Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Mol Neurobiol ; 43(6): 2769-2783, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36580209

RESUMO

Whole exome sequencing (WES) has been used to detect rare causative variants in neurological diseases. However, the efficacy of WES in genetic diagnosis of clinically heterogeneous familial stroke remains inconclusive. We prospectively searched for disease-causing variants in unrelated probands with defined familial stroke by candidate gene/hotspot screening and/or WES, depending on stroke subtypes and neuroimaging features at a referral center. The clinical significance of each variant was determined according to the American College of Medical Genetics guidelines. Among 161 probands (mean age at onset 53.2 ± 13.7 years; male 63.4%), 33 participants (20.5%) had been identified with 19 pathogenic/likely pathogenic variants (PVs; WES applied 152/161 = 94.4%). Across subtypes, the highest hit rate (HR) was intracerebral hemorrhage (ICH, 7/18 = 38.9%), particularly with the etiological subtype of structural vasculopathy (4/4 = 100%, PVs in ENG, KRIT1, PKD1, RNF213); followed by ischemic small vessel disease (SVD, 15/48 = 31.3%; PVs in NOTCH3, HTRA1, HBB). In contrast, large artery atherosclerosis (LAA, 4/44 = 9.1%) and cardioembolism (0/11 = 0%) had the lowest HR. NOTCH3 was the most common causative gene (16/161 = 9.9%), presenting with multiple subtypes of SVD (n = 13), ICH (n = 2), or LAA (n = 1). Importantly, we disclosed two previously unreported PVs, KRIT1 p.E379* in a familial cerebral cavernous malformation, and F2 p.F382L in a familial cerebral venous sinus thrombosis. The contribution of monogenic etiologies was particularly high in familial ICH and SVD subtypes in our Taiwanese cohort. Utilizing subtype-guided hotspot screening and/or subsequent WES, we unraveled monogenic causes in 20.5% familial stroke probands, including 1.2% novel PVs. Genetic diagnosis may enable early diagnosis, management and lifestyle modification. Among 161 familial stroke probands, 33 (20.5%) had been identified pathogenic or likely pathogenic monogenic variants related to stroke. The positive hit rate among all subtypes was high in intracerebral hemorrhage (ICH) and ischemic small vessel disease (SVD). Notably, two previously unreported variants, KRIT1 p.E379* in a familial cerebral cavernous malformation and F2 p.F382L in familial cerebral venous sinus thrombosis, were disclosed. CVT cerebral venous thrombosis; HTN Hypertensive subtype; LAA large artery atherosclerosis; SV structural vasculopathy; U Undetermined.


Assuntos
Aterosclerose , AVC Isquêmico , Trombose dos Seios Intracranianos , Acidente Vascular Cerebral , Humanos , Masculino , Adulto , Pessoa de Meia-Idade , Idoso , Sequenciamento do Exoma , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/diagnóstico , Hemorragia Cerebral/complicações , Hemorragia Cerebral/diagnóstico por imagem , Hemorragia Cerebral/genética , Aterosclerose/complicações , Isquemia/complicações , Trombose dos Seios Intracranianos/complicações , Adenosina Trifosfatases , Ubiquitina-Proteína Ligases
2.
Int J Mol Sci ; 22(15)2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34361012

RESUMO

Loss-of-function mutations in the KV4.3 channel-encoding KCND3 gene are linked to neurodegenerative cerebellar ataxia. Patients suffering from neurodegeneration associated with iron deposition may also present with cerebellar ataxia. The mechanism underlying brain iron accumulation remains unclear. Here, we aim to ascertain the potential pathogenic role of KCND3 variant in iron accumulation-related cerebellar ataxia. We presented a patient with slowly progressive cerebellar ataxia, parkinsonism, cognitive impairment, and iron accumulation in the basal ganglia and the cerebellum. Whole exome sequencing analyses identified in the patient a heterozygous KCND3 c.1256G>A (p.R419H) variant predicted to be disease-causing by multiple bioinformatic analyses. In vitro biochemical and immunofluorescence examinations revealed that, compared to the human KV4.3 wild-type channel, the p.R419H variant exhibited normal protein abundance and subcellular localization pattern. Electrophysiological investigation, however, demonstrated that the KV4.3 p.R419H variant was associated with a dominant increase in potassium current amplitudes, as well as notable changes in voltage-dependent gating properties leading to enhanced potassium window current. These observations indicate that, in direct contrast with the loss-of-function KCND3 mutations previously reported in cerebellar ataxia patients, we identified a rare gain-of-function KCND3 variant that may expand the clinical and molecular spectra of neurodegenerative cerebellar disorders associated with brain iron accumulation.


Assuntos
Disfunção Cognitiva/genética , Mutação com Ganho de Função , Ferro/metabolismo , Transtornos Parkinsonianos/genética , Canais de Potássio Shal/genética , Ataxias Espinocerebelares/genética , Potenciais de Ação , Idoso , Encéfalo/metabolismo , Disfunção Cognitiva/patologia , Células HEK293 , Humanos , Masculino , Transtornos Parkinsonianos/patologia , Domínios Proteicos , Canais de Potássio Shal/química , Canais de Potássio Shal/metabolismo , Ataxias Espinocerebelares/patologia
3.
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
4.
J Neurosci ; 37(9): 2485-2503, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28167673

RESUMO

Voltage-gated CaV2.1 channels comprise a pore-forming α1A subunit with auxiliary α2δ and ß subunits. CaV2.1 channels play an essential role in regulating synaptic signaling. Mutations in the human gene encoding the CaV2.1 subunit are associated with the cerebellar disease episodic ataxia type 2 (EA2). Several EA2-causing mutants exhibit impaired protein stability and exert dominant-negative suppression of CaV2.1 wild-type (WT) protein expression via aberrant proteasomal degradation. Here, we set out to delineate the protein degradation mechanism of human CaV2.1 subunit by identifying RNF138, an E3 ubiquitin ligase, as a novel CaV2.1-binding partner. In neurons, RNF138 and CaV2.1 coexist in the same protein complex and display notable subcellular colocalization at presynaptic and postsynaptic regions. Overexpression of RNF138 promotes polyubiquitination and accelerates protein turnover of CaV2.1. Disrupting endogenous RNF138 function with a mutant (RNF138-H36E) or shRNA infection significantly upregulates the CaV2.1 protein level and enhances CaV2.1 protein stability. Disrupting endogenous RNF138 function also effectively rescues the defective protein expression of EA2 mutants, as well as fully reversing EA2 mutant-induced excessive proteasomal degradation of CaV2.1 WT subunits. RNF138-H36E coexpression only partially restores the dominant-negative effect of EA2 mutants on CaV2.1 WT functional expression, which can be attributed to defective membrane trafficking of CaV2.1 WT in the presence of EA2 mutants. We propose that RNF138 plays a critical role in the homeostatic regulation of CaV2.1 protein level and functional expression and that RNF138 serves as the primary E3 ubiquitin ligase promoting EA2-associated aberrant degradation of human CaV2.1 subunits.SIGNIFICANCE STATEMENT Loss-of-function mutations in the human CaV2.1 subunit are linked to episodic ataxia type 2 (EA2), a dominantly inherited disease characterized by paroxysmal attacks of ataxia and nystagmus. EA2-causing mutants may exert dominant-negative effects on the CaV2.1 wild-type subunit via aberrant proteasomal degradation. The molecular nature of the CaV2.1 ubiquitin-proteasome degradation pathway is currently unknown. The present study reports the first identification of an E3 ubiquitin ligase for CaV2.1, RNF138. CaV2.1 protein stability is dynamically regulated by RNF138 and auxiliary α2δ and ß subunits. We provide a proof of concept that protecting the human CaV2.1 subunit from excessive proteasomal degradation with specific interruption of endogenous RNF138 function may partially contribute to the future development of a novel therapeutic strategy for EA2 patients.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Ataxia/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Canais de Cálcio Tipo N/genética , Linhagem Celular , Células Cultivadas , Córtex Cerebral/citologia , Cicloeximida/farmacologia , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Mutação/genética , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nistagmo Patológico/genética , Oócitos , Inibidores da Síntese de Proteínas/farmacologia , Proteólise/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/genética , Xenopus
5.
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
6.
PLoS One ; 9(10): e110423, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333352

RESUMO

Eag (Kv10) and Erg (Kv11) belong to two distinct subfamilies of the ether-à-go-go K+ channel family (KCNH). While Erg channels are characterized by an inward-rectifying current-voltage relationship that results from a C-type inactivation, mammalian Eag channels display little or no voltage-dependent inactivation. Although the amino (N)-terminal region such as the eag domain is not required for the C-type inactivation of Erg channels, an N-terminal deletion in mouse Eag1 has been shown to produce a voltage-dependent inactivation. To further discern the role of the eag domain in the inactivation of Eag1 channels, we generated N-terminal chimeras between rat Eag (rEag1) and human Erg (hERG1) channels that involved swapping the eag domain alone or the complete cytoplasmic N-terminal region. Functional analyses indicated that introduction of the homologous hERG1 eag domain led to both a fast phase and a slow phase of channel inactivation in the rEag1 chimeras. By contrast, the inactivation features were retained in the reverse hERG1 chimeras. Furthermore, an eag domain-lacking rEag1 deletion mutant also showed the fast phase of inactivation that was notably attenuated upon co-expression with the rEag1 eag domain fragment, but not with the hERG1 eag domain fragment. Additionally, we have identified a point mutation in the S4-S5 linker region of rEag1 that resulted in a similar inactivation phenotype. Biophysical analyses of these mutant constructs suggested that the inactivation gating of rEag1 was distinctly different from that of hERG1. Overall, our findings are consistent with the notion that the eag domain plays a critical role in regulating the inactivation gating of rEag1. We propose that the eag domain may destabilize or mask an inherent voltage-dependent inactivation of rEag1 K+ channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Ativação do Canal Iônico/fisiologia , Animais , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Feminino , Células HEK293 , Humanos , Camundongos , Oócitos/fisiologia , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , RNA Complementar/metabolismo , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Transfecção , Xenopus/crescimento & desenvolvimento
7.
J Biol Chem ; 289(33): 22815-22834, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25008323

RESUMO

A functional voltage-gated K(+) (Kv) channel comprises four pore-forming α-subunits, and only members of the same Kv channel subfamily may co-assemble to form heterotetramers. The ether-à-go-go family of Kv channels (KCNH) encompasses three distinct subfamilies: Eag (Kv10), Erg (Kv11), and Elk (Kv12). Members of different ether-à-go-go subfamilies, such as Eag and Erg, fail to form heterotetramers. Although a short stretch of amino acid sequences in the distal C-terminal section has been implicated in subfamily-specific subunit assembly, it remains unclear whether this region serves as the sole and/or principal subfamily recognition domain for Eag and Erg. Here we aim to ascertain the structural basis underlying the subfamily specificity of ether-à-go-go channels by generating various chimeric constructs between rat Eag1 and human Erg subunits. Biochemical and electrophysiological characterizations of the subunit interaction properties of a series of different chimeric and truncation constructs over the C terminus suggested that the putative C-terminal recognition domain is dispensable for subfamily-specific assembly. Further chimeric analyses over the N terminus revealed that the N-terminal region may also harbor a subfamily recognition domain. Importantly, exchanging either the N-terminal or the C-terminal domain alone led to a virtual loss of the intersubfamily assembly boundary. By contrast, simultaneously swapping both recognition domains resulted in a reversal of subfamily specificity. Our observations are consistent with the notion that both the N-terminal and the C-terminal recognition domains are required to sustain the subfamily-specific assembly of rat Eag1 and human Erg.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Animais , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Ratos , Xenopus laevis
8.
BMC Neurosci ; 15: 23, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24495567

RESUMO

BACKGROUND: In mammals, Eag K+ channels (KV10) are exclusively expressed in the brain and comprise two isoforms: Eag1 (KV10.1) and Eag2 (KV10.2). Despite their wide presence in various regions of the brain, the functional role of Eag K+ channels remains obscure. Here we address this question by characterizing the subcellular localization of rat Eag1 (rEag1) and rat Eag2 (rEag2) in hippocampal neurons, as well as determining the structural basis underlying their different localization patterns. RESULTS: Immunofluorescence analysis of young and mature hippocampal neurons in culture revealed that endogenous rEag1 and rEag2 K+ channels were present in both the dendrosomatic and the axonal compartments. Only rEag1 channels displayed a punctate immunostaining pattern and showed significant co-localization with PSD-95. Subcellular fractionation analysis further demonstrated a distinct enrichment of rEag1 in the synaptosomal fraction. Over-expression of recombinant GFP-tagged Eag constructs in hippocampal neurons also showed a significant punctate localization of rEag1 channels. To identify the protein region dictating the Eag channel subcellular localization pattern, we generated a variety of different chimeric constructs between rEag1 and rEag2. Quantitative studies of neurons over-expressing these GFP-tagged chimeras indicated that punctate localization was conferred by a segment (A723-R807) within the proximal post-cyclic nucleotide-binding homology domain (post-CNBHD) region in the rEag1 carboxyl terminus. CONCLUSIONS: Our findings suggest that Eag1 and Eag2 K+ channels may modulate membrane excitability in both the dendrosomatic and the axonal compartments and that Eag1 may additionally regulate neurotransmitter release and postsynaptic signaling. Furthermore, we present the first evidence showing that the proximal post-CNBHD region seems to govern the Eag K+ channel subcellular localization pattern.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Hipocampo/química , Neurônios/química , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Frações Subcelulares , Distribuição Tecidual
9.
PLoS One ; 8(11): e79350, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24236122

RESUMO

The astrocytic syncytium plays a critical role in maintaining the homeostasis of the brain through the regulation of gap junction intercellular communication (GJIC). Changes to GJIC in response to inflammatory stimuli in astrocytes may have serious effects on the brain. We have previously shown that lipopolysaccharide (LPS) reduces connexin43 (Cx43) expression and GJIC in cultured rat astrocytes via a toll-like receptor 4-mediated signaling pathway. In the present study, treatment of astrocytes with LPS resulted in a significant increase in levels of the phosphorylated forms of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) -1, -2, and -3 for up to 18 h. An increase in nuclear transcription factor NF-κB levels was also observed after 8 h of LPS treatment and was sustained for up to 18 h. The LPS-induced decrease in Cx43 protein levels and inhibition of GJIC were blocked by the SAPK/JNK inhibitor SP600125, but not by the NF-κB inhibitor BAY11-7082. Following blockade of de novo protein synthesis by cycloheximide, LPS accelerated Cx43 degradation. Moreover, the LPS-induced downregulation of Cx43 was blocked following inhibition of 26S proteasome activity using the reversible proteasome inhibitor MG132 or the irreversible proteasome inhibitor lactacystin. Immunoprecipitation analyses revealed an increased association of Cx43 with both ubiquitin and E3 ubiquitin ligase Nedd4 in astrocytes after LPS stimulation for 6 h and this effect was prevented by SP600125. Taken together, these results suggest that LPS stimulation leads to downregulation of Cx43 expression and GJIC in rat astrocytes by activation of SAPK/JNK and the ubiquitin-proteasome proteolytic pathway.


Assuntos
Astrócitos/metabolismo , Conexina 43/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitinas/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Antracenos/farmacologia , Astrócitos/efeitos dos fármacos , Conexina 43/genética , Regulação para Baixo/efeitos dos fármacos , Junções Comunicantes/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leupeptinas/farmacologia , Lipopolissacarídeos , NF-kappa B/metabolismo , Proteólise , Ratos , Ubiquitinação
10.
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
11.
J Biomed Sci ; 19: 80, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22931352

RESUMO

BACKGROUND: Daidzein, a phytoestrogen found in isoflavone, is known to exert neurotrophic and neuroprotective effects on the nervous system. Using primary rat dorsal root ganglion (DRG) neuronal cultures, we have examined the potential neurite outgrowth effect of daidzein. METHODS: Dissociated dorsal root ganglia (DRG) cultures were used to study the signaling mechanism of daidzein-induced neuritogenesis by immunocytochemistry and Western blotting. RESULTS: In response to daidzein treatment, DRG neurons showed a significant increase in total neurite length and in tip number per neuron. The neuritogenic effect of daidzein was significantly hampered by specific blockers for Src, protein kinase C delta (PKCδ) and mitogen-activated protein kinase/extracellular signal-regulated kinase kinases (MEK/ERK), but not by those for estrogen receptor (ER). Moreover, daidzein induced phosphorylation of Src, PKCδ and ERK. The activation of PKCδ by daidzein was attenuated in the presence of a Src kinase inhibitor, and that of ERK by daidzein was diminished in the presence of either a Src or PKCδ inhibitor. CONCLUSION: Daidzein may stimulate neurite outgrowth of DRG neurons depending on Src kinase, PKCδ and ERK signaling pathway.


Assuntos
Gânglios Espinais , Isoflavonas/farmacologia , Neuritos , Fármacos Neuroprotetores/farmacologia , Animais , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/crescimento & desenvolvimento , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Proteína Oncogênica pp60(v-src)/antagonistas & inibidores , Proteína Oncogênica pp60(v-src)/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais
12.
J Biol Chem ; 286(31): 27183-96, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21646358

RESUMO

The assembly of four pore-forming α-subunits into tetramers is a prerequisite for the formation of functional K(+) channels. A short carboxyl assembly domain (CAD) in the distal end of the cytoplasmic carboxyl terminus has been implicated in the assembly of Eag α-subunits, a subfamily of the ether-à-go-go K(+) channel family. The precise role of CAD in the formation of Eag tetrameric channels, however, remains unclear. Moreover, it has not been determined whether other protein regions also contribute to the assembly of Eag subunits. We addressed these questions by studying the biophysical properties of a series of different rat Eag1 (rEag1) truncation mutants. Two truncation mutants without CAD (K848X and W823X) yielded functional phenotypes similar to those for wild-type (WT) rEag1 channels. Furthermore, nonfunctional rEag1 truncation mutants lacking the distal region of the carboxyl terminus displayed substantial dominant-negative effects on the functional expression of WT as well as K848X and W823X channels. Our co-immunoprecipitation studies further revealed that truncation mutants containing no CAD indeed displayed significant association with rEag1-WT subunits. Finally, surface biotinylation and protein glycosylation analyses demonstrated that progressive truncations of the carboxyl terminus resulted in aggravating disruptions of membrane trafficking and glycosylation of rEag1 proteins. Overall, our data suggest that the distal carboxyl terminus, including CAD, is dispensable for the assembly of rEag1 K(+) channels but may instead be essential for ensuring proper protein biosynthesis. We propose that the S6 segment and the proximal carboxyl terminus may constitute the principal subunit recognition site for the assembly of rEag1 channels.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Animais , Linhagem Celular , DNA Complementar , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Imunofluorescência , Glicosilação , Humanos , Mutação , Técnicas de Patch-Clamp , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Chem Res Toxicol ; 22(11): 1817-26, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19848398

RESUMO

Dehydroeburicoic acid (DeEA) is a triterpene purified from medicinal fungi such as Antrodia camphorate, the crude extract of which is known to exert cytotoxic effects against several types of cancer cells. We aim to test the hypothesis that DeEA possesses significant cytotoxic effects against glioblastomas, one of the most frequent and malignant brain tumors in adults. 3-(4,5-Dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase release assays indicated that DeEA inhibited the proliferation of the human glioblastoma cell U87MG. In addition, Annexin V and propidium iodide staining showed that DeEA treatment led to a rapid increase of glioblastomas in the necrotic/late apoptotic fraction, whereas cell cycle analysis revealed that DeEA failed to significantly enhance the population of U87MG cells in the hypodiploid (sub-G1) fraction. Using electron microscopy, we found that DeEA induced significant cell enlargements, massive cytoplasmic vacuolization, and loss of mitochondrial membrane integrity. DeEA treatment triggered an intracellular Ca(2+) increase, and DeEA-induced cell death was significantly attenuated by BAPTA-AM but not ethylenediaminetetraacetic acid or ethylene glycol tetraacetic acid. DeEA instigated a reduction of both mitochondrial transmembrane potential and intracellular ATP level. Moreover, DeEA induced proteolysis of alpha-spectrin by calpain, and DeEA cytotoxicity in U87MG cells was caspase-independent but was effectively blocked by calpain inhibitor. Interestingly, DeEA also caused autophagic response that was prevented by calpain inhibitor. Taken together, these results suggest that in human glioblastomas, DeEA induces necrotic cell death that involves Ca(2+) overload, mitochondrial dysfunction, and calpain activation.


Assuntos
Antineoplásicos/toxicidade , Cálcio/metabolismo , Calpaína/metabolismo , Glioblastoma/metabolismo , Lanosterol/análogos & derivados , Triterpenos/toxicidade , Apoptose , Caspases/metabolismo , Linhagem Celular Tumoral , Glioblastoma/ultraestrutura , Humanos , Lactato Desidrogenases/metabolismo , Lanosterol/química , Lanosterol/uso terapêutico , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Necrose
14.
Histol Histopathol ; 23(11): 1299-308, 2008 11.
Artigo em Inglês | MEDLINE | ID: mdl-18785111

RESUMO

Beta-lapachone, an o-naphthoquinone, induces various carcinoma cells to undergo apoptosis, but the mechanism is poorly understood. In the present study, we found that the beta-lapachone-induced apoptosis of DU145 human prostate carcinoma cells was associated with endoplasmic reticulum (ER) stress, as shown by increased intracellular calcium levels and induction of GRP-78 and GADD-153 proteins, suggesting that the endoplasmic reticulum is a target of beta-lapachone. Beta-Lapachone-induced DU145 cell apoptosis was dose-dependent and accompanied by cleavage of procaspase-12 and phosphorylation of p38, ERK, and JNK, followed by activation of the executioner caspases, caspase-7 and calpain. However, pretreatment with the general caspase inhibitor, z-VAD-FMK, or calpain inhibitors, including ALLM or ALLN, failed to prevent beta-lapachone-induced apoptotic cell death. Blocking the enzyme activity of NQO1 with dicoumarol, a known NQO1 inhibitor, or preventing an increase in intracellular calcium levels using BAPTA-AM, an intracellular calcium chelator, substantially inhibited MAPK phosphorylation, abolished the activation of calpain, caspase-12 and caspase-7, and provided significant protection of beta-lapachone-treated cells. These findings show that beta-lapachone-induced ER stress and MAP kinase phosphorylation is a novel signaling pathway underlying the molecular mechanism of the anticancer effect of beta-lapachone.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Naftoquinonas/farmacologia , Neoplasias da Próstata/enzimologia , Cálcio/metabolismo , Calpaína/antagonistas & inibidores , Calpaína/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Retículo Endoplasmático/enzimologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Homeostase , Humanos , Masculino , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fosforilação , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico , Fatores de Tempo
15.
Biochem Biophys Res Commun ; 366(2): 393-400, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18062921

RESUMO

We aim to study the mechanisms underlying the neurotrophic effect of daidzein (Dz) in hippocampal neurons. Dz-enhanced axonal outgrowths manifested growth cone formation and increased immunostaining intensity of growth-associated protein 43 (GAP-43) in growth cones. Consistent with this, Dz increased GAP-43 phosphorylation and its membrane translocation without affecting total GAP-43 levels. In the presence of Dz, significant increase in the immunoreactivity for estrogen receptor (ER) beta, but not ERalpha, was observed on the membrane of cell bodies and growing axons. Dz also induced the activation of protein kinase C alpha (PKCalpha), which was inhibited by the ICI182,780 pretreatment. Similarly, Dz-promoted axonal elongation was blocked by ICI182,780 and Gö6976. Moreover, Dz-stimulated activation of GAP-43 was specifically abolished by Gö6976, suggesting PKCalpha being the upstream effector of GAP-43. Taken together, our data suggest that Dz triggers an ERbeta/PKCalpha/GAP-43 signaling cascade to promote axonal outgrowths in cultured hippocampal neurons.


Assuntos
Axônios/fisiologia , Hipocampo/fisiologia , Isoflavonas/administração & dosagem , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Axônios/efeitos dos fármacos , Axônios/ultraestrutura , Crescimento Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Fitoestrógenos/administração & dosagem , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
16.
Neurosci Lett ; 431(1): 12-6, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18063306

RESUMO

Despite of their wide expression in the brain, the precise neurophysiological role of rat Eag1 (rEag1) and Eag2 (rEag2) K(+) channels remains elusive. Our previous studies in hippocampal pyramidal neurons demonstrate a somatodendritic localization of rEag1 and rEag2 channels, suggesting that the two channel isoforms may contribute to setting the membrane excitability of somas and dendrites. Here, we aim to further characterize the cellular and subcellular localization patterns of rEag1 and rEag2 proteins by studying their laminar distribution in the retina. Confocal microscopic analyses of immunofluorescence data revealed that rEag1 and rEag2 K(+) channels exhibit distinct cellular expression pattern in the retina. rEag1 immunoreactivity was most prominent in the outer half of the inner plexiform layer, whereas strong rEag2 immunostain was found in the outer and inner segments of photoreceptor cells, the outer plexiform layer, and the inner nuclear layer. These results suggest that rEag1 and rEag2 K(+) channels may play a significant role in the transmission of electrical signals along the retinal neuronal circuits. We also performed double-labeling experiments to demonstrate that rEag1 and rEag2 are predominantly expressed in the somatodendritic compartment of retinal neurons. In addition, we presented evidence suggesting that rEag1 channels may be expressed in the GABAergic amacrine cell. Finally, based on their different immunostaining patterns over the inner region of the retina, we propose that compared to rEag2, rEag1 expression encompasses a significantly broader range of the somatodendritic compartment of the retinal ganglion cell.


Assuntos
Membrana Celular/metabolismo , Dendritos/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Retina/metabolismo , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Compartimento Celular/fisiologia , Dendritos/ultraestrutura , Imuno-Histoquímica , Potenciais da Membrana/fisiologia , Microscopia Confocal , Células Fotorreceptoras/citologia , Células Fotorreceptoras/metabolismo , Ratos , Ratos Wistar , Retina/citologia , Retina/efeitos dos fármacos , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo
17.
J Cell Biochem ; 103(1): 67-77, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17486586

RESUMO

Our previous study has shown that anti-Thy-1 antibody promotes neurite outgrowth of cultured dorsal root ganglion (DRG) neurons in a protein kinase A (PKA)-dependent manner. The present study provided another intracellular signaling pathway for the neurotrophic effect of anti-Thy-1 antibody. In DMSO-treated control cells, Thy-1 was enriched in microdomain-like structures on cell membranes by immunofluorescence observation. Treatment of DRG neurons with anti-Thy-1 antibody not only stimulated neurite outgrowth, but also increased the branching complexity of the neurites in both small and large neurons. We have previously shown that anti-Thy-1 antibody causes a time-dependent activation of mitogen-activated protein kinase (MEK) and of cyclic AMP response-element binding protein (CREB). Here, anti-Thy-1 antibody elicited a transient activation of c-Src kinase, and the activation of c-Src kinase appeared occurring upstream of the activation of MEK and CREB, since pretreatment with the Src kinase inhibitor, PP2, effectively abolished the anti-Thy-1 antibody-induced neurite outgrowth and the phosphorylation of MEK and CREB. CREB phosphorylation might result in upregulation of certain neurite outgrowth-related proteins. We therefore conclude that anti-Thy-1 antibody activates the c-Src kinase-MEK-CREB cascade and overcomes the inhibitory effect of Thy-1 on neurite outgrowth in DRG neurons.


Assuntos
Anticorpos/imunologia , Gânglios Espinais/enzimologia , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neuritos/enzimologia , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Antígenos Thy-1/imunologia , Animais , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ativação Enzimática , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/imunologia , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Neuritos/efeitos dos fármacos , Neuritos/imunologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas pp60(c-src)/antagonistas & inibidores , Ratos , Ratos Wistar
18.
J Cell Biochem ; 101(3): 566-75, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17177293

RESUMO

Thy-1 is highly expressed in the mammalian nervous system. Our previous study showed that addition of anti-Thy-1 antibody to cultured dorsal root ganglionic (DRG) neurons promotes neurite outgrowth. In this study, we identified a novel signaling pathway mediating this event. Treatment with function-blocking anti-Thy-1 antibodies enhanced neurite outgrowth of DRG neurons in terms of total neurite length, longest neurite length, and total neurite branching points. To elucidate the possible signal transduction pathway involved, activation of kinases was evaluated by Western blotting. Transient phosphorylation of protein kinase A (PKA) and mitogen-activated kinase kinase (MEK) was induced after 15 min of anti-Thy-1 antibody treatment. Pretreatment with a PKA inhibitor (PKI) or an MEK inhibitor, PD98059, significantly decreased the neurite outgrowth response triggered by anti-Thy-1 antibody, indicating the involvement of both kinases. In addition, anti-Thy-1 antibody treatment also induced transient phosphorylation of cyclic AMP-response element-binding protein (CREB) and this effect was also blocked by a PKI or PD98059. Furthermore, the fact that PKI abolished anti-Thy-1 antibody-induced MEK phosphorylation showed that PKA acts upstream of the MEK-CREB cascade. In summary, the PKA-MEK-CREB pathway is a new pathway involved in the neurite outgrowth-promoting effect of anti-Thy-1 antibody.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Gânglios Espinais/efeitos dos fármacos , Isoanticorpos/farmacologia , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Western Blotting , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Flavonoides/farmacologia , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Imuno-Histoquímica , Masculino , Proteínas dos Microfilamentos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
19.
J Biomed Sci ; 12(2): 347-61, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15917998

RESUMO

Tanshinone IIA, a major component extracted from the traditional herbal medicine, Salvia miltiorrhiza Bunge, is known to exhibit potent cytotoxicity against various human carcinoma cells in vitro. However, the mechanism by which tanshinone IIA produces this anti-tumor effect remains unknown. Since anti-neovascularization has generally been regarded as an effective strategy for anti-cancer therapy, we decided to investigate the mechanism underlying tanshinone IIA-mediated death of human endothelial cells. In this study, we demonstrate that tanshinone IIA elicits human endothelial cell death independent of oxidative stress. These events are partially calcium-dependent and actually dependent upon NAD(P)H: quinone oxidoreductase (NQO1) activity. Tanshinone IIA induces an increase in intracellular calcium, which triggers the release of cytochrome c, thus causing loss of the mitochondrial membrane potential (MMP), resulting in the subsequent activation of caspases. Blocking the induction of Ca2+ perturbation with BAPTA-AM partially rescued cells from tanshinone IIA-induced cytotoxicity. Additionally, blocking NQO1 activity with dicoumoral or inhibiting caspase activities with the general caspase inhibitor, z-VAD-fmk, prevented cell death induced by tanshinone IIA. Therefore, our results imply that tanshinone IIA-mediated cytotoxicity against human endothelial cells may occur through activation of NQO1, which induces a calcium imbalance and mitochondrial dysfunction, thus stimulating caspase activity.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Células Endoteliais/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fenantrenos/farmacologia , Salvia miltiorrhiza/metabolismo , Abietanos , Laranja de Acridina/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Antineoplásicos/farmacologia , Apoptose , Western Blotting , Cálcio/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Ciclo Celular , Morte Celular , Citocromos c/metabolismo , Dicumarol/farmacologia , Medicamentos de Ervas Chinesas , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Eletroforese em Gel de Poliacrilamida , Células Endoteliais/patologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Potenciais da Membrana , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Modelos Biológicos , Estresse Oxidativo , Extratos Vegetais , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
20.
Neuroreport ; 16(3): 229-33, 2005 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-15706225

RESUMO

Two isoforms of rat ether-a-go-go (Eag) K channels, rEag1 and rEag2, are widely expressed in many regions of the brain. The neurophysiological roles of these channels, however, are unclear. We addressed this issue by studying their subcellular localizations in hippocampal neurons. Immunofluorescence studies using markers for different compartments of neurons demonstrated a differential expression pattern of rEag1 and rEag2 K channels in the somatodendritic region. Furthermore, rEag1 K channels were in close proximity to synaptophysin and densin-180, but not GAD65. Our data suggest that both rEag1 and rEag2 K channels may play a pivotal role in the regulation of the excitability of dendrites and somas, and that rEag1 K channels may modulate the postsynaptic signaling of glutamatergic synapses.


Assuntos
Hipocampo/citologia , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Potássio/metabolismo , Animais , Western Blotting/métodos , Linhagem Celular , Dendritos/metabolismo , Canais de Potássio Éter-A-Go-Go , Regulação da Expressão Gênica/fisiologia , Glutamato Descarboxilase/metabolismo , Humanos , Imuno-Histoquímica/métodos , Isoenzimas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Canais de Potássio/química , Canais de Potássio/imunologia , Ratos , Sialoglicoproteínas/metabolismo , Frações Subcelulares/metabolismo , Sinapses/metabolismo , Sinaptofisina/metabolismo , Transfecção/métodos , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA