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.
Cell Biosci ; 13(1): 18, 2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36717938

RESUMO

BACKGROUND: Mutations in the human gene encoding the neuron-specific Eag1 (KV10.1; KCNH1) potassium channel are linked to congenital neurodevelopmental diseases. Disease-causing mutant Eag1 channels manifest aberrant gating function and defective protein homeostasis. Both the E3 ubiquitin ligase cullin 7 (Cul7) and the small acid protein 14-3-3 serve as binding partners of Eag1. Cul7 mediates proteasomal and lysosomal degradation of Eag1 protein, whereas over-expression of 14-3-3 notably reduces Eag1 channel activity. It remains unclear whether 14-3-3 may also contribute to Eag1 protein homeostasis. RESULTS: In human cell line and native rat neurons, disruptions of endogenous 14-3-3 function with the peptide inhibitor difopein or specific RNA interference up-regulated Eag1 protein level in a transcription-independent manner. Difopein hindered Eag1 protein ubiquitination at the endoplasmic reticulum and the plasma membrane, effectively promoting the stability of both immature and mature Eag1 proteins. Suppression of endogenous 14-3-3 function also reduced excitotoxicity-associated Eag1 degradation in neurons. Difopein diminished Cul7-mediated Eag1 degradation, and Cul7 knock-down abolished the effect of difopein on Eag1. Inhibition of endogenous 14-3-3 function substantially perturbed the interaction of Eag1 with Cul7. Further structural analyses suggested that the intracellular Per-Arnt-Sim (PAS) domain and cyclic nucleotide-binding homology domain (CNBHD) of Eag1 are essential for the regulatory effect of 14-3-3 proteins. Significantly, suppression of endogenous 14-3-3 function reduced Cul7-mediated degradation of disease-associated Eag1 mutant proteins. CONCLUSION: Overall these results highlight a chaperone-like role of endogenous 14-3-3 proteins in regulating Eag1 protein homeostasis, as well as a therapeutic potential of 14-3-3 modulators in correcting defective protein expression of disease-causing Eag1 mutants.

2.
J Biol Chem ; 296: 100484, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33647316

RESUMO

Mutations in the human gene encoding the neuron-specific Eag1 voltage-gated K+ channel are associated with neurodevelopmental diseases, indicating an important role of Eag1 during brain development. A disease-causing Eag1 mutation is linked to decreased protein stability that involves enhanced protein degradation by the E3 ubiquitin ligase cullin 7 (CUL7). The general mechanisms governing protein homeostasis of plasma membrane- and endoplasmic reticulum (ER)-localized Eag1 K+ channels, however, remain unclear. By using yeast two-hybrid screening, we identified another E3 ubiquitin ligase, makorin ring finger protein 1 (MKRN1), as a novel binding partner primarily interacting with the carboxyl-terminal region of Eag1. MKRN1 mainly interacts with ER-localized immature core-glycosylated, as well as nascent nonglycosylated, Eag1 proteins. MKRN1 promotes polyubiquitination and ER-associated proteasomal degradation of immature Eag1 proteins. Although both CUL7 and MKRN1 contribute to ER quality control of immature core-glycosylated Eag1 proteins, MKRN1, but not CUL7, associates with and promotes degradation of nascent, nonglycosylated Eag1 proteins at the ER. In direct contrast to the role of CUL7 in regulating both ER and peripheral quality controls of Eag1, MKRN1 is exclusively responsible for the early stage of Eag1 maturation at the ER. We further demonstrated that both CUL7 and MKRN1 contribute to protein quality control of additional disease-causing Eag1 mutants associated with defective protein homeostasis. Our data suggest that the presence of this dual ubiquitination system differentially maintains Eag1 protein homeostasis and may ensure efficient removal of disease-associated misfolded Eag1 mutant channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Ribonucleoproteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Células Cultivadas , Retículo Endoplasmático/metabolismo , Proteólise , Proteostase , Ratos , Ratos Sprague-Dawley , Técnicas do Sistema de Duplo-Híbrido
3.
Sci Rep ; 7: 40825, 2017 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-28098200

RESUMO

Mammalian Eag1 (Kv10.1) potassium (K+) channels are widely expressed in the brain. Several mutations in the gene encoding human Eag1 K+ channel have been associated with congenital neurodevelopmental anomalies. Currently very little is known about the molecules mediating protein synthesis and degradation of Eag1 channels. Herein we aim to ascertain the protein degradation mechanism of rat Eag1 (rEag1). We identified cullin 7 (Cul7), a member of the cullin-based E3 ubiquitin ligase family, as a novel rEag1 binding partner. Immunoprecipitation analyses confirmed the interaction between Cul7 and rEag1 in heterologous cells and neuronal tissues. Cul7 and rEag1 also exhibited significant co-localization at synaptic regions in neurons. Over-expression of Cul7 led to reduced protein level, enhanced ubiquitination, accelerated protein turn-over, and decreased current density of rEag1 channels. We provided further biochemical and morphological evidence suggesting that Cul7 targeted endoplasmic reticulum (ER)- and plasma membrane-localized rEag1 to the proteasome and the lysosome, respectively, for protein degradation. Cul7 also contributed to protein degradation of a disease-associated rEag1 mutant. Together, these results indicate that Cul7 mediates both proteasomal and lysosomal degradations of rEag1. Our findings provide a novel insight to the mechanisms underlying ER and peripheral protein quality controls of Eag1 channels.


Assuntos
Proteínas Culina/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Lisossomos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Membrana Celular/metabolismo , Proteínas Culina/genética , Cicloeximida/farmacologia , Retículo Endoplasmático/metabolismo , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Leupeptinas/farmacologia , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Ratos
4.
FEBS Open Bio ; 6(4): 349-57, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27239447

RESUMO

Eag1 is neuron-specific K(+) channel abundantly expressed in the brain and retina. Subcellular localization and physiological analyses in neurons reveal that Eag1 may participate in Ca(2+)-signaling processes in the synapse. Here, we searched for rat Eag1 (rEag1)-binding proteins that may contribute to Ca(2+) regulation of the K(+) channel. Yeast two-hybrid screening identified centrin 4, a member of the centrin family of Ca(2+)-binding proteins. GST pull-down and immunoprecipitation assays in brain and retina lysates confirm the interaction of centrin with rEag1 in neurons. Centrin 4 binds to rEag1 in the absence of Ca(2+). Raising Ca(2+) concentration enhances the association efficiency of centrin 4 and rEag1, and is required for the suppression of rEag1 currents by centrin 4. Altogether, our data suggest that centrin 4 is a novel binding partner that may contribute to Ca(2+) regulation of rEag1 in neurons.

5.
Cell Biochem Biophys ; 67(2): 773-83, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23516094

RESUMO

In the central nervous system, densin-180 (densin) is one of the major components of the post-synaptic density (PSD) of excitatory synapses. Through its intricate interaction with various post-synaptic proteins, this scaffold protein may play a key role in synaptic regulation. Initial structural analyses suggest that densin is a transmembrane protein and may participate in cell-adhesion function between pre- and post-synaptic membranes. Whereas recent biochemical and mass spectrometry studies indicate that densin may instead be a membrane-associated protein with no extracellular domain. To further investigate the structural topology of densin, we began with examining the extracellular accessibility of multiple epitopes in densin. We have provided immunofluorescence evidence showing that none of the tested epitope sites in densin was accessible to extracellularly applied antibodies. In addition, both protease digestion and surface biotinylation data failed to affirm the presence of extracellular domain for densin. However, protein extraction experiments indicated that densin exhibited a significant hydrophobic interaction with the cell membrane that was not expected of cytosolic proteins. Our data therefore do not support the transmembrane model, but rather are consistent with the idea that the topology of densin involves the membrane association configuration.


Assuntos
Sialoglicoproteínas/metabolismo , Animais , Membrana Celular/metabolismo , Espaço Extracelular/metabolismo , Células HEK293 , Humanos , Ratos , Ratos Sprague-Dawley , Sialoglicoproteínas/química
6.
PLoS One ; 7(7): e41203, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22911758

RESUMO

The ether-à-go-go (Eag) potassium (K(+)) channel belongs to the superfamily of voltage-gated K(+) channel. In mammals, the expression of Eag channels is neuron-specific but their neurophysiological role remains obscure. We have applied the yeast two-hybrid screening system to identify rat Eag1 (rEag1)-interacting proteins from a rat brain cDNA library. One of the clones we identified was 14-3-3θ, which belongs to a family of small acidic protein abundantly expressed in the brain. Data from in vitro yeast two-hybrid and GST pull-down assays suggested that the direct association with 14-3-3θ was mediated by both the N- and the C-termini of rEag1. Co-precipitation of the two proteins was confirmed in both heterologous HEK293T cells and native hippocampal neurons. Electrophysiological studies showed that over-expression of 14-3-3θ led to a sizable suppression of rEag1 K(+) currents with no apparent alteration of the steady-state voltage dependence and gating kinetics. Furthermore, co-expression with 14-3-3θ failed to affect the total protein level, membrane trafficking, and single channel conductance of rEag1, implying that 14-3-3θ binding may render a fraction of the channel locked in a non-conducting state. Together these data suggest that 14-3-3θ is a binding partner of rEag1 and may modulate the functional expression of the K(+) channel in neurons.


Assuntos
Proteínas 14-3-3/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas 14-3-3/genética , Animais , Proteínas de Transporte , Linhagem Celular , Canais de Potássio Éter-A-Go-Go/química , Expressão Gênica , Ordem dos Genes , Humanos , Neurônios/metabolismo , Fosforilação , Ligação Proteica , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...