Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 146(4): 1652-61, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17478049

RESUMO

The Slo1 gene, also known as KCNMA1, encodes the pore-forming subunits of large-conductance Ca2+-activated K+ (BK(Ca)) channels. Products of this gene are widely expressed in vertebrate tissues, and occur in a large number (>or=20) of alternatively spliced variants that vary in their gating properties, susceptibility to modulation, and trafficking to the plasma membrane. Motifs in the large cytoplasmic C-terminal are especially important in determining the functional properties of BK(Ca) channels. Here we report that chick ciliary ganglion neurons express transcripts and proteins of two Slo1 splice variants that differ at the extreme C-terminal. We refer to these variants as VEDEC and QEDRL (or QEERL for the orthologous mammalian versions), after the five terminal amino acid residues in each isoform. Individual ciliary ganglion neurons preferentially express these variants in different subcellular compartments. Moreover, QEERL channels show markedly higher levels of constitutive expression on the plasma membrane than VEDEC channels in HEK293T and NG108-15 cells. However, growth factor treatment can stimulate surface expression of VEDEC channels to levels comparable to those seen with QEERL. In addition, we show that co-expression of a soluble protein composed of VEDEC C-terminal tail residues markedly increases cell surface expression of full-length VEDEC channels, suggesting that this region binds to proteins that cause retention of the these channels in intracellular stores.


Assuntos
Processamento Alternativo/fisiologia , Regulação da Expressão Gênica/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Neurônios/metabolismo , Análise de Variância , Animais , Biotinilação/métodos , Células Cultivadas , Embrião de Galinha , Gânglios Parassimpáticos/citologia , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Potenciais da Membrana/genética , Camundongos , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , RNA Mensageiro/biossíntese , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transfecção/métodos
2.
Neuroscience ; 143(3): 655-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17081697

RESUMO

Myasthenia gravis can be induced in mice by injecting the extracellular domain of rat muscle-specific kinase (MuSK), a transmembrane receptor tyrosine kinase involved in agrin signaling at the neuromuscular junction. About 5-10% of human myasthenia gravis patients have autoantibodies against MuSK. Here we have examined mouse neuromuscular junctions following MuSK immunization in two groups of muscles that can be distinguished on the basis of the timing of neuromuscular synaptogenesis and their response to perturbation of agrin signaling. We used confocal microscopy to characterize the distribution and expression of nicotinic acetylcoline receptors and of two presynaptic makers, neurofilament protein and synaptophysin. We observed disruption of neuromuscular junctions in all muscles examined in this model of myasthenia gravis. However delayed-synapsing muscles, including the diaphragm, sternomastoid and tibialis posterior, were significantly more severely affected than fast-synapsing muscles, including the intercostal, adductor longus and tibialis anterior. These results suggest a basis for the differential susceptibility of muscles in different classes of myasthenia gravis patients, including patients with autoantibodies against MuSK.


Assuntos
Músculos/patologia , Miastenia Gravis/patologia , Junção Neuromuscular/metabolismo , Receptores Proteína Tirosina Quinases , Receptores Colinérgicos , Agrina/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal/métodos , Músculos/metabolismo , Miastenia Gravis/induzido quimicamente , Proteínas de Neurofilamentos/metabolismo , Ratos , Receptores Nicotínicos/metabolismo , Sinaptofisina/metabolismo , Fatores de Tempo
3.
Neuroscience ; 135(4): 1013-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16165293

RESUMO

Endogenous beta-neuregulin-1 is required for the plasma membrane expression of large-conductance (BK-type) Ca2+-activated K+ channels in developing chick ciliary neurons of the chick ciliary ganglion. During normal development, beta-neuregulin-1 acts in concert with transforming growth factor-beta1 to stimulate movement of large-conductance Ca2+-activated K+ channels from intracellular stores into the plasma membrane, although these two growth factors preferentially act on different intracellular pools. We have previously shown that actions of transforming growth factor-beta1 on ciliary neurons require activation of phosphoinositol 3-kinase and Akt, as well as a parallel cascade composed of the small GTPase Ras and a mitogen-activated protein kinase (extracellular signal-regulated kinase). In addition, we have shown that the actions of beta-neuregulin-1 require activation of phosphoinositol 3-kinase and the protein kinase Akt. Here we examine whether beta-neuregulin-1-evoked mobilization of large-conductance Ca2+-activated K+ channels also requires activation of a Ras-extracellular signal-regulated kinase signaling cascade. We observed that application of beta-neuregulin-1 caused a robust and MEK1/2-dependent increase in extracellular signal-regulated kinase diphosphorylation that indicates activation of this signaling cascade in ciliary ganglion neurons, similar to what we have previously observed for transforming growth factor-beta1. However, activation of this cascade is not necessary for beta-neuregulin-1-evoked mobilization because stimulation of macroscopic large-conductance Ca2+-activated K+ channels persisted in cells treated with the MEK1/2 inhibitors PD98059 or U0126, in cells over-expressing dominant-negative forms of extracellular signal-regulated kinase, and in cells treated with the Ras inhibitor FTI-277. These results indicate that the mechanisms that underlie beta-neuregulin-1 and transforming growth factor-beta1 mobilization of large-conductance Ca2+-activated K+ channels are only partly overlapping, possibly because they cause recruitment of spatially distinct signaling complexes.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neuregulina-1/metabolismo , Neurônios/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Proteínas ras/metabolismo , Animais , Células Cultivadas , Embrião de Galinha , Ativação Enzimática/fisiologia , Immunoblotting , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
4.
Nature ; 411(6835): 313-7, 2001 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-11357134

RESUMO

Cryptochromes are flavin/pterin-containing proteins that are involved in circadian clock function in Drosophila and mice. In mice, the cryptochromes Cry1 and Cry2 are integral components of the circadian oscillator within the brain and contribute to circadian photoreception in the retina. In Drosophila, cryptochrome (CRY) acts as a photoreceptor that mediates light input to circadian oscillators in both brain and peripheral tissue. A Drosophila cry mutant, cryb, leaves circadian oscillator function intact in central circadian pacemaker neurons but renders peripheral circadian oscillators largely arrhythmic. Although this arrhythmicity could be caused by a loss of light entrainment, it is also consistent with a role for CRY in the oscillator. A peripheral oscillator drives circadian olfactory responses in Drosophila antennae. Here we show that CRY contributes to oscillator function and physiological output rhythms in the antenna during and after entrainment to light-dark cycles and after photic input is eliminated by entraining flies to temperature cycles. These results demonstrate a photoreceptor-independent role for CRY in the periphery and imply fundamental differences between central and peripheral oscillator mechanisms in Drosophila.


Assuntos
Relógios Biológicos/fisiologia , Ritmo Circadiano/fisiologia , Proteínas de Drosophila , Drosophila melanogaster/fisiologia , Proteínas do Olho , Flavoproteínas/metabolismo , Animais , Relógios Biológicos/genética , Encéfalo/citologia , Encéfalo/fisiologia , Criptocromos , Escuridão , Drosophila melanogaster/genética , Extremidades/fisiologia , Flavoproteínas/genética , Regulação da Expressão Gênica , Genes Reporter/genética , Luz , Mutação/genética , Neurônios/fisiologia , Fenótipo , Estimulação Luminosa , Células Fotorreceptoras de Invertebrados/metabolismo , Receptores Acoplados a Proteínas G , Olfato/fisiologia , Temperatura , Fatores de Tempo
5.
Neuron ; 29(1): 255-66, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11182096

RESUMO

cGMP-gated channels are essential for phototransduction in the vertebrate retina. Here we show that the affinity of these channels for cGMP in chick cones is substantially higher during the subjective night than during the subjective day. This effect persists in constant environmental conditions after entrainment to 12:12 hr light-dark cycles in vitro or in ovo. Circadian modulation of ligand affinity is a posttranslational effect and is driven by rhythms in the activities of two protein kinases: Erk and Ca2+/calmodulin-dependent protein kinase II (CaMKII). Erk is maximally active during the subjective night, whereas CaMKII is maximally active during the subjective day. Acute inhibition of these signaling pathways causes phase-dependent changes in the affinity of the channels for cGMP.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Ritmo Circadiano/fisiologia , Canais Iônicos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Animais , Relógios Biológicos/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Células Cultivadas , Embrião de Galinha , GMP Cíclico/metabolismo , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Escuridão , Inibidores Enzimáticos/farmacologia , Proteínas do Olho/metabolismo , Ativação do Canal Iônico/fisiologia , Ligantes , Luz , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Células Fotorreceptoras Retinianas Cones/citologia , Transdução de Sinais
6.
Proc Natl Acad Sci U S A ; 98(5): 2832-6, 2001 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11226326

RESUMO

The development of functional Ca(2+)-activated K(+) channels (K(Ca)) in chick ciliary ganglion (CG) neurons requires interactions with afferent preganglionic nerve terminals. Here we show that the essential preganglionic differentiation factor is an isoform of beta-neuregulin-1. beta-Neuregulin-1 transcripts are expressed in the midbrain preganglionic Edinger-Westphal nucleus at developmental stages that coincide with or precede the normal onset of macroscopic K(Ca) in CG neurons. Injection of beta-neuregulin-1 peptide into the brains of developing embryos evoked a robust stimulation of functional K(Ca) channels at stages before the normal appearance of these channels in CG neurons developing in vivo. Conversely, injection of a neutralizing antiserum specific for beta-neuregulin-1 inhibited the development of K(Ca) channels in CG neurons. Low concentrations of beta-neuregulin-1 evoked a robust increase in whole-cell K(Ca) in CG neurons cocultured with iris target tissues. By contrast, culturing CG neurons with iris cells or low concentrations of beta-neuregulin-1 by themselves was insufficient to stimulate K(Ca). These data suggest that the preganglionic factor required for the development of K(Ca) in ciliary ganglion neurons is an isoform of beta-neuregulin-1, and that this factor acts in concert with target-derived trophic molecules to regulate the differentiation of excitability.


Assuntos
Cálcio/metabolismo , Neuregulina-1/fisiologia , Neurônios/metabolismo , Sistema Nervoso Parassimpático/metabolismo , Canais de Potássio/fisiologia , Animais , Sequência de Bases , Embrião de Galinha , Técnicas de Cocultura , Primers do DNA , DNA Complementar , Neuregulina-1/genética , Sistema Nervoso Parassimpático/citologia , Sistema Nervoso Parassimpático/embriologia , Canais de Potássio/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
J Neurosci ; 21(1): 221-9, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11150339

RESUMO

The cyclic GMP-gated cationic channels of vertebrate photoreceptors are essential for visual phototransduction. We have examined the developmental regulation of cGMP-gated channels in morphologically identified cones in the chick retina. Expression of cone-type cGMP-gated channel mRNA can be detected at embryonic day 6 (E6), but expression of functional channels, as accessed by patch-clamp recordings, cannot be detected until E8. Plasma membrane channels in embryonic cones have a high turnover rate because inhibition of protein synthesis or disruption of the Golgi apparatus causes an almost complete loss of functional cGMP-gated channels within 12 hr. Different subpopulations of cones begin to express functional channels at different developmental stages, but all cones express channels by E10. Expression of cGMP-gated channels in at least one cone subpopulation appears to require one or more soluble differentiation factors, which are presumably present in the normal microenvironment of the developing retina. Application of chick embryo extract (CEE), a rich source of trophic factors, causes marked stimulation of cGMP-gated channel expression in chick cones at E8, but not at E6. Inhibition of MAP kinase (Erk) signaling using PD98059, or inhibition of PI3 kinase signaling by LY294002, blocked the stimulatory effects of CEE on E8 cones. Several recombinant trophic factors were also tested, but none could mimic the stimulatory effects of CEE on channel expression. In summary, the developmental expression of cGMP-gated cationic channels in embryonic cones appears to be regulated by epigenetic factors. The ability of cones to respond to these epigenetic factors is also developmentally regulated.


Assuntos
GMP Cíclico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Canais Iônicos/biossíntese , Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Extratos Celulares/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Cromonas/farmacologia , GMP Cíclico/antagonistas & inibidores , GMP Cíclico/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/genética , Sistema de Sinalização das MAP Quinases , Morfolinas/farmacologia , Técnicas de Patch-Clamp , Inibidores de Fosfoinositídeo-3 Quinase , RNA Mensageiro/biossíntese , Retina/efeitos dos fármacos , Retina/embriologia , Células Fotorreceptoras Retinianas Cones/efeitos dos fármacos
8.
J Neurophysiol ; 84(6): 2767-76, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11110807

RESUMO

The intrinsic electrical properties of identified choroid and ciliary neurons of the chick ciliary ganglion were examined by patch-clamp recording methods. These neurons are derived from a common pool of mesencephalic neural crest precursor cells but innervate different target tissues and have markedly different action potential waveforms and intrinsic patterns of repetitive spike discharge. Therefore it is important to determine whether these cell types express different types of plasma membrane ionic channels, and to ascertain the developmental stages at which these cell types begin to diverge. This study has focused on large-conductance Ca(2+)-activated K(+) channels (K(Ca)), which are known to regulate spike waveform and repetitive firing in many cell types. Both ciliary ganglion cell types, identified on the basis of size and somatostatin immunoreactivity, express a robust macroscopic K(Ca) carried by a kinetically homogeneous population of large-conductance (BK-type) K(Ca) channels. However, the kinetic properties of these channels are different in the two cell types. Steady-state fluctuation analyses of macroscopic K(Ca) produced power spectra that could be fitted with a single Lorentzian curve in both cell types. However, the resulting corner frequency was significantly lower in choroid neurons than in ciliary neurons, suggesting that the underlying K(Ca) channels have a longer mean open-time in choroid neurons. Consistent with fluctuation analyses, significantly slower gating of K(Ca) channels in choroid neurons was also observed during macroscopic activation and deactivation at membrane potentials positive to -30 mV. Differences in the kinetic properties of K(Ca) channels could also be observed directly in single-channel recordings from identified embryonic day 13 choroid and ciliary neurons. The mean open-time of large-conductance K(Ca) channels was significantly greater in choroid neurons than in ciliary neurons in excised inside-out patches. The developmental expression of functional K(Ca) channels appears to be regulated differently in the two cell types. Although both cell types acquire functional K(Ca) at the same developmental stages (embryonic days 9-13), functional expression of these channels in ciliary neurons requires target-derived trophic factors. In contrast, expression of functional K(Ca) channels proceeds normally in choroid neurons developing in vitro in the absence of target-derived trophic factors. Consistent with this, extracts of ciliary neuron target tissues (striated muscle of the iris/ciliary body) contain K(Ca) stimulatory activity. However, K(Ca) stimulatory activity cannot be detected in extracts of the smooth muscle targets of choroid neurons.


Assuntos
Gânglios Parassimpáticos/metabolismo , Neurônios/metabolismo , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Animais , Cálcio/metabolismo , Cálcio/farmacologia , Tamanho Celular , Células Cultivadas , Embrião de Galinha , Corioide/química , Corioide/embriologia , Corioide/inervação , Corpo Ciliar/química , Corpo Ciliar/embriologia , Corpo Ciliar/inervação , Relação Dose-Resposta a Droga , Gânglios Parassimpáticos/citologia , Gânglios Parassimpáticos/embriologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/classificação , Neurônios/citologia , Técnicas de Patch-Clamp , Potássio/metabolismo , Potássio/farmacocinética , Extratos de Tecidos/farmacologia
9.
J Neurosci ; 20(15): 5616-22, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10908598

RESUMO

An avian ortholog of transforming growth factor beta1 (TGFbeta1) is the target-derived factor responsible for the developmental expression of large-conductance Ca(2+)-activated K(+) (K(Ca)) channels in chick ciliary ganglion (CG) neurons developing in vivo and in vitro. Application of TGFbeta1 evokes an acute stimulation of K(Ca) that can be observed immediately after cessation of a 12 hr exposure to this factor, that persists in the presence of protein synthesis inhibitors, and that is therefore mediated by posttranslational events. Here we show that a single 3 hr exposure to TGFbeta1 can also induce long-lasting stimulation of macroscopic K(Ca) that persists for at least 3.5 d after the end of the treatment. In contrast to the acute stimulation, this sustained effect is dependent on the transcription and synthesis of new proteins at approximately the time of TGFbeta1 treatment. However TGFbeta1 does not cause increases in the levels of slowpoke alpha subunit transcripts in CG neurons, suggesting that induction of some other protein or proteins is required for sustained enhancement of macroscopic K(Ca). In addition, application of TGFbeta1 evoked an almost immediate but transient phosphorylation of the mitogen-activated protein kinase Erk in CG neurons. TGFbeta1-evoked Erk activation was blocked by the specific MEK1 inhibitor 2- (2'-amino-3'-methoxyphenyl)-oxanaphthalen-4-one (PD98059). Moreover, application of PD98059 blocked both acute and sustained K(Ca) stimulation evoked by TGFbeta1. These results indicate that TGFbeta1 elicits a biphasic stimulation of K(Ca) via activation of an MEK1-Erk pathway and raise the possibility that other neuronal effects of TGFbeta superfamily members entail Erk activation.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/enzimologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/genética , Processamento Pós-Transcricional do RNA/fisiologia , Transcrição Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Células Cultivadas , Embrião de Galinha , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Gânglios Parassimpáticos/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Neurônios/química , Neurônios/citologia , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo
10.
J Neurochem ; 74(3): 1026-33, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10693933

RESUMO

Application of arachidonic acid evoked robust activation of large-conductance K+ channels in cell-attached and excised inside-out patches from acutely isolated chick ciliary ganglion neurons. A similar effect was produced by 5,8,11,14-eicosatetraynoic acid, a nonmetabolizable analogue of arachidonic acid. The unitary conductance of fatty acid-activated channels was 35-40 pS at +20 mV with physiological gradients of K+ and 165 pS at +20 mV with an extracellular K+ concentration of 37.5 mM and an intracellular K+ concentration of 150 mM. Gating of these channels in cell-attached patches was potentiated by membrane stretch. Channel gating evoked by both lipids was concentration-dependent, with detectable activation apparent at 4 microM in the majority of patches and maximal activation occurring between 32 and 64 microM. Gating was relatively voltage-independent. Large-conductance K+ channels were also activated in inside-out patches by the monounsaturated fatty acid 11-cis-eicosenoic acid but not by the fully saturated fatty acid arachidic acid. Application of 100 microM H2O2, an agent that activates cytosolic phospholipase A2, also caused activation of large-conductance K+ channels in intact neurons. The stimulatory effects of H2O2 were blocked by pretreatment with 20 microM 4-bromophenacyl bromide, an irreversible inhibitor of phospholipase A2. Therefore, mobilization of endogenous fatty acids can cause activation of large-conductance K+ channels in autonomic neurons.


Assuntos
Ácidos Graxos/fisiologia , Gânglios Parassimpáticos/metabolismo , Neurônios/metabolismo , Canais de Potássio/metabolismo , Ácido 5,8,11,14-Eicosatetrainoico/farmacologia , Animais , Ácido Araquidônico/farmacologia , Embrião de Galinha , Ativação Enzimática/fisiologia , Ácidos Graxos não Esterificados/fisiologia , Gânglios Parassimpáticos/citologia , Gânglios Parassimpáticos/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Ativação do Canal Iônico , Neurônios/efeitos dos fármacos , Fosfolipases A/metabolismo , Fosfolipases A2 , Canais de Potássio/efeitos dos fármacos
11.
J Neurophysiol ; 82(3): 1627-31, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10482777

RESUMO

The developmental expression of Ca2+-activated K+ channels (KCa) in chick ciliary ganglion (CG) neurons is regulated by a target-derived avian isoform of TGFbeta1, which evokes a robust increase in the number of functional large-conductance (BK) KCa channels but which produces no change in their kinetics. However, CG neurons express multiple KCa channel subtypes. Here we show that TGFbeta1 regulates the gating properties of intermediate-conductance (IK) KCa channels in developing CG neurons. IK channels in inside-out patches excised from control E9 CG neurons became active on exposure to 1-5 microM free Ca2+ but then remained active on return to Ca2+-free salines. In contrast, IK channels in TGFbeta1-treated cells became active on exposure to 1-5 microM Ca2+, but became quiescent immediately on return to Ca2+-free salines. In contrast to its effects on BK channels, TGFbeta1 had no effect on the mean number of IK channels detected in excised patches. IK channels were not activated in cell-attached patches on E9 neurons depolarized by bath application of 145 mM KCl in the presence of 5 mM external Ca2+. However, BK channels were activated immediately by this procedure and were detected at a higher density in TGFbeta1-treated cells. In addition, analyses of macroscopic KCa fluctuations, and the voltage-dependence of KCa tail currents, suggest that IK channels do not contribute to voltage-evoked whole cell KCa. IK channels therefore may have some other function. These results indicate that the effects of TGFbeta1 on CG neurons entail distinct actions on multiple KCa channel subtypes.


Assuntos
Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , Neurônios/fisiologia , Sistema Nervoso Parassimpático/embriologia , Canais de Potássio/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Linhagem Celular , Embrião de Galinha/citologia , Embrião de Galinha/fisiologia , Condutividade Elétrica , Humanos , Proteínas Recombinantes
12.
Development ; 126(18): 4157-64, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10457024

RESUMO

The developmental expression of macroscopic Ca(2+)-activated K(+) currents in chick ciliary ganglion neurons is dependent on an avian ortholog of TGFbeta1, known as TGFbeta4, secreted from target tissues in the eye. Here we report that a different isoform, TGFbeta3, is also expressed in a target tissue of ciliary ganglion neurons. Application of TGFbeta3 inhibits the functional expression of whole-cell Ca(2+)-activated K(+) currents evoked by 12 hour treatment with either TGFbeta1 or beta-neuregulin-1 in ciliary ganglion neurons developing in vitro. TGFbeta3 had no effect on voltage-activated Ca(2+) currents. A neutralizing antiserum specific for TGFbeta3 potentiates stimulation of Ca(2+)-activated K(+) currents evoked by a target tissue (iris) extract in cultured ciliary ganglion neurons, indicating that TGFbeta3 is an inhibitory component of these extracts. Intraocular injection of TGFbeta3 causes a modest but significant inhibition of the expression of Ca(2+)-activated K(+) currents in ciliary ganglion neurons developing in vivo. Further, intraocular injection of a TGFbeta3-neutralizing antiserum stimulates expression of Ca(2+)-activated K(+) currents in ciliary ganglion neurons developing in vivo, indicating that endogenous TGFbeta3 regulates the functional expression of this current. The normal developmental expression of functional Ca(2+)-activated K(+) currents in ciliary ganglion neurons developing in vivo is therefore regulated by two different target-derived isoforms of TGFbeta, which produce opposing effects on the electrophysiological differentiation of these neurons.


Assuntos
Iris/embriologia , Canais de Potássio/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Cálcio/metabolismo , Embrião de Galinha , Técnicas de Cultura , Eletrofisiologia , Gânglios/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/metabolismo , Soros Imunes , Iris/metabolismo , Neurregulinas , Neurônios/metabolismo , Isoformas de Proteínas , Extratos de Tecidos , Transcrição Gênica , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia
13.
Nature ; 400(6742): 375-8, 1999 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-10432117

RESUMO

The core mechanism of circadian timekeeping in arthropods and vertebrates consists of feedback loops involving several clock genes, including period (per) and timeless (tim). In the fruitfly Drosophila, circadian oscillations in per expression occur in chemosensory cells of the antennae, even when the antennae are excised and maintained in isolated organ culture. Here we demonstrate a robust circadian rhythm in Drosophila in electrophysiological responses to two classes of olfactory stimuli. These rhythms are observed in wild-type flies during light-dark cycles and in constant darkness, but are abolished in per or tim null-mutant flies (per01 and tim01) which lack rhythms in adult emergence and locomotor behaviour. Olfactory rhythms are also abolished in the per 7.2:2 transgenic line in which per expression is restricted to the lateral neurons of the optic lobe. Because per 7.2:2 flies do not express per in peripheral oscillators, our results provide evidence that peripheral circadian oscillators are necessary for circadian rhythms in olfactory responses. As olfaction is essential for food acquisition, social interactions and predator avoidance in many animals, circadian regulation of olfactory systems could have profound effects on the behaviour of organisms that rely on this sensory modality.


Assuntos
Ritmo Circadiano/fisiologia , Drosophila melanogaster/fisiologia , Olfato/fisiologia , Animais , Animais Geneticamente Modificados , Eletrofisiologia , Feminino , Odorantes
14.
Neuron ; 21(5): 1045-53, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9856460

RESUMO

The functional expression of Ca2+-activated K+ channels (KCa) in developing chick ciliary ganglion (CG) neurons requires interactions with target tissues and preganglionic innervation. Here, we show that the stimulatory effects of target tissues are mediated by an isoform of TGFbeta. Exposure of cultured CG neurons to TGFbeta1, but not TGFbeta2 or TGFbeta3, caused robust stimulation of KCa. The KCa stimulatory effects of target tissue extracts were blocked by a neutralizing pan-TGFbeta antiserum but not by specific TGFbeta2 or TGFbeta3 antisera. Intraocular injection of TGFbeta1 caused robust stimulation of KCa, whereas intraocular injection of pan-TGFbeta antiserum inhibited expression of KCa in CG neurons developing in vivo. The effects of TGFbeta1 were potentiated by beta-neuregulin-1, a differentiation factor expressed in preganglionic neurons.


Assuntos
Neurônios/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Cálcio/fisiologia , Células Cultivadas , Embrião de Galinha , Sinergismo Farmacológico , Olho/efeitos dos fármacos , Olho/embriologia , Gânglios Parassimpáticos/efeitos dos fármacos , Gânglios Parassimpáticos/embriologia , Gânglios Parassimpáticos/metabolismo , Glicoproteínas/farmacologia , Soros Imunes/farmacologia , Neurregulinas , Neurônios/citologia , Neurônios/efeitos dos fármacos , Canais de Potássio/genética , Fator de Crescimento Transformador beta/imunologia
15.
J Neurobiol ; 37(1): 23-36, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9777730

RESUMO

The functional expression of the Ca2+-activated K+ current (IK[Ca]) is dependent on cell-cell interactions in developing chick autonomic neurons. In chick ciliary ganglion (CG) neurons, expression of macroscopic IK[Ca] coincides with the formation of synapses with target tissues. CG neurons that develop in vivo in the absence of normal target tissues fail to express functional IK[Ca], although voltage-activated Ca2+ currents and most other ionic currents are expressed at normal amplitudes and densities. CG neurons placed in cell culture prior to formation of synapses with target tissues also fail to express macroscopic IK[Ca]. However, CG neurons cultured in the presence of a heat- and trypsin-sensitive extract of target tissues express IK[Ca] at normal levels. Similarly, interactions with target tissue appear to regulate the expression of whole-cell IK[Ca] in developing chick sympathetic ganglion neurons, although the relevant trophic factors appear to be different from those required by CG neurons. In addition to target tissue interactions, an intact preganglionic innervation is required for the normal in vivo development of IK[Ca] in chick CG neurons. The trophic effects of the afferent innervation do not require synaptic activation of the CG neurons, indicating secretion of a trophic factor, possibly an isoform of beta-neuregulin. The results are consistent with the hypothesis that target- and nerve terminal-derived trophic factors interact at a posttranslational level in the regulation of a functional IK[Ca]. Together, this body of data demonstrates an essential role for cell-cell interactions in the differentiation of neuronal excitability.


Assuntos
Cálcio/metabolismo , Comunicação Celular , Gânglios Autônomos/embriologia , Neurônios/citologia , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular , Embrião de Galinha , Gânglios Autônomos/citologia , Glicoproteínas/metabolismo , Dados de Sequência Molecular , Neurregulinas , Neurônios/metabolismo , Sinapses/fisiologia
16.
Brain Res ; 789(1): 162-6, 1998 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-9602108

RESUMO

A-currents (IA) of chick ciliary ganglion (CG) neurons were blocked reversibly by arachidonic acid and a non-metabolizable analog of arachidonic acid, 5,8,11,14-eicosatetraynoic acid. Inhibition of IA by both lipids was observed in whole-cell recordings and in excised inside-out patches, suggesting that Kv4 (Shal) subunits contribute to functional IA channels in CG neurons. Consistent with this, Kv4.2 and Kv4.3 cDNAs were isolated by RT-PCR from chick CG neurons.


Assuntos
Ácido Araquidônico/farmacologia , Gânglios Parassimpáticos/fisiologia , Neurônios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , RNA Mensageiro/metabolismo , Animais , Embrião de Galinha , Condutividade Elétrica , Gânglios Parassimpáticos/citologia , Gânglios Parassimpáticos/embriologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Canais de Potássio Shal
17.
Endocrinology ; 138(8): 3141-53, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9231761

RESUMO

Endothelin-1 (ET-1) inhibits PRL secretion from cultured rat lactotrophs. However, ET-1 stimulates PRL secretion after cultured lactotrophs have been exposed for 48 h to dopamine or D2 dopamine agonists. In the present study, we have used cell-attached and inside-out patch recordings to establish an ionic basis for these effects. Bath application of 20 nM ET-1 to untreated lactotrophs evoked a robust and persistent activation of large-conductance K+ channels in cell-attached patches. This effect of ET-1 had a long latency to onset, was maintained for as long as ET-1 was present, and required at least 10 min of washing in control saline before complete recovery was achieved. The stimulatory effect of 20 nM ET-1 on these channels was markedly attenuated in the presence of the selective ET(A) receptor antagonist BQ-610 (200 nM), or after pertussis toxin (200 ng/ml, 16 h) pretreatment. The unitary slope conductance of the ET-1 activated channels in cell attached patches was 165 and 95 pS when the recording electrodes contained 150 and 5.4 mM KCl, respectively. These channels were voltage-sensitive and their activity increased upon patch depolarization. Previously activated channels in cell-attached patches became quiescent immediately upon patch excision into Ca2+-free bath saline. Exposure of the intracellular surface to 0.1 microM Ca2+ restored the activity of these channels similar to the level seen before patch excision. In addition, preincubating the cells with the membrane-permeable Ca2+-chelator BAPTA-AM, or using Ca2+-free solution in the recording pipettes, prevented the activation of these channels by ET-1. The ET-1 activated large-conductance Ca2+-dependent K+ (BK(Ca)) channels were blocked by 20 mM tetraethylammonium but were insensitive to the K+ channel blockers apamin (1 microM), charybdotoxin (200 nM), or iberiotoxin (200 nM). Acute application of 10 microM dopamine and 20 nM ET-1 caused activation of BK(Ca) channels with indistinguishable kinetic properties, although the effect of dopamine occurred with shorter latency. After 48-h exposure to the specific D2 dopamine receptor agonist (+/-)-2-(N-phenyl-N-propyl) amino-5-hydroxytetralin hydrochloride (PPHT, 500 nM), bath application of 20 nM ET-1 resulted in inhibition of spontaneously active BK(Ca) channels. These data suggest that both the stimulatory and inhibitory effects of ET-1 on PRL secretion are mediated, at least in part, by actions on BK(Ca) channels, and that long term exposure to dopamine or D2 agonists alters the signaling pathways from the ET(A) receptor to BK(Ca) channels.


Assuntos
Agonistas de Dopamina/farmacologia , Endotelinas/farmacologia , Fenetilaminas/farmacologia , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Canais de Potássio/efeitos dos fármacos , Animais , Apamina/farmacologia , Cálcio/fisiologia , Células Cultivadas , Charibdotoxina/farmacologia , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Estimulação Elétrica , Antagonistas dos Receptores de Endotelina , Feminino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Toxina Pertussis , Adeno-Hipófise/química , Canais de Potássio/análise , Canais de Potássio/fisiologia , Prolactina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Endotelina/análise , Receptores de Endotelina/fisiologia , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia , Fatores de Tempo , Fatores de Virulência de Bordetella/farmacologia
18.
Biol Signals ; 6(4-6): 212-6, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9500658

RESUMO

Free-running circadian rhythms in melatonin secretion persist in dissociated chick pineal cells. Calcium and cyclic AMP interact at several levels in the regulation of melatonin biosynthesis and secretion. Extracellular Ca2+ is required for optimal stimulation of melatonin secretion by cAMP analogues and protagonists. Increased Ca2+ influx during the circadian night is thought to play a role in the circadian clock regulation of melatonin secretion. We have recently described a nonselective cationic channel, ILOT, in chick pineal cells that is regulated by the intrinsic circadian oscillator. Active ILOT channels are detected only during the nighttime and may explain the nocturnal increase in Ca2+ influx. The mechanism by which the activity of ILOT is regulated by the circadian oscillator is not known. In the present study, the effect of the translational inhibitor anisomycin (10(-6) M) on the nighttime activity of ILOT channels was examined. The results show that protein synthesis is required for the detection of ILOT channel activity during the nighttime in cells maintained on light-dark cycles or constant dark conditions.


Assuntos
Ritmo Circadiano/fisiologia , Canais Iônicos/metabolismo , Glândula Pineal/citologia , Glândula Pineal/metabolismo , Biossíntese de Proteínas , Animais , Anisomicina/farmacologia , Células Cultivadas , Galinhas , Ritmo Circadiano/efeitos dos fármacos , Escuridão , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Luz , Técnicas de Patch-Clamp , Inibidores da Síntese de Proteínas/farmacologia , Proteínas/fisiologia
19.
Proc Natl Acad Sci U S A ; 94(11): 5934-8, 1997 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-9159178

RESUMO

The developmental expression of macroscopic Ca2+-activated K+ currents (IK[Ca]) in chicken ciliary ganglion (CG) neurons is dependent in part on trophic factors released from preganglionic nerve terminals. Neuregulins are expressed in the preganglionic neurons that innervate the chicken CG and are therefore plausible candidates for this activity. Application of 1 nM beta1-neuregulin peptide for 12 hr evokes a large (7- to 10-fold) increase in IK[Ca] in embryonic day 9 CG neurons, even in the presence of a translational inhibitor. A similar posttranslational effect is produced by high concentrations (10 nM) of epidermal growth factor and type alpha transforming growth factor but not by 10 nM alpha2-neuregulin peptide or by neurotrophins at 40 ng.ml-1. beta1-neuregulin treatment for 12 hr also confers Ca2+ sensitivity onto large-conductance (285 pS) K+ channels observed in inside-out patches. beta-Neuregulins have no effect on voltage-activated Ca2+ currents of CG neurons. These data support the hypothesis that beta-neuregulins mediate the trophic effects of preganglionic nerve terminals on the electrophysiological differentiation of developing CG neurons.


Assuntos
Gânglios Parassimpáticos/fisiologia , Glicoproteínas/farmacologia , Fatores de Crescimento Neural/farmacologia , Neurônios/fisiologia , Canais de Potássio/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Animais , Células Cultivadas , Embrião de Galinha , Receptores ErbB/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Terminações Nervosas/fisiologia , Neurregulinas , Neurônios/efeitos dos fármacos
20.
Science ; 276(5312): 599-603, 1997 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-9110980

RESUMO

ARIA (for acetylcholine receptor-inducing activity), a protein purified on the basis of its ability to stimulate acetylcholine receptor (AChR) synthesis in cultured myotubes, is a member of the neuregulin family and is present at motor endplates. This suggests an important role for neuregulins in mediating the nerve-dependent accumulation of AChRs in the postsynaptic membrane. Nerve-muscle synapses have now been analyzed in neuregulin-deficient animals. Mice that are heterozygous for the deletion of neuregulin isoforms containing an immunoglobulin-like domain are myasthenic. Postsynaptic AChR density is significantly reduced, as judged by the decrease in the mean amplitude of spontaneous miniature endplate potentials and bungarotoxin binding. On the other hand, the mean amplitude of evoked endplate potentials was not decreased, due to an increase in the number of quanta released per impulse, a compensation that has been observed in other myasthenic states. Thus, the density of AChRs in the postsynaptic membrane depends on immunoglobulin-containing neuregulin isoforms throughout the life of the animal.


Assuntos
Glicoproteínas/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Junção Neuromuscular/metabolismo , Receptores Colinérgicos/metabolismo , Acetilcolina/metabolismo , Animais , Bungarotoxinas/metabolismo , Glicoproteínas/genética , Heterozigoto , Imunoglobulinas/análise , Técnicas In Vitro , Potenciais da Membrana , Camundongos , Placa Motora/metabolismo , Placa Motora/fisiologia , Debilidade Muscular/etiologia , Proteínas do Tecido Nervoso/genética , Neuregulina-1 , Neurregulinas , Receptores Colinérgicos/genética , Transmissão Sináptica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...