Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros








Base de dados
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.
Neuroscience ; 217: 67-76, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22554781

RESUMO

The firing properties of dopamine (DA) neurons in the substantia nigra (SN) pars compacta are strongly influenced by the activity of apamin-sensitive small conductance Ca(2+)-activated K(+) (SK) channels. Of the three SK channel genes expressed in central neurons, only SK3 expression has been identified in DA neurons. The present findings show that SK2 was also expressed in DA neurons. Immuno-electron microscopy (iEM) showed that SK2 was primarily expressed in the distal dendrites, while SK3 was heavily expressed in the soma and, to a lesser extent, throughout the dendritic arbor. Electrophysiological recordings of the effects of the SK channel blocker apamin on DA neurons from wild type and SK(-/-) mice show that SK2-containing channels contributed to the precision of action potential (AP) timing, while SK3-containing channels influenced AP frequency. The expression of SK2 in DA neurons may endow distinct signaling and subcellular localization to SK2-containing channels.


Assuntos
Potenciais de Ação/fisiologia , Neurônios Dopaminérgicos/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Apamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Camundongos , Camundongos Knockout , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
3.
J Physiol ; 548(Pt 3): 881-91, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12626668

RESUMO

Rat and sheep cardiac myocytes become binucleate as they complete the 'terminal differentiation' process soon after birth and are not able to divide thereafter. Angiotensin II (Ang II) is known to stimulate hypertrophic changes in rodent cardiomyocytes under both in vivo and in vitro conditions via the AT1 receptor and intracellular extracellular regulated kinase (ERK) signalling cascade. We sought to develop culture methods for immature sheep cardiomyocytes in order to test the hypothesis that Ang II is a hypertrophic agent in the immature myocardium of the sheep. We isolated fetal sheep cardiomyocytes and cultured them for 96 h, added Ang II and phenylephrine (PE) for 48 h, and measured footprint area and proliferation (5-bromo-2'-deoxyuridine (BrdU) uptake) separately in mono- vs. binucleate myocytes. We found that neither Ang II nor PE changed the footprint area of mononucleated cells. PE stimulated an increase in footprint area of binucleate cells but Ang II did not. Ang II increased myocyte BrdU uptake compared to serum free conditions, but PE did not affect BrdU uptake. The MAP kinase kinase (MEK) inhibitor UO126 prevented BrdU uptake in Ang II-stimulated cells and prevented cell hypertrophy in PE-stimulated cells. This paper establishes culture methods for immature sheep cardiomyocytes and reports that: (1) Ang II is not a hypertrophic agent; (2) Ang II stimulates hyperplastic growth among mononucleate myocytes; (3) PE is a hypertrophic agent in binucleate myocytes; and (4) the ERK cascade is required for the proliferation effect of Ang II and the hypertrophic effect of PE.


Assuntos
Angiotensina II/farmacologia , Coração/fisiologia , Miocárdio/patologia , Função Ventricular , Animais , Bromodesoxiuridina/farmacocinética , Técnicas de Cultura de Células/métodos , Células Cultivadas , Feminino , Feto , Idade Gestacional , Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Hiperplasia , Hipertrofia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Gravidez , Ovinos
4.
J Physiol ; 536(Pt 2): 397-407, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11600675

RESUMO

1. Hyperexcitability in denervated skeletal muscle is associated with the expression of SK3, a small-conductance Ca2+-activated K+ channel (SK channel). SK currents were examined in dissociated fibres from flexor digitorum brevis (FDB) muscle using the whole-cell patch clamp configuration. 2. Depolarization activated a K+-selective, apamin-sensitive and iberiotoxin-insensitive current, detected as a tail current upon repolarization, in fibres from denervated but not innervated muscle. Dialysis of the fibres with 20 mM EGTA in the patch pipette solution eliminated the tail current, consistent with this current reflecting Ca2+-activated SK channels expressed only in denervated muscle. 3. Activation of SK tail currents depended on the duration of the depolarizing pulse, consistent with a rise in intracellular Ca2+ due to release from the sarcoplasmic reticulum (SR) and influx through voltage-gated Ca2+ channels. 4. The envelope of SK tail currents was diminished by 10 microM ryanodine for all pulse durations, whereas 2 mM cobalt reduced the SK tail current for pulses greater than 80 ms, demonstrating that Ca2+ release from the SR during short pulses primarily activated SK channels. 5. In current clamp mode with the resting membrane potential set at -70 mV, denervation decreased the action potential threshold by approximately 8 mV. Application of apamin increased the action potential threshold in denervated fibres to that measured in innervated fibres, suggesting that SK channel activity modulates the apparent action potential threshold. 6. These results are consistent with a model in which SK channel activity in the T-tubules of denervated skeletal muscle causes a local increase in K+ concentration that results in hyperexcitability.


Assuntos
Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Apamina/farmacologia , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Quelantes/farmacologia , Cobalto/farmacologia , Ácido Egtázico/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Denervação Muscular , Músculo Esquelético/citologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Potássio/metabolismo , Retículo Sarcoplasmático/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa
6.
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
7.
Am J Physiol Regul Integr Comp Physiol ; 279(4): R1157-64, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11003978

RESUMO

The effects of right ventricular (RV) systolic pressure (RVSP) load on fetal myocyte size and maturation were studied. Pulmonary artery (PA) pressure was increased by PA occlusion from mean 47.4 +/- 5.0 (+/-SD) to 71 +/- 13.6 mmHg (P < 0.0001) in eight RVSP-loaded near-term fetal sheep for 10 days. The maximal pressure generated by the RV with acute PA occlusion increased after RVSP load: 78 +/- 7 to 101 +/- 15 mmHg (P < 0.005). RVSP-load hearts were heavier (44.7 +/- 8.4 g) than five nonloaded hearts (31.8 +/- 0.2 g; P < 0.03); heart-to-body weight ratio (10.9 +/- 1.1 and 6.5 +/- 0.9 g/kg, respectively; P < 0.0001). RVSP-RV myocytes were longer (101.3 +/- 10.2 microm) than nonloaded RV myocytes (88.2 +/- 8.1 microm; P < 0. 02) and were more often binucleated (82 +/- 13%) than nonloaded myocytes (63 +/- 7%; P < 0.02). RVSP-loaded myocytes had less myofibrillar volume than did nonloaded hearts (44.1 +/- 4.4% and 56. 1 +/- 2.6%; P < 0.002). We conclude that RV systolic load 1) leads to RV myocyte enlargement, 2) has minor effects on left ventricular myocyte size, and 3) stimulates maturation (increased RV myocyte binucleation). Myocyte volume data suggest that RV systolic loading stimulates both hyperplastic and hypertrophic growth.


Assuntos
Pressão Sanguínea , Coração/embriologia , Hemodinâmica/fisiologia , Miocárdio/citologia , Artéria Pulmonar/embriologia , Função Ventricular Direita/fisiologia , Animais , Função do Átrio Direito , Divisão Celular , Feminino , Feto , Idade Gestacional , Miocárdio/ultraestrutura , Gravidez , Ovinos , Sístole
8.
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
9.
Nat Genet ; 25(1): 105-9, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10802667

RESUMO

Expansion of a CTG trinucleotide repeat in the 3' UTR of the gene DMPK at the DM1 locus on chromosome 19 causes myotonic dystrophy, a dominantly inherited disease characterized by skeletal muscle dystrophy and myotonia, cataracts and cardiac conduction defects. Targeted deletion of Dm15, the mouse orthologue of human DMPK, produced mice with a mild myopathy and cardiac conduction abnormalities, but without other features of myotonic dystrophy, such as myotonia and cataracts. We, and others, have demonstrated that repeat expansion decreases expression of the adjacent gene SIX5 (refs 7,8), which encodes a homeodomain transcription factor. To determine whether SIX5 deficiency contributes to the myotonic dystrophy phenotype, we disrupted mouse Six5 by replacing the first exon with a beta-galactosidase reporter. Six5-mutant mice showed reporter expression in multiple tissues, including the developing lens. Homozygous mutant mice had no apparent abnormalities of skeletal muscle function, but developed lenticular opacities at a higher rate than controls. Our results suggest that SIX5 deficiency contributes to the cataract phenotype in myotonic dystrophy, and that myotonic dystrophy represents a multigenic disorder.


Assuntos
Catarata/etiologia , Catarata/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Distrofia Miotônica/genética , Regiões 3' não Traduzidas/genética , Animais , Catarata/enzimologia , Catarata/patologia , Éxons/genética , Marcação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distrofia Miotônica/enzimologia , Miotonina Proteína Quinase , Proteínas Serina-Treonina Quinases/genética , Expansão das Repetições de Trinucleotídeos/genética
10.
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
11.
Biophys J ; 77(4): 1905-13, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10512811

RESUMO

The activation of small-conductance calcium-activated potassium channels (SK) has a profound effect on membrane excitability. In hippocampal pyramidal neurons, SK channel activation by Ca2+ entry from a preceding burst of action potentials generates the slow afterhyperpolarization (AHP). Stimulation of a number of receptor types suppresses the slow AHP, inhibiting spike frequency adaptation and causing these neurons to fire tonically. Little is known of the gating properties of native SK channels in CNS neurons. By using excised inside-out patches, a small-amplitude channel has been resolved that was half-activated by approximately 0.6 microM Ca2+ in a voltage-independent manner. The channel possessed a slope conductance of 10 pS and exhibited nonstationary gating. These properties are in accord with those of cloned SK channels. The measured Ca2+ sensitivity of hippocampal SK channels suggests that the slow AHP is generated by activation of SK channels from a local rise of intracellular Ca2+.


Assuntos
Hipocampo/citologia , Ativação do Canal Iônico , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Células Piramidais/metabolismo , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Condutividade Elétrica , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Canais de Potássio Ativados por Cálcio de Condutância Baixa
12.
Pflugers Arch ; 438(3): 314-21, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10398861

RESUMO

Small-conductance calcium-activated potassium channels (SK channels) are gated solely by intracellular calcium ions and their activity is responsible for the slow afterhyperpolarization (AHP) that follows an action potential in many excitable cells. Brain slice studies commonly employ a methyl derivative of bicuculline (bicuculline-m), a GABAA (gamma-aminobutyric acid) receptor antagonist, to diminish the tonic inhibitory influences of GABAergic synapses, or to investigate the role of these synapses in specialized neural networks. However, recent evidence suggests that bicuculline-m may not be specific for GABAA receptors and may also block the slow AHP. Therefore, the effects of bicuculline-m on cloned apamin-sensitive SK2 and apamin-insensitive SK1 channels were examined following expression in Xenopus oocytes. The results show that at concentrations employed for slice recordings, bicuculline-m potently blocks both apamin-sensitive SK2 currents and apamin-insensitive SK1 currents when applied to outside-out patches. Apamin-insensitive SK1 currents run down in excised patches. The potency of bicuculline-m block also decreases with time after patch excision. Site-directed mutagenesis that changes two residues in the outer vestibule of the SK1 pore that confers apamin sensitivity also reduces run down of the current in patches, and endows stable sensitivity to bicuculline-m indistinguishable from SK2. Therefore, the use of bicuculline-m in slice recordings may mask apamin-sensitive slow AHPs that are important determinants of neuronal excitability. In addition, bicuculline-m-insensitive slow AHPs may indicate that the underlying channels have run down.


Assuntos
Bicuculina/farmacologia , Cálcio/farmacologia , Antagonistas GABAérgicos/farmacologia , Bloqueadores dos Canais de Potássio , Canais de Potássio/fisiologia , Animais , Apamina/farmacologia , Condutividade Elétrica , Feminino , Expressão Gênica , Cinética , Oócitos/metabolismo , Canais de Potássio/genética , Proteínas Recombinantes , Xenopus
13.
Ann N Y Acad Sci ; 868: 370-8, 1999 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-10414306

RESUMO

SK channels play a fundamental role in all excitable cells. SK channels are potassium selective and are activated by an increase in the level of intracellular calcium, such as occurs during an action potential. Their activation causes membrane hyperpolarization, which inhibits cell firing and limits the firing frequency of repetitive action potentials. The intracellular calcium increase evoked by action potential firing decays slowly, allowing SK channel activation to generate a long-lasting hyperpolarization termed the slow afterhyperpolarization (sAHP). This spike-frequency adaptation protects the cell from the deleterious effects of continuous tetanic activity and is essential for normal neurotransmission. Slow AHPs can be classified into two groups, based on sensitivity to the bee venom toxin apamin. In general, apamin-sensitive sAHPs activate rapidly following a single action potential and decay with a time constant of approximately 150 ms. In contrast, apamin-insensitive sAHPs rise slowly and decay with a time constant of approximately 1.5 s. The basis for this kinetic difference is not yet understood. Apamin-sensitive and apamin-insensitive SK channels have recently been cloned. This chapter will compare with different classes of sAHPs, discuss the cloned SK channels and how they are gated by calcium ions, describe the molecular basis for their different pharmacologies, and review the possible role of SK channels in several pathological conditions.


Assuntos
Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Potenciais de Ação/fisiologia , Animais , Apamina , Cálcio/metabolismo , Clonagem Molecular , Humanos , Ativação do Canal Iônico , Cinética , Modelos Moleculares , Distrofia Miotônica/genética , Neurotransmissores/farmacologia , Canais de Potássio/genética , Transtornos do Sono-Vigília/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa
14.
Muscle Nerve ; 22(6): 742-50, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10366228

RESUMO

Skeletal muscle becomes hyperexcitable following denervation and when cultured in the absence of nerve cells. In these circumstances, the bee venom peptide toxin apamin, a blocker of small-conductance calcium-activated potassium (SK) channels, dramatically reduces the hyperexcitability. In this report, we show that SK3 channels are expressed in denervated skeletal muscle and in L6 cells. Action potentials evoked from normal innervated rat skeletal muscle did not exhibit an afterhyperpolarization, indicating a lack of SK channel activity; very low levels of apamin binding sites, SK3 protein, or SK3 mRNA were present. However, denervation resulted in apamin-sensitive afterhyperpolarizations and increased apamin binding sites, SK3 protein, and SK3 mRNA. Cultured rat L6 myoblasts and differentiated L6 myotubes contained similar levels of SK3 mRNA, although apamin-sensitive SK currents and apamin binding sites were detected only following myotube differentiation. Therefore, different molecular mechanisms govern SK3 expression levels in denervated muscle compared with muscle cells differentiated in culture.


Assuntos
Cálcio/farmacologia , Músculo Esquelético/fisiologia , Condução Nervosa , Canais de Potássio/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Mapeamento Cromossômico , Genoma Humano , Humanos , Denervação Muscular , Ratos , Células-Tronco/efeitos dos fármacos
15.
Nature ; 395(6701): 503-7, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9774106

RESUMO

The slow afterhyperpolarization that follows an action potential is generated by the activation of small-conductance calcium-activated potassium channels (SK channels). The slow afterhyperpolarization limits the firing frequency of repetitive action potentials (spike-frequency adaptation) and is essential for normal neurotransmission. SK channels are voltage-independent and activated by submicromolar concentrations of intracellular calcium. They are high-affinity calcium sensors that transduce fluctuations in intracellular calcium concentrations into changes in membrane potential. Here we study the mechanism of calcium gating and find that SK channels are not gated by calcium binding directly to the channel alpha-subunits. Instead, the functional SK channels are heteromeric complexes with calmodulin, which is constitutively associated with the alpha-subunits in a calcium-independent manner. Our data support a model in which calcium gating of SK channels is mediated by binding of calcium to calmodulin and subsequent conformational alterations in the channel protein.


Assuntos
Cálcio/metabolismo , Ativação do Canal Iônico , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Animais , Sítios de Ligação , Calmodulina/genética , Calmodulina/metabolismo , Eletrofisiologia , Glutationa Transferase/metabolismo , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Xenopus
16.
FEBS Lett ; 431(3): 461-4, 1998 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-9714564

RESUMO

Episodic ataxia (EA) is a rare inherited neurological disorder due to mutation in the voltage-dependent Kv1.1 potassium channel. In nine unrelated families, a different missense point mutation at highly conserved positions has been reported. We have previously characterized six of the EA mutants. In this study, three recently identified mutations were introduced into the human Kv1.1 cDNA and expressed in Xenopus oocytes. Compared to wild type, T226A and T226M reduced the current amplitude by > 95%, shifted the voltage dependence by 15 mV, and slowed activation and deactivation kinetics. Currents from G311S were approximately 25% of wild type, less steeply voltage-dependent and had more pronounced C-type inactivation. These altered gating properties will reduce the delayed-rectifier potassium current which may underlie the symptoms of EA.


Assuntos
Ataxia/genética , Mutação , Canais de Potássio/genética , Animais , Humanos , Xenopus
17.
J Biol Chem ; 273(34): 21542-53, 1998 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-9705284

RESUMO

We have cloned from murine erythroleukemia (MEL) cells, thymus, and stomach the cDNA encoding the Ca2+-gated K+ (KCa) channel, mIK1, the mouse homolog of hIK1 (Ishii, T. M., Silvia, C., Hirschberg, B., Bond, C. T., Adelman, J. P., and Maylie, J. (1997) Proc. Natl. Acad. Sci.(U. S. A. 94, 11651-11656). mIK1 mRNA was detected at varied levels in many tissue types. mIK1 KCa channel activity expressed in Xenopus oocytes closely resembled the Kca of red cells (Gardos channel) and MEL cells in its single channel conductance, lack of voltage-sensitivity of activation, inward rectification, and Ca2+ concentration dependence. mIK1 also resembled the erythroid channel in its pharmacological properties, mediating whole cell and unitary currents sensitive to low nM concentrations of both clotrimazole (CLT) and its des-imidazolyl metabolite, 2-chlorophenyl-bisphenyl-methanol, and to low nM concentrations of iodocharybdotoxin. Whereas control oocytes subjected to hypotonic swelling remained swollen, mIK1 expression conferred on oocytes a novel, Ca2+-dependent, CLT-sensitive regulatory volume decrease response. Hypotonic swelling of voltage-clamped mIK1-expressing oocytes increased outward currents that were Ca2+-dependent, CLT-sensitive, and reversed near the K+ equilibrium potential. mIK1 mRNA levels in ES cells increased steadily during erythroid differentiation in culture, in contrast to other KCa mRNAs examined. Low nanomolar concentrations of CLT inhibited proliferation and erythroid differentiation of peripheral blood stem cells in liquid culture.


Assuntos
Canais de Potássio Cálcio-Ativados , Canais de Potássio/genética , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular , Clonagem Molecular , Clotrimazol/farmacologia , DNA Complementar/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Vírus da Leucemia Murina de Friend , Glicosilação , Soluções Hipotônicas , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Ativação do Canal Iônico , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Dados de Sequência Molecular , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas , Xenopus
19.
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
20.
Biophys J ; 74(5): 2299-305, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9591657

RESUMO

The channel underlying the slow component of the voltage-dependent delayed outward rectifier K+ current, I(Ks), in heart is composed of the minK and KvLQT1 proteins. Expression of the minK protein in Xenopus oocytes results in I(Ks)-like currents, I(sK), due to coassembly with the endogenous XKvLQT1. The kinetics and voltage-dependent characteristics of I(sK) suggest a distinct mechanism for voltage-dependent gating. Currents recorded at 40 mV from holding potentials between -60 and -120 mV showed an unusual "cross-over," with the currents obtained from more depolarized holding potentials activating more slowly and deviating from the Cole-Moore prediction. Analysis of the current traces revealed two components with fast and slow kinetics that were not affected by the holding potential. Rather, the relative contribution of the fast component decreased with depolarized holding potentials. Deactivation and reactivation, after a short period of repolarization (100 ms), was markedly faster than the fast component of activation. These gating properties suggest a physiological mechanism by which cardiac I(Ks) may suppress premature action potentials.


Assuntos
Membrana Celular/fisiologia , Ativação do Canal Iônico/fisiologia , Oócitos/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Animais , Feminino , Coração/fisiologia , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Potenciais da Membrana , Modelos Biológicos , Canais de Potássio/biossíntese , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA