Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 287(52): 43853-61, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23118233

RESUMO

The L-type Ca(2+) channel (dihydropyridine receptor (DHPR) in skeletal muscle acts as the voltage sensor for excitation-contraction coupling. To better resolve the spatial organization of the DHPR subunits (α(1s) or Ca(V)1.1, α(2), ß(1a), δ1, and γ), we created transgenic mice expressing a recombinant ß(1a) subunit with YFP and a biotin acceptor domain attached to its N- and C- termini, respectively. DHPR complexes were purified from skeletal muscle, negatively stained, imaged by electron microscopy, and subjected to single-particle image analysis. The resulting 19.1-Å resolution, three-dimensional reconstruction shows a main body of 17 × 11 × 8 nm with five corners along its perimeter. Two protrusions emerge from either face of the main body: the larger one attributed to the α(2)-δ1 subunit that forms a flexible hook-shaped feature and a smaller protrusion on the opposite side that corresponds to the II-III loop of Ca(V)1.1 as revealed by antibody labeling. Novel features discernible in the electron density accommodate the atomic coordinates of a voltage-gated sodium channel and of the ß subunit in a single docking possibility that defines the α1-ß interaction. The ß subunit appears more closely associated to the membrane than expected, which may better account for both its role in localizing the α(1s) subunit to the membrane and its suggested role in excitation-contraction coupling.


Assuntos
Canais de Cálcio Tipo L/ultraestrutura , Simulação de Acoplamento Molecular , Proteínas Musculares/ultraestrutura , Músculo Esquelético/ultraestrutura , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Subunidades Proteicas
2.
PLoS One ; 7(9): e45581, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029114

RESUMO

Neurodegeneration is a major cause of human disease. Within the cerebellum, neuronal degeneration and/or dysfunction has been associated with many diseases, including several forms of cerebellar ataxia, since normal cerebellar function is paramount for proper motor coordination, balance, and motor learning. The cerebellum represents a well-established neural circuit. Determining the effects of neuronal loss is of great importance for understanding the fundamental workings of the cerebellum and disease-associated dysfunctions. This paper presents computational modeling of cerebellar function in relation to neurodegeneration either affecting a specific cerebellar cell type, such as granule cells or Purkinje cells, or more generally affecting cerebellar cells and the implications on effects in relation to performance degradation throughout the progression of cell death. The results of the models show that the overall number of cells, as a percentage of the total cell number in the model, of a particular type and, primarily, their proximity to the circuit output, and not the neuronal convergence due to the relative number of cells of a particular type, is the main indicator of the gravity of the functional deficit caused by the degradation of that cell type. Specifically, the greater the percentage loss of neurons of a specific type and the closer proximity of those cells to the deep cerebellar neurons, the greater the deficit caused by the neuronal cell loss. These findings contribute to the understanding of the functional consequences of neurodegeneration and the functional importance of specific connectivity within a neuronal circuit.


Assuntos
Cerebelo/fisiologia , Modelos Neurológicos , Cerebelo/citologia , Cerebelo/fisiopatologia , Simulação por Computador , Humanos , Neurônios/fisiologia
3.
Channels (Austin) ; 6(1): 26-40, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22522946

RESUMO

In skeletal muscle, L-type calcium channels (DHPRs), localized to plasma membrane sarcoplasmic reticulum junctions, are tightly packed into groups of four termed tetrads. Here, we have used bimolecular fluorescence complementation (BiFC) and targeted biotinylation to probe the structure and organization of ß1a subunits associated with native CaV 1.1 in DHPRs of myotubes. The construct YN-ß1a-YC, in which the non-fluorescent fragments of YFP ("YN" corresponding to YFP residues 1-158, and "YC" corresponding to YFP residues 159-238) were fused, respectively, to the N- and C-termini of ß1a, was fully functional and displayed yellow fluorescence within DHPR tetrads after expression in ß1-knockout (ß1KO) myotubes; this yellow fluorescence demonstrated the occurrence of BiFC of YN and YC on the ß1a N- and C-termini. In these experiments, we avoided overexpression because control experiments in non-muscle cells indicated that this could result in non-specific BiFC. BiFC of YN-ß1a-YC in DHPR tetrads appeared to be intramolecular between N- and C-termini of individual ß1a subunits rather than between adjacent DHPRs because BiFC (1) was observed for YN-ß1a-YC co-expressed with CaV 1.2 (which does not form tetrads) and (2) was not observed after co-expression of YN-ß1a-YN plus YC-ß1a-YC in ß1KO myotubes. Thus, ß1a function is compatible with N- and C-termini being close enough together to allow BiFC. However, both termini appeared to have positional freedom and not to be closely opposed by other junctional proteins since both were accessible to gold-streptavidin conjugates. Based on these results, a model is proposed for the arrangement of ß1a subunits in DHPR tetrads.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Músculo Esquelético/fisiologia , Animais , Proteínas de Bactérias/genética , Biotina/genética , Biotinilação , Linhagem Celular , Fluorescência , Humanos , Proteínas Luminescentes/genética , Fibras Musculares Esqueléticas/fisiologia , Subunidades Proteicas/fisiologia , Transfecção
4.
Channels (Austin) ; 2(3): 163-79, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18836296

RESUMO

Calcium ions play an important role in the electrical excitability of nerve and muscle, as well as serving as a critical second messenger for diverse cellular functions. As a result, mutations of genes encoding calcium channels may have subtle affects on channel function yet strongly perturb cellular behavior. This review discusses the effects of calcium channel mutations on channel function, the pathological consequences for cellular physiology, and possible links between altered channel function and disease. Many cellular functions are directly or indirectly regulated by the free cytosolic calcium concentration. Thus, calcium levels must be very tightly regulated in time and space. Intracellular calcium ions are essential second messengers and play a role in many functions including, action potential generation, neurotransmitter and hormone release, muscle contraction, neurite outgrowth, synaptogenesis, calcium-dependent gene expression, synaptic plasticity and cell death. Calcium ions that control cell activity can be supplied to the cell cytosol from two major sources: the extracellular space or intracellular stores. Voltage-gated and ligand-gated channels are the primary way in which Ca(2+) ions enter from the extracellular space. The sarcoplasm reticulum (SR) in muscle and the endoplasmic reticulum in non-muscle cells are the main intracellular Ca(2+) stores: the ryanodine receptor (RyR) and inositol-triphosphate receptor channels are the major contributors of calcium release from internal stores.


Assuntos
Canais de Cálcio/genética , Mutação , Animais , Ataxia/genética , Cálcio/química , Canais de Cálcio Tipo L/metabolismo , Citosol/metabolismo , Feminino , Heterozigoto , Humanos , Íons , Masculino , Camundongos , Cegueira Noturna/genética , Retículo Sarcoplasmático/metabolismo
5.
J Gen Physiol ; 130(4): 379-88, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17893191

RESUMO

In skeletal muscle, the dihydropyridine receptor (DHPR) in the plasma membrane (PM) serves as a Ca(2+) channel and as the voltage sensor for excitation-contraction (EC coupling), triggering Ca(2+) release via the type 1 ryanodine receptor (RyR1) in the sarcoplasmic reticulum (SR) membrane. In addition to being functionally linked, these two proteins are also structurally linked to one another, but the identity of these links remains unknown. As an approach to address this issue, we have expressed DHPR alpha(1S) or beta(1a) subunits, with a biotin acceptor domain fused to targeted sites, in myotubes null for the corresponding, endogenous DHPR subunit. After saponin permeabilization, the approximately 60-kD streptavidin molecule had access to the beta(1a) N and C termini and to the alpha(1S) N terminus and proximal II-III loop (residues 671-686). Steptavidin also had access to these sites after injection into living myotubes. However, sites of the alpha(1S) C terminus were either inaccessible or conditionally accessible in saponin- permeabilized myotubes, suggesting that these C-terminal regions may exist in conformations that are occluded by other proteins in PM/SR junction (e.g., RyR1). The binding of injected streptavidin to the beta(1a) N or C terminus, or to the alpha(1S) N terminus, had no effect on electrically evoked contractions. By contrast, binding of streptavidin to the proximal alpha(1S) II-III loop abolished such contractions, without affecting agonist-induced Ca(2+) release via RyR1. Moreover, the block of EC coupling did not appear to result from global distortion of the DHPR and supports the hypothesis that conformational changes of the alpha(1S) II-III loop are necessary for EC coupling in skeletal muscle.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio , Contração Muscular/fisiologia , Estreptavidina/metabolismo , Animais , Sítios de Ligação , Canais de Cálcio Tipo L/química , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Células Cultivadas , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Subunidades Proteicas/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Estreptavidina/química , Relação Estrutura-Atividade
6.
J Biol Chem ; 279(42): 44057-64, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15280388

RESUMO

Excitation-contraction coupling in skeletal muscle involves conformational coupling between dihydropyridine receptors (DHPRs) in the plasma membrane and ryanodine receptors (RyRs) in the sarcoplasmic reticulum. However, it remains uncertain what regions, if any, of the two proteins interact with one another. Toward this end, it would be valuable to know the spatial interrelationships of DHPRs and RyRs within plasma membrane/sarcoplasmic reticulum junctions. Here we describe a new approach based on metabolic incorporation of biotin into targeted sites of the DHPR. To accomplish this, cDNAs were constructed with a biotin acceptor domain (BAD) fused to selected sites of the DHPR, with fluorescent protein (XFP) attached at a second site. All of the BAD-tagged constructs properly targeted to junctions (as indicted by small puncta of XFP) and were functional for excitation-contraction coupling. To determine whether the introduced BAD was biotinylated and accessible to avidin (approximately 60 kDa), myotubes were fixed, permeablized, and exposed to fluorescently labeled avidin. Upon expression in beta1-null or dysgenic (alpha1S-null) myotubes, punctate avidin fluorescence co-localized with the XFP puncta for BAD attached to the beta1a N- or C-terminals, or the alpha1S N-terminal or II-III loop. However, BAD fused to the alpha1S C-terminal was inaccessible to avidin in dysgenic myotubes (containing RyR1). In contrast, this site was accessible to avidin when the identical construct was expressed in dyspedic myotubes lacking RyR1. These results indicate that avidin has access to a number of sites of the DHPR within fully assembled (RyR1-containing) junctions, but not to the alpha1S C-terminal, which appears to be occluded by the presence of RyR1.


Assuntos
Membrana Celular/fisiologia , Músculo Esquelético/fisiologia , Retículo Sarcoplasmático/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/análise , Biotinilação , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/fisiologia , Proteínas Luminescentes/análise , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteínas Recombinantes de Fusão/química , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA