Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Neurosci ; 40(8): 3179-88, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25080203

RESUMO

Global cerebral ischemia following cardiac arrest and cardiopulmonary resuscitation (CA/CPR) causes injury to hippocampal CA1 pyramidal neurons and impairs cognition. Small conductance Ca(2+)-activated potassium channels type 2 (SK2), expressed in CA1 pyramidal neurons, have been implicated as potential protective targets. Here we showed that, in mice, hippocampal long-term potentiation (LTP) was impaired as early as 3 h after recovery from CA/CPR and LTP remained impaired for at least 30 days. Treatment with the SK2 channel agonist 1-Ethyl-2-benzimidazolinone (1-EBIO) at 30 min after CA provided sustained protection from plasticity deficits, with LTP being maintained at control levels at 30 days after recovery from CA/CPR. Minimal changes in glutamate release probability were observed at delayed times after CA/CPR, implicating post-synaptic mechanisms. Real-time quantitative reverse transcriptase-polymerase chain reaction indicated that CA/CPR did not cause a loss of N-methyl-D-aspartate (NMDA) receptor mRNA at 7 or 30 days after CA/CPR. Similarly, no change in synaptic NMDA receptor protein levels was observed at 7 or 30 days after CA/CPR. Further, patch-clamp experiments demonstrated no change in functional synaptic NMDA receptors at 7 or 30 days after CA/CPR. Electrophysiology recordings showed that synaptic SK channel activity was reduced for the duration of experiments performed (up to 30 days) and that, surprisingly, treatment with 1-EBIO did not prevent the CA/CPR-induced loss of synaptic SK channel function. We concluded that CA/CPR caused alterations in post-synaptic signaling that were prevented by treatment with the SK2 agonist 1-EBIO, indicating that activators of SK2 channels may be useful therapeutic agents to prevent ischemic injury and cognitive impairments.


Assuntos
Isquemia Encefálica/fisiopatologia , Hipocampo/fisiopatologia , Potenciação de Longa Duração , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Animais , Benzimidazóis/farmacologia , Isquemia Encefálica/prevenção & controle , Agonistas dos Canais de Cálcio/farmacologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de AMPA/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia
2.
Science ; 235(4795): 1514-7, 1987 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-3547652

RESUMO

This report describes the characterization of a genomic locus in the rat that encodes overlapping genes occupying both strands of the same piece of DNA. One gene (strand) encodes gonadotropin-releasing hormone (GnRH). A second gene, SH, is transcribed from the other DNA strand to produce RNA of undefined function. The RNAs transcribed from each DNA strand are spliced and polyadenylated, and share significant exon domains. GnRH is expressed in the central nervous system while SH transcripts are present in the heart. Thus, the genome of a mammalian organism encodes two distinct genes by using both strands of the same DNA.


Assuntos
DNA/genética , Genes , Hormônio Liberador de Gonadotropina/genética , RNA Mensageiro/genética , Animais , Sequência de Bases , Éxons , Coração/fisiologia , Hipotálamo/fisiologia , Íntrons , Splicing de RNA , Ratos , Moldes Genéticos , Transcrição Gênica
3.
Science ; 255(5052): 1705-7, 1992 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-1553557

RESUMO

A slowly activating, voltage-dependent potassium channel protein cloned from rat kidney was expressed in Xenopus oocytes. Two activators of protein kinase C, 1-oleoyl-2-acetyl-rac-glycerol and phorbol 12,13-didecanoate, inhibited the current. This inhibition was blocked by the kinase inhibitor staurosporine. Inhibition of the current was not seen in channels in which Ser103 was replaced by Ala, although other properties of the current were unchanged. These results indicate that inhibition of the potassium current results from direct phosphorylation of the channel subunit protein at Ser103.


Assuntos
Canais de Potássio/fisiologia , Proteína Quinase C/metabolismo , Sequência de Aminoácidos , Animais , DNA/genética , Diglicerídeos/farmacologia , Ativação do Canal Iônico , Potenciais da Membrana , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ésteres de Forbol/farmacologia , Fosforilação , Ratos , Relação Estrutura-Atividade
4.
Science ; 244(4901): 221-4, 1989 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-2539643

RESUMO

Potassium channels are ubiquitous membrane proteins with essential roles in nervous tissue, but little is known about the relation between their function and their molecular structure. A complementary DNA library was made from rat hippocampus, and a complementary DNA clone (RBK-1) was isolated. The predicted sequence of the 495-amino acid protein is homologous to potassium channel proteins encoded by the Shaker locus of Drosophila and differs by only three amino acids from the expected product of a mouse clone MBK-1. Messenger RNA transcribed from RBK-1 in vitro directed the expression of potassium channels when it was injected into Xenopus oocytes. The potassium current through the expressed channels resembles both the transient (or A) and the delayed rectifier currents reported in mammalian neurons and is sensitive to both 4-aminopyridine and tetraethylammonium.


Assuntos
Encéfalo/metabolismo , Oócitos/metabolismo , Canais de Potássio/metabolismo , 4-Aminopiridina , Sequência de Aminoácidos , Aminopiridinas/farmacologia , Animais , Sequência de Bases , Clonagem Molecular , Potenciais da Membrana , Proteínas de Membrana/genética , Dados de Sequência Molecular , Potássio/fisiologia , Canais de Potássio/efeitos dos fármacos , Ratos , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia , Xenopus
5.
Science ; 273(5282): 1709-14, 1996 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-8781233

RESUMO

Members of a previously unidentified family of potassium channel subunits were cloned from rat and human brain. The messenger RNAs encoding these subunits were widely expressed in brain with distinct yet overlapping patterns, as well as in several peripheral tissues. Expression of the messenger RNAs in Xenopus oocytes resulted in calcium-activated, voltage-independent potassium channels. The channels that formed from the various subunits displayed differential sensitivity to apamin and tubocurare. The distribution, function, and pharmacology of these channels are consistent with the SK class of small-conductance, calcium-activated potassium channels, which contribute to the afterhyperpolarization in central neurons and other cell types.


Assuntos
Química Encefálica , Cálcio/metabolismo , Neurônios/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Sequência de Aminoácidos , Animais , Elementos Antissenso (Genética) , Apamina/farmacologia , Cálcio/farmacologia , Clonagem Molecular , Condutividade Elétrica , Feminino , Humanos , Potenciais da Membrana , Dados de Sequência Molecular , Oócitos , Técnicas de Patch-Clamp , Potássio/metabolismo , Bloqueadores dos Canais de Potássio , Canais de Potássio/análise , Canais de Potássio/química , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Xenopus
6.
Science ; 289(5486): 1942-6, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10988076

RESUMO

In excitable cells, small-conductance Ca2+-activated potassium channels (SK channels) are responsible for the slow after-hyperpolarization that often follows an action potential. Three SK channel subunits have been molecularly characterized. The SK3 gene was targeted by homologous recombination for the insertion of a gene switch that permitted experimental regulation of SK3 expression while retaining normal SK3 promoter function. An absence of SK3 did not present overt phenotypic consequences. However, SK3 overexpression induced abnormal respiratory responses to hypoxia and compromised parturition. Both conditions were corrected by silencing the gene. The results implicate SK3 channels as potential therapeutic targets for disorders such as sleep apnea or sudden infant death syndrome and for regulating uterine contractions during labor.


Assuntos
Trabalho de Parto/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Fenômenos Fisiológicos Respiratórios , Regiões 5' não Traduzidas , Potenciais de Ação , Animais , Encéfalo/metabolismo , Cruzamentos Genéticos , Técnicas de Cultura , Doxiciclina/farmacologia , Feminino , Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Marcação de Genes , Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Canais de Potássio/genética , Gravidez , Canais de Potássio Ativados por Cálcio de Condutância Baixa
7.
Neuron ; 14(2): 407-12, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7857648

RESUMO

Expression of the structurally and functionally distinct min K channel in Xenopus oocytes results in voltage-dependent potassium currents that activate with a characteristic slow time course. Application of a membrane-impermeable chemical cross-linking agent to oocytes expressing min K decreased the time-dependent current, increased its rate of activation, and induced persistently activated inward and outward potassium currents. These effects required membrane depolarization, demonstrating use dependence. Persistently activated channels retained potassium selectivity and sensitivity to block by clofilium and barium. These results suggest that a major conformational change occurs during min K channel gating, which can be stabilized by chemical cross-linking, and are consistent with a model in which min K channels activate by voltage-dependent subunit aggregation.


Assuntos
Canais de Potássio/fisiologia , Succinimidas/farmacologia , Animais , Bário/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Ativação do Canal Iônico , Substâncias Macromoleculares , Potenciais da Membrana/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Canais de Potássio/biossíntese , Compostos de Amônio Quaternário/farmacologia , RNA Mensageiro/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Xenopus laevis
8.
Neuron ; 15(6): 1449-54, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8845167

RESUMO

Episodic ataxia (EA) is an autosomal dominant human disorder that produces persistent myokymia and attacks of generalized ataxia. Recently, familial EA has been linked to the voltage-dependent delayed rectifier, Kv1.1, on chromosome 12. Six EA families have been identified that carry distinct Kv1.1 missense mutations; all individuals are heterozygous. Expression in Xenopus oocytes demonstrates that two of the EA subunits form homomeric channels with altered gating properties. V408A channels have voltage dependence similar to that of wild-type channels, but with faster kinetics and increased C-type inactivation, while the voltage dependence of F184C channels is shifted 20 mV positive. The other four EA subunits do not produce functional homomeric channels but reduce the potassium current when coassembled with wild-type subunits. The results suggest a cellular mechanism underlying EA in which the affected nerve cells cannot efficiently repolarize following an action potential because of altered delayed rectifier function.


Assuntos
Ataxia/genética , Canais de Potássio/fisiologia , Animais , Ataxia/fisiopatologia , Eletrofisiologia , Feminino , Humanos , Ativação do Canal Iônico , Cinética , Mutação Puntual , Recidiva , Xenopus
9.
Neuron ; 14(5): 1039-45, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7748551

RESUMO

Two inward rectifier potassium channels, the G protein-dependent GIRK1 and the G protein-independent BIR10, display large differences in rectification and macroscopic kinetics. A chimeric channel was constructed in which the putative intracellular carboxy-terminal domain of the G protein-dependent channel replaced the corresponding domain of the G protein-independent channel. The chimeric channel conducted potassium ions without the requirement of activated G proteins, yet displayed activation and deactivation kinetics and rectification properties similar to those of the G protein-dependent channel. The results demonstrate that structural elements in the C-terminus can independently control gating but not G protein signal transduction. The voltage dependence, time course, and kinetics of gating suggest a mechanism in which the pore may be occluded by reversible interactions with charged residues in the C-terminus.


Assuntos
Ativação do Canal Iônico/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/química , Canais de Potássio/fisiologia , Sequência de Aminoácidos , Condutividade Elétrica , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Proteínas de Ligação ao GTP/fisiologia , Cinética , Potenciais da Membrana , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/fisiologia , Transdução de Sinais , Relação Estrutura-Atividade
10.
Neuron ; 8(5): 975-82, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1586488

RESUMO

Tetraethylammonium ion (TEA+) blocks voltage-gated K+ channels by acting at two sites located at opposite ends of the aqueous pore. This allowed us to test two predictions made by models of ion permeation, namely that K+ channels can be simultaneously occupied by multiple ions and that the ions repel each other. We show that externally applied TEA+ antagonize block by internal TEA+ and vice versa. The antagonism is less than predicted for competitive binding, hence TEA+ may occupy both sites simultaneously. External TEA+ and internal TEA+ reduce each others affinity 4- to 5-fold. In addition, K+ antagonizes block by TEA+ at the opposite side of the membrane, and external TEA+ antagonizes is block by internal Ba2+. The antagonism between ions applied at opposite sides of the membrane may be common to all cations binding to K+ channels.


Assuntos
Canais de Potássio/fisiologia , Compostos de Tetraetilamônio/farmacologia , Animais , Bário/farmacologia , Ligação Competitiva , Cátions , Eletrofisiologia , Feminino , Oócitos/fisiologia , Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , RNA Mensageiro/genética , Tetraetilamônio , Compostos de Tetraetilamônio/antagonistas & inibidores , Compostos de Tetraetilamônio/metabolismo , Transfecção , Xenopus laevis
11.
Neuron ; 22(3): 549-58, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10197534

RESUMO

Elevated intracellular Ca2+ triggers inactivation of L-type calcium channels, providing negative Ca2+ feedback in many cells. Ca2+ binding to the main alpha1c channel subunit has been widely proposed to initiate such Ca2+ -dependent inactivation. Here, we find that overexpression of mutant, Ca2+ -insensitive calmodulin (CaM) ablates Ca2+ -dependent inactivation in a "dominant-negative" manner. This result demonstrates that CaM is the actual Ca2+ sensor for inactivation and suggests that CaM is constitutively tethered to the channel complex. Inactivation is likely to occur via Ca2+ -dependent interaction of tethered CaM with an IQ-like motif on the carboxyl tail of alpha1c. CaM also binds to analogous IQ regions of N-, P/Q-, and R-type calcium channels, suggesting that CaM-mediated effects may be widespread in the calcium channel family.


Assuntos
Canais de Cálcio Tipo N , Canais de Cálcio/fisiologia , Sinalização do Cálcio/fisiologia , Cálcio/fisiologia , Calmodulina/fisiologia , Sequência de Aminoácidos , Animais , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L , Calmodulina/biossíntese , Calmodulina/genética , Calmodulina/metabolismo , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Retroalimentação/fisiologia , Potenciais da Membrana , Dados de Sequência Molecular , Mutação , Técnicas de Patch-Clamp , Ratos
12.
Neuron ; 4(3): 405-11, 1990 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2317379

RESUMO

Voltage-dependent potassium currents were measured in Xenopus oocytes previously injected with RNAs generated in vitro from each of three cloned cDNAs (RBK1, RBK2, and RGK5). The currents differed in their sensitivities to blockade by tetraethylammonium (TEA; respective KDs 0.3, greater than 100, and 10 mM) and in their inactivation during a depolarizing pulse. Injections of RNA combinations (RBK1/RBK2 and RBK1/RGK5) caused currents that had TEA sensitivities different from those expected from the sum, in any proportion, of the two native channels. It is concluded that novel potassium channels are formed by the oocytes injected with two RNAs, presumably by heteropolymerization of subunits; such heteropolymerization would contribute functional diversity to voltage-dependent potassium channels in addition to that provided by a large gene family.


Assuntos
Proteínas de Membrana/fisiologia , Oócitos/fisiologia , Canais de Potássio/fisiologia , Animais , Encéfalo/metabolismo , Clonagem Molecular , DNA/genética , Feminino , Cinética , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Oócitos/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Ratos , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia , Xenopus laevis
13.
Neuron ; 8(3): 493-7, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1550674

RESUMO

RNAs encoding a wild-type (RBK1) and a mutant (RBK1(Y379V,V381T); RBK1*) subunit of voltage-dependent potassium channels were injected into Xenopus oocytes. When expressed separately, they made homotetrameric channels that differed about 100-fold in sensitivity to tetraethylammonium (TEA). Mixtures of channels having one, two, or three low affinity subunits were expressed by injecting various proportions of RBK1 and RBK1* RNAs. The affinity for TEA of these three channel species was deduced by fitting concentration-response curves for the inhibition of potassium currents. DNAs were also concatenated to construct a sequence that encoded two connected subunits, and channels that contained four, two, or no TEA-sensitive subunits were expressed. The results suggest that bound TEA interacts simultaneously with all four subunits.


Assuntos
Canais de Potássio/metabolismo , Compostos de Tetraetilamônio/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Técnicas In Vitro , Substâncias Macromoleculares , Potenciais da Membrana , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos/química , Oócitos , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Xenopus laevis
14.
Neuron ; 9(2): 209-16, 1992 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1497890

RESUMO

Calcium-activated potassium channels were expressed in Xenopus oocytes by injection of RNA transcribed in vitro from complementary DNAs derived from the slo locus of Drosophila melanogaster. Many cDNAs were found that encode closely related proteins of about 1200 aa. The predicted sequences of these proteins differ by the substitution of blocks of amino acids at five identified positions within the putative intracellular region between residues 327 and 797. Excised inside-out membrane patches showed potassium channel openings only with micromolar calcium present at the cytoplasmic side; activity increased steeply both with depolarization and with increasing calcium concentration. The single-channel conductance was 126 pS with symmetrical potassium concentrations. The mean open time of the channels was clearly different for channels having different substituent blocks of amino acids. The results suggest that alternative splicing gives rise to a large family of functionally diverse, calcium-activated potassium channels.


Assuntos
Cálcio/farmacologia , DNA/genética , Drosophila melanogaster/genética , Expressão Gênica , Canais de Potássio/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Cálcio/metabolismo , Clonagem Molecular , Condutividade Elétrica , Dados de Sequência Molecular , Oócitos/metabolismo , Reação em Cadeia da Polimerase , Canais de Potássio/química , Canais de Potássio/fisiologia , RNA Mensageiro/genética , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia , Transcrição Gênica , Transfecção , Xenopus
15.
Mol Cell Biol ; 11(5): 2547-57, 1991 May.
Artigo em Inglês | MEDLINE | ID: mdl-2017164

RESUMO

A novel human keratinocyte-derived autocrine factor (KAF) was purified from conditioned medium by using heparin affinity chromatography as the first step. Purified KAF stimulated the growth of normal human keratinocytes, mouse AKR-2B cells, and a mouse keratinocyte cell line (BALB/MK). Heparin sulfate inhibited KAF mitogenic activity on all cell types tested and inhibited the ability of KAF to compete with epidermal growth factor for cell surface binding. Interestingly, KAF stimulated the growth of BALB/MK cells at high cell density but failed to stimulate these cells at clonal density. Protein microsequencing of the first 20 NH2-terminal amino acid residues of purified KAF revealed identity to the NH2 terminus of human amphiregulin (AR). Northern (RNA) blot analysis with AR-specific cRNA demonstrated that human keratinocytes, as well as mammary epithelial cell cultures, expressed high levels of AR mRNA. In contrast, AR mRNA was not detected in normal human fibroblasts or melanocytes and was present at reduced levels in several mammary tumor cell lines. The mitogenic activity of purified AR was also shown to be inhibited by heparin sulfate, and an AR-specific enzyme-linked immunosorbent assay (ELISA) revealed that KAF and AR are antigenically related. We have previously shown that human keratinocytes can grow in an autocrine manner. Our present study demonstrates that one of the growth factors responsible for this autocrine growth (KAF) is similar or identical to AR and that KAF and AR bioactivity can be negatively regulated by heparin sulfate.


Assuntos
Glicoproteínas/genética , Substâncias de Crescimento/genética , Heparitina Sulfato/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular , Queratinócitos/fisiologia , Sequência de Aminoácidos , Anfirregulina , Animais , Sequência de Bases , Ligação Competitiva , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Replicação do DNA/efeitos dos fármacos , Família de Proteínas EGF , Fator de Crescimento Epidérmico/metabolismo , Glicoproteínas/isolamento & purificação , Glicoproteínas/farmacologia , Substâncias de Crescimento/isolamento & purificação , Substâncias de Crescimento/farmacologia , Humanos , Queratinócitos/efeitos dos fármacos , Masculino , Camundongos , Dados de Sequência Molecular , Sondas de Oligonucleotídeos , RNA Mensageiro/genética , Homologia de Sequência do Ácido Nucleico , Pele/citologia
16.
Curr Opin Neurobiol ; 5(3): 286-95, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7580150

RESUMO

Voltage-gated ion channels are composed of pore-forming subunits, as well as auxiliary subunits that modify the functions of these channels. In addition, the channels interact with other modulatory proteins in a more transient manner, although with significant effects on channel activity. Even though many second-messenger systems influence the voltage-gated ion channels, only in a few cases has clear evidence for direct protein-protein interactions been demonstrated. Recent biochemical and genetic studies have helped to elucidate the scope of the interactions between these ion channels and various modulatory proteins by determining the structures and functions of nonpore-forming subunits.


Assuntos
Canais Iônicos/fisiologia , Proteínas/metabolismo , Animais , Canais de Cálcio/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Estrutura Molecular , Canais de Potássio/fisiologia , Canais de Sódio/fisiologia
17.
Curr Opin Neurobiol ; 8(3): 321-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9687354

RESUMO

Calcium-activated potassium channels are fundamental regulators of neuronal excitability, participating in interspike interval and spike-frequency adaptation. For large-conductance calcium-activated potassium (BK) channels, recent experiments have illuminated the fundamental biophysical mechanisms of gating, demonstrating that BK channels are voltage gated and calcium modulated. Structurally, BK channels have been shown to possess an extracellular amino-terminal domain, different from other potassium channels. Domains and residues involved in calcium-gating, and perhaps calcium binding itself, have been identified. For small- and intermediate-conductance calcium-activated potassium channels, SK and IK channels, clones have only recently become available, and they show that SK channels are a distinct subfamily of potassium channels. The biophysical properties of SK channels demonstrate that kinetic differences between apamin-sensitive and apamin-insensitive slow afterhyperpolarizations are not attributable to intrinsic gating differences between the two subtypes. Interestingly, SK and IK channels may prove effective drug targets for diseases such as myotonic muscular dystrophy and sickle cell anemia.


Assuntos
Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Canais de Potássio Ativados por Cálcio de Condutância Alta , Dados de Sequência Molecular , Canais de Potássio/química , Estrutura Terciária de Proteína
18.
J Neurosci ; 19(20): 8830-8, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10516302

RESUMO

Small conductance Ca(2+)-activated potassium channels (SK channels) are coassembled complexes of pore-forming SK alpha subunits and calmodulin. We proposed a model for channel activation in which Ca2+ binding to calmodulin induces conformational rearrangements in calmodulin and the alpha subunits that result in channel gating. We now report fluorescence measurements that indicate conformational changes in the alpha subunit after calmodulin binding and Ca2+ binding to the alpha subunit-calmodulin complex. Two-hybrid experiments showed that the Ca(2+)-independent interaction of calmodulin with the alpha subunits requires only the C-terminal domain of calmodulin and is mediated by two noncontiguous subregions; the ability of the E-F hands to bind Ca2+ is not required. Although SK alpha subunits lack a consensus calmodulin-binding motif, mutagenesis experiments identified two positively charged residues required for Ca(2+)-independent interactions with calmodulin. Electrophysiological recordings of SK2 channels in membrane patches from oocytes coexpressing mutant calmodulins revealed that channel gating is mediated by Ca2+ binding to the first and second E-F hand motifs in the N-terminal domain of calmodulin. Taken together, the results support a calmodulin- and Ca(2+)-calmodulin-dependent conformational change in the channel alpha subunits, in which different domains of calmodulin are responsible for Ca(2+)-dependent and Ca(2+)-independent interactions. In addition, calmodulin is associated with each alpha subunit and must bind at least one Ca2+ ion for channel gating. Based on these results, a state model for Ca2+ gating was developed that simulates alterations in SK channel Ca2+ sensitivity and cooperativity associated with mutations in CaM.


Assuntos
Cálcio/fisiologia , Calmodulina/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Animais , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Feminino , Ativação do Canal Iônico/fisiologia , Modelos Biológicos , Oócitos , Canais de Potássio/genética , Ratos , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Xenopus
19.
Biochim Biophys Acta ; 1518(1-2): 36-46, 2001 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-11267657

RESUMO

Small conductance calcium-gated K(+) channels (SK channels) are encoded by the three SK genes, SK1, SK2, and SK3. These channels likely contribute to slow synaptic afterhyperpolarizations of apamin-sensitive and apamin-insensitive types. SK channels are also widely expressed outside the nervous system. The mouse SK1 gene comprises at least 12 exons extending across 19.8 kb of genomic DNA. This gene encodes a complex pattern of alternatively spliced SK1 transcripts widely expressed among mouse tissues. These transcripts exhibit at least four distinct 5'-nucleotide sequence variants encoding at least two N-terminal amino acid sequences. Optional inclusion of exons 7 and 9, together with two alternate splice donor sites in exon 8, yields transcripts encoding eight variant C-terminal amino acid sequences for SK1. These include an altered putative S6 transmembrane span, modification of the C-terminal cytoplasmic domain binding site for calmodulin, and generation of two alternate predicted binding sites for PDZ domain-containing proteins. 20 of the 32 predicted mouse SK1 transcripts are expressed in brain at levels sufficient to allow consistent detection, and encode 16 SK1 polypeptide variants. Only four of these 16 polypeptides preserve the ability to bind calmodulin in a Ca(2+)-independent manner. Mouse SK1 also exhibits novel, strain-specific, length polymorphism of a polyglutamate repeat in the N-terminal cytoplasmic domain. The evolutionary conservation of this complex transcription pattern suggests a possible role in the tuning of SK1 channel function.


Assuntos
Perfilação da Expressão Gênica , Canais de Potássio Cálcio-Ativados , Canais de Potássio/genética , Transcrição Gênica , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Calmodulina/metabolismo , Clonagem Molecular , Citoplasma/metabolismo , DNA Complementar , Humanos , Camundongos , Dados de Sequência Molecular , Peptídeos/genética , Ácido Poliglutâmico , Polimorfismo Genético , Canais de Potássio/metabolismo , RNA Mensageiro , Ratos , Sequências Repetitivas de Aminoácidos , Homologia de Sequência de Aminoácidos , Canais de Potássio Ativados por Cálcio de Condutância Baixa
20.
J Gen Physiol ; 111(4): 565-81, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9524139

RESUMO

Small-conductance Ca-activated K+ channels play an important role in modulating excitability in many cell types. These channels are activated by submicromolar concentrations of intracellular Ca2+, but little is known about the gating kinetics upon activation by Ca2+. In this study, single channel currents were recorded from Xenopus oocytes expressing the apamin-sensitive clone rSK2. Channel activity was detectable in 0.2 micro M Ca2+ and was maximal above 2 micro M Ca2+. Analysis of stationary currents revealed two open times and three closed times, with only the longest closed time being Ca dependent, decreasing with increasing Ca2+ concentrations. In addition, elevated Ca2+ concentrations resulted in a larger percentage of long openings and short closures. Membrane voltage did not have significant effects on either open or closed times. The open probability was approximately 0.6 in 1 micro M free Ca2+. A lower open probability of approximately 0.05 in 1 micro M Ca2+ was also observed, and channels switched spontaneously between behaviors. The occurrence of these switches and the amount of time channels spent displaying high open probability behavior was Ca2+ dependent. The two behaviors shared many features including the open times and the short and intermediate closed times, but the low open probability behavior was characterized by a different, long Ca2+-dependent closed time in the range of hundreds of milliseconds to seconds. Small-conductance Ca- activated K+ channel gating was modeled by a gating scheme consisting of four closed and two open states. This model yielded a close representation of the single channel data and predicted a macroscopic activation time course similar to that observed upon fast application of Ca2+ to excised inside-out patches.


Assuntos
Cálcio/farmacologia , Ativação do Canal Iônico/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/efeitos dos fármacos , Animais , Apamina/farmacologia , Cálcio/fisiologia , Condutividade Elétrica , Eletrofisiologia , Feminino , Membranas Intracelulares/metabolismo , Cinética , Modelos Biológicos , Oócitos , Canais de Potássio/fisiologia , Ratos , Proteínas Recombinantes , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Fatores de Tempo , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA