studied S4S5 and S6 single alanine mutants and corresponding double mutants. Both transitory and steady-state inactivation were modified by these mutations, and we quantified the mutational effects based on apparent affinities for the respective inactivated states. Double-mutant cycle analyses revealed strong functional coupling of the S6 residues V404 and I412 to all tested S4S5 residues. To examine whether dynamic S4S5/S6 binding occurs within individual α-subunits or between neighboring α-subunits, we performed a double-mutant cycle analysis with Kv4.2 tandem-dimer constructs. The constructs carried either an S4S5/S6 double mutation in the first α-subunit and no mutation in the second (concatenated) α-subunit or an S4S5 point mutation in the first α-subunit and an S6 point mutation in the second α-subunit. Our results support the notion that CSI in Kv4.2 channels is a multistep process that involves dynamic binding both within individual α-subunits and between neighboring α-subunits.


Assuntos
Ativação do Canal Iônico , Subunidades Proteicas/metabolismo , Canais de Potássio Shal/química , Canais de Potássio Shal/metabolismo , Animais , Humanos , Cinética , Modelos Moleculares , Mutação Puntual , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Canais de Potássio Shal/genética , Xenopus/genética
13.
J Biol Chem ; 290(37): 22724-33, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26209633

RESUMO

Kv4 is a member of the voltage-gated K(+) channel family and forms a complex with various accessory subunits. Dipeptidyl aminopeptidase-like protein (DPP) is one of the auxiliary subunits for the Kv4 channel. Although DPP has been well characterized and is known to increase the current amplitude and accelerate the inactivation and recovery from inactivation of Kv4 current, it remains to be determined how many DPPs bind to one Kv4 channel. To examine whether the expression level of DPP changes the biophysical properties of Kv4, we expressed Kv4.2 and DPP10 in different ratios in Xenopus oocytes and analyzed the currents under two-electrode voltage clamp. The current amplitude and the speed of recovery from inactivation of Kv4.2 changed depending on the co-expression level of DPP10. This raised the possibility that the stoichiometry of the Kv4.2-DPP10 complex is variable and affects the biophysical properties of Kv4.2. We next determined the stoichiometry of DPP10 alone by subunit counting using single-molecule imaging. Approximately 70% of the DPP10 formed dimers in the plasma membrane, and the rest existed as monomers in the absence of Kv4.2. We next determined the stoichiometry of the Kv4.2-DPP10 complex; Kv4.2-mCherry and mEGFP-DPP10 were co-expressed in different ratios and the stoichiometries of Kv4.2-DPP10 complexes were evaluated by the subunit counting method. The stoichiometry of the Kv4.2-DPP10 complex was variable depending on the relative expression level of each subunit, with a preference for 4:2 stoichiometry. This preference may come from the bulky dimeric structure of the extracellular domain of DPP10.


Assuntos
Membrana Celular/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Complexos Multiproteicos/metabolismo , Canais de Potássio Shal/metabolismo , Animais , Membrana Celular/genética , Dipeptidil Peptidases e Tripeptidil Peptidases/química , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Canais de Potássio Shal/química , Canais de Potássio Shal/genética , Xenopus laevis
14.
Biophys J ; 108(11): 2658-69, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26039167

RESUMO

Auxiliary Kv channel-interacting proteins 1-4 (KChIPs1-4) coassemble with pore-forming Kv4 α-subunits to form channel complexes underlying somatodendritic subthreshold A-type current that regulates neuronal excitability. It has been hypothesized that different KChIPs can competitively bind to Kv4 α-subunit to form variable channel complexes that can exhibit distinct biophysical properties for modulation of neural function. In this study, we use single-molecule subunit counting by total internal reflection fluorescence microscopy in combinations with electrophysiology and biochemistry to investigate whether different isoforms of auxiliary KChIPs, KChIP4a, and KChIP4bl, can compete for binding of Kv4.3 to coassemble heteromultimeric channel complexes for modulation of channel function. To count the number of photobleaching steps solely from cell membrane, we take advantage of a membrane tethered k-ras-CAAX peptide that anchors cytosolic KChIP4 proteins to the surface for reduction of background noise. Single-molecule subunit counting reveals that the number of KChIP4 isoforms in Kv4.3-KChIP4 complexes can vary depending on the KChIP4 expression level. Increasing the amount of KChIP4bl gradually reduces bleaching steps of KChIP4a isoform proteins, and vice versa. Further analysis of channel gating kinetics from different Kv4-KChIP4 subunit compositions confirms that both KChIP4a and KChIP4bl can modulate the channel complex function upon coassembly. Taken together, our findings show that auxiliary KChIPs can heteroassemble with Kv4 in a competitive manner to form heteromultimeric Kv4-KChIP4 channel complexes that are biophysically distinct and regulated under physiological or pathological conditions.


Assuntos
Ligação Competitiva , Proteínas Interatuantes com Canais de Kv/química , Proteínas Interatuantes com Canais de Kv/metabolismo , Multimerização Proteica , Subunidades Proteicas/química , Canais de Potássio Shal/química , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica , Células HEK293 , Humanos , Ativação do Canal Iônico , Cinética , Porosidade , Subunidades Proteicas/metabolismo , Canais de Potássio Shal/metabolismo , Xenopus
15.
J Biol Chem ; 289(46): 32153-32165, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25190807

RESUMO

Dipeptidyl peptidase-like protein 6 (DPP6) is an auxiliary subunit of the Kv4 family of voltage-gated K(+) channels known to enhance channel surface expression and potently accelerate their kinetics. DPP6 is a single transmembrane protein, which is structurally remarkable for its large extracellular domain. Included in this domain is a cysteine-rich motif, the function of which is unknown. Here we show that this cysteine-rich domain of DPP6 is required for its export from the ER and expression on the cell surface. Disulfide bridges formed at C349/C356 and C465/C468 of the cysteine-rich domain are necessary for the enhancement of Kv4.2 channel surface expression but not its interaction with Kv4.2 subunits. The short intracellular N-terminal and transmembrane domains of DPP6 associates with and accelerates the recovery from inactivation of Kv4.2, but the entire extracellular domain is necessary to enhance Kv4.2 surface expression and stabilization. Our findings show that the cysteine-rich domain of DPP6 plays an important role in protein folding of DPP6 that is required for transport of DPP6/Kv4.2 complexes out of the ER.


Assuntos
Dipeptidil Peptidases e Tripeptidil Peptidases/química , Dipeptidil Peptidases e Tripeptidil Peptidases/fisiologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio/química , Canais de Potássio/fisiologia , Animais , Biotinilação , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Cisteína/química , Dissulfetos/química , Eletrofisiologia , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Potenciais da Membrana , Neurônios/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Canais de Potássio Shal/química
16.
Elife ; 3: e03255, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25030910

RESUMO

Temperature-sensitive transient receptor potential (TRP) ion channels are members of the large tetrameric cation channels superfamily but are considered to be uniquely sensitive to heat, which has been presumed to be due to the existence of an unidentified temperature-sensing domain. Here we report that the homologous voltage-gated potassium (Kv) channels also exhibit high temperature sensitivity comparable to that of TRPV1, which is detectable under specific conditions when the voltage sensor is functionally decoupled from the activation gate through either intrinsic mechanisms or mutations. Interestingly, mutations could tune Shaker channel to be either heat-activated or heat-deactivated. Therefore, high temperature sensitivity is intrinsic to both TRP and Kv channels. Our findings suggest important physiological roles of heat-induced variation in Kv channel activities. Mechanistically our findings indicate that temperature-sensing TRP channels may not contain a specialized heat-sensor domain; instead, non-obligatory allosteric gating permits the intrinsic heat sensitivity to drive channel activation, allowing temperature-sensitive TRP channels to function as polymodal nociceptors.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/química , Potássio/metabolismo , Canais de Potássio Shab/química , Superfamília Shaker de Canais de Potássio/química , Canais de Potássio Shal/química , Canais de Cátion TRPV/química , Regulação Alostérica , Animais , Expressão Gênica , Células HEK293 , Temperatura Alta , Humanos , Ativação do Canal Iônico , Transporte de Íons , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Canais de Potássio Shab/genética , Canais de Potássio Shab/metabolismo , Superfamília Shaker de Canais de Potássio/genética , Superfamília Shaker de Canais de Potássio/metabolismo , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Transgenes
17.
Anal Biochem ; 449: 99-105, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24361715

RESUMO

The specific binding of auxiliary Kv channel-interacting proteins (KChIPs) to the N terminus of Kv4 pore-forming α-subunits results in modulation of gating properties, surface expression, and subunit assembly of Kv4 channels. However, the interactions between KChIPs and Kv4 remain elusive. Thus, affinity capillary electrophoresis (ACE) was employed to quantitatively evaluate the interactions between KChIPs and Kv4.3 N terminus (KvN) and between KChIP4a/related mutants and Ca(2+) for the first time. The mobility ratio, derivatives calculated from the mobility shift method, was used to deduce the binding constants (Kb). As a result, the binding constants for KChIP4a/KvN and KChIP1/KvN complexes were (8.32±1.66)×10(6) L mol(-1) and (5.26±0.71)×10(6) L mol(-1), respectively. In addition, in the presence of calcium (10 µmol L(-1)), the binding constant of KChIP4a/KvN increased to (6.72±1.66)×10(7) L mol(-1). In addition, the binding constant of KChIP4a with Ca(2+) was (7.1±1.5)×10(7) L mol(-1). Besides, studies on the effect of truncated mutants revealed that the third EF hand of KChIP4a was related to high-affinity binding with Ca(2+), and the integrity of the molecular structure of KChIP4a was important for Ca(2+) binding. This method profits from small samples, rapid analysis, and simple operation without being time-consuming.


Assuntos
Eletroforese Capilar/métodos , Proteínas Interatuantes com Canais de Kv/metabolismo , Mapeamento de Interação de Proteínas/métodos , Canais de Potássio Shal/metabolismo , Cálcio/metabolismo , Humanos , Proteínas Interatuantes com Canais de Kv/química , Proteínas Interatuantes com Canais de Kv/genética , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Canais de Potássio Shal/química
18.
Cell Physiol Biochem ; 31(6): 968-80, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23839156

RESUMO

BACKGROUND/AIMS: Potassium channels are tetrameric proteins providing potassium selective passage through lipid embedded proteinaceous pores with highest fidelity. The selectivity results from binding to discrete potassium binding sites and stabilization of a hydrated potassium ion in a central internal cavity. The four potassium binding sites, generated by the conserved TTxGYGD signature sequence are formed by the backbone carbonyls of the amino acids TXGYG. Residues KV1.5-Val481, KV4.3-Leu368 and KV7.1- Ile 313 represent the amino acids in the X position of the respective channels. METHODS: Here, we study the impact of these residues on ion selectivity, permeation and inactivation kinetics as well as the modulation by ß-subunits using site-specific mutagenesis, electrophysiological analyses and molecular dynamics simulations. RESULTS: We identify this position as key in modulation of slow inactivation by structurally dissimilar ß-subunits in different KV channels. CONCLUSION: We propose a model in which structural changes accompanying activation and ß-subunit modulation allosterically constrain the backbone carbonyl oxygen atoms via the side chain of the respective X-residue in the signature sequence to reduce conductance during slow inactivation.


Assuntos
Canal de Potássio KCNQ1/metabolismo , Canal de Potássio Kv1.5/metabolismo , Canais de Potássio Shal/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Humanos , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/genética , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Potássio/metabolismo , Estrutura Quaternária de Proteína , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Alinhamento de Sequência , Canais de Potássio Shal/química , Canais de Potássio Shal/genética , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
19.
J Neurochem ; 126(4): 462-72, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23692269

RESUMO

K channel-interacting proteins (KChIPs) enhance functional expression of Kv4 channels by binding to an N-terminal regulatory region located in the first 40 amino acids of Kv4.2 that we call the functional expression regulating N-terminal (FERN) domain. Mutating two residues in the FERN domain to alanines, W8A and F11A, disrupts KChIP binding and regulation of Kv4.2 without eliminating the FERN domain's control of basal expression level or regulation by DPP6. When Kv4.2(W8A,F11A) is co-expressed with wild type Kv4.2 and KChIP3 subunits, a dominant negative effect is seen where the current expression is reduced to levels normally seen without KChIP addition. The dominant negative effect correlates with heteromultimeric channels remaining on intracellular membranes despite KChIP binding to non-mutant Kv4.2 subunits. In contrast, the deletion mutant Kv4.2(Δ1-40), eliminating both KChIP binding and the FERN domain, has no dominant negative effect even though the maximal conductance level is 5x lower than seen with KChIP3. The 5x increased expression seen with KChIP integration into the channel is fully apparent even when a reduced number of KChIP subunits are incorporated as long as all FERN domains are bound. Our results support the hypothesis that KChIPs enhances Kv4.2 functional expression by a 1 : 1 suppression of the N-terminal FERN domain and by producing additional positive regulatory effects on functional channel expression.


Assuntos
Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo , Animais , Células CHO , Células COS , Chlorocebus aethiops , Clonagem Molecular , Cricetinae , Proteínas de Fluorescência Verde/genética , Membranas Intracelulares/metabolismo , Ativação do Canal Iônico/fisiologia , Proteínas Interatuantes com Canais de Kv/química , Potenciais da Membrana/fisiologia , Mutagênese/fisiologia , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transporte Proteico/fisiologia , Proteínas Repressoras/química , Canais de Potássio Shal/química
20.
Channels (Austin) ; 7(2): 74-84, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23334377

RESUMO

Polyunsaturated fatty acids such as arachidonic acid (AA) exhibit inhibitory modulation of Kv4 potassium channels. Molecular docking approaches using a Kv4.2 homology model predicted a membrane-embedded binding pocket for AA comprised of the S4-S5 linker on one subunit and several hydrophobic residues within S3, S5 and S6 from an adjacent subunit. The pocket is conserved among Kv4 channels. We tested the hypothesis that modulatory effects of AA on Kv4.2/KChIP channels require access to this site. Targeted mutation of a polar residue (K318) and a nonpolar residue (G314) within the S4-S5 linker as well as a nonpolar residue in S3 (V261) significantly impaired the effects of AA on K (+) currents in Xenopus oocytes. These residues may be important in stabilizing (K318) or regulating access to (V261, G314) the negatively charged carboxylate moiety on the fatty acid. Structural specificity was supported by the lack of disruption of AA effects observed with mutations at residues located near, but not within the predicted binding pocket. Furthermore, we found that the crystal structure of the related Kv1.2/2.1 chimera lacks the structural features present in the proposed AA docking site of Kv4.2 and the Kv1.2/2.1 K (+) currents were unaffected by AA. We simulated the mutagenic substitutions in our Kv4.2 model to demonstrate how specific mutations may disrupt the putative AA binding pocket. We conclude that AA inhibits Kv4 channel currents and facilitates current decay by binding within a hydrophobic pocket in the channel in which K318 within the S4-S5 linker is a critical residue for AA interaction.


Assuntos
Ácido Araquidônico/farmacologia , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Bloqueadores dos Canais de Potássio/farmacologia , Homologia de Sequência de Aminoácidos , Canais de Potássio Shal/química , Sequência de Aminoácidos , Animais , Ácido Araquidônico/química , Sítios de Ligação , Humanos , Dados de Sequência Molecular , Bloqueadores dos Canais de Potássio/química , Ratos , Canais de Potássio Shal/antagonistas & inibidores , Canais de Potássio Shal/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...