Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Pharm Bull ; 46(3): 379-381, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36858566

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disease. We previously identified Midnolin (MIDN) to be a genetic risk factor for PD in both Yamagata (Japan) and British populations. However, the scale of our previous study was not sufficient to identify MIDN structural variants in the ascertained control of Yamagata Prefecture. We, therefore, reanalyzed MIDN variants in 3021 individuals from Yamagata Prefecture to compare with that in our previous British cohort study. MIDN copy number loss was only found in two cases (0.0662%), which was a lower frequency than that (1.64%) of the previously studied British cohort. Between the Yamagata and British groups, there was significant difference for rs3746106, located in the 5'-UTR of MIDN mRNA (p = 0.0003344, odds ratio 1.143), and for rs3746107, which corresponds to Ala34 (p < 2.2 × 10-16, odds ratio 5.89401). This study indicates that MIDN loss is relatively rare in the general Japanese population. Considering our previous studies that the frequency of MIDN loss is high among patients with PD (10.5 and 6.55% in Yamagata and Britain, respectively), the MIDN variants are much higher genetic risk factors for PD in a Japanese population than in a British population.


Asunto(s)
Proteínas Nucleares , Enfermedad de Parkinson , Humanos , Estudios de Cohortes , Enfermedad de Parkinson/genética , Factores de Riesgo , Proteínas Nucleares/genética , Japón
2.
J Pharmacol Exp Ther ; 381(2): 68-78, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35241633

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disease. Although many monogenic variants have been identified that cause familial PD, most cases are sporadic and the mechanisms of sporadic PD onset remain unclear. We previously identified midnolin (MIDN) as a novel genetic risk factor for PD in a Japanese population. MIDN copy number loss was strongly associated with sporadic PD, which was replicated in a British population. Furthermore, suppression of MIDN expression in rat pheochromocytoma cells inhibits neurite outgrowth and expression of Parkin ubiquitin ligase. However, the detailed molecular mechanisms of MIDN expression are unknown. We, therefore, investigated the molecular mechanism of MIDN expression in human neuroblastoma SH-SY5Y cells. We found that MIDN expression was promoted by insulin via extracellular-signal regulated kinase1/2 and phosphoinositide 3-kinase-dependent pathways. In addition, MIDN promoter activity was enhanced by mutations at transcription factor AP-2 consensus sequences and reduced by mutations at cAMP response element-binding protein and activator protein 1 (AP-1) consensus sequences. The dominant-negative cAMP response element-binding protein mutant did not block MIDN promoter activity, but both the pharmacological inhibitor and decoy oligodeoxynucleotide for AP-1 significantly blocked its activity. Additionally, DNA binding of c-FOS and c-JUN to the AP-1 consensus sequence in the MIDN promoter was enhanced by insulin as determined by chromatin immunoprecipitation, which suggested that AP-1 positively regulated MIDN expression. Taken together, this study reveals molecular mechanisms of MIDN gene expression induced by insulin in neuronal cells, and drugs which promote MIDN expression may have potential to be a novel medicine for PD. SIGNIFICANCE STATEMENT: We demonstrated that insulin promotes midnolin expression via extracellular-signal regulated kinase 1/2 and phosphoinositide 3-kinase pathways. Furthermore, we identified the important region of the MIDN promoter and showed that transcription factors, including activator protein 1, positively regulate MIDN expression, whereas transcription factor AP-2 negatively regulates basal and insulin-induced MIDN expression. We believe that our observations are important and that they contribute to the development of novel drugs to treat Parkinson's disease.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Humanos , Insulina/farmacología , Proteínas Nucleares , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Factores de Riesgo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Transcripción Genética
3.
J Pharmacol Sci ; 148(3): 326-330, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35177212

RESUMEN

Extracellular signal-regulated protein kinase 5 (ERK5) has various physiological functions. However, the physiological role of ERK5 in the treatment of mice with an illicit drug such as methamphetamine (METH) remains unknown. We revealed that mice treated with METH showed hyperactivity, and increased p-ERK5 and Iba1 (a microglia marker) levels in the striatum. Additionally, these changes were inhibited by pretreatment with the ERK5 inhibitor BIX02189. The results suggest that METH-induced hyperactivity is associated with the activation of microglia via p-ERK5 in the striatum. Thus, the ERK5 pathway components in the central nervous system are potential therapeutic targets for preventing METH addiction.


Asunto(s)
Compuestos de Anilina/farmacología , Cuerpo Estriado/citología , Hipercinesia/inducido químicamente , Hipercinesia/tratamiento farmacológico , Indoles/farmacología , Metantelina/efectos adversos , Microglía/efectos de los fármacos , Proteína Quinasa 7 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Compuestos de Anilina/uso terapéutico , Animales , Proteínas de Unión al Calcio/metabolismo , Cuerpo Estriado/metabolismo , Indoles/uso terapéutico , Ratones , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Agitación Psicomotora , Trastornos Relacionados con Sustancias/prevención & control
4.
Int J Mol Sci ; 23(15)2022 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-35955582

RESUMEN

(1) Background: Extracellular signal-regulating kinase 5 (ERK5) has been implicated in many cellular functions, including survival, proliferation, and vascularization. Our objectives were to examine the expression and effect of ERK5 in clear cell renal cell carcinoma (ccRCC). (2) Methods: The expressions of ERK5 and its regulating micro-RNA miR-143 were investigated using immunohistochemistry and quantitative reverse transcriptase PCR in surgical specimens of ccRCC patients. With invitro and in vivo studies, we used pharmacologic ERK5 inhibitor XMD8-92, RNA interference, pre-miR-143 transduction, Western blotting, MTS assay, apoptosis assay, and subcutaneous xenograft model. (3) Results: A strong ERK5 expression in surgical specimen was associated with high-grade (p = 0.01), high-recurrence free rate (p = 0.02), and high cancer-specific survival (p = 0.03). Expression levels of ERK5 and miR-143 expression level were correlated (p = 0.049). Pre-miR-143 transduction into ccRCC cell A498 suppressed ERK5 expression. ERK5 inhibition enhanced cyclin-dependent kinase inhibitor p21 expression and decreased anti-apoptotic molecules BCL2, resulting in decreased cell proliferation and survival both in ccRCC and endothelial cells. In the xenograft model, ERK5 inhibitor XMD8-92 suppressed tumor growth. (4) Conclusions: ERK5 is regulated by miR-143, and ERK5 inhibition is a promising target for ccRCC treatment.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , MicroARNs , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , MicroARNs/metabolismo
5.
J Biol Chem ; 294(44): 16049-16061, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31506297

RESUMEN

Pulmonary veins (PVs) are the major origin of atrial fibrillation. Recently, we recorded hyperpolarization-activated Cl- current (ICl, h) in rat PV cardiomyocytes. Unlike the well-known chloride channel protein 2 (CLCN2) current, the activation curve of ICl, h was hyperpolarized as the Cl- ion concentration ([Cl-] i ) increased. This current could account for spontaneous activity in PV cardiomyocytes linked to atrial fibrillation. In this study, we aimed to identify the channel underlying ICl, h Using RT-PCR amplification specific for Clcn2 or its homologs, a chloride channel was cloned from rat PV and detected in rat PV cardiomyocytes using immunocytochemistry. The gene sequence and electrophysiological functions of the protein were identical to those previously reported for Clcn2, with protein activity observed as a hyperpolarization-activated current by the patch-clamp method. However, the [Cl-] i dependence of activation was entirely different from the observed ICl, h of PV cardiomyocytes; the activation curve of the Clcn2-transfected cells shifted toward positive potential with increased [Cl-] i , whereas the ICl, h of PV and left ventricular cardiomyocytes showed a leftward shift. Therefore, we used MS to explore the possibility of additional proteins interacting with CLCN2 and identified an individual 71-kDa protein, HSPA8, that was strongly expressed in rat PV cardiomyocytes. With co-expression of HSPA8 in HEK293 and PC12 cells, the CLCN2 current showed voltage-dependent activation and shifted to negative potential with increasing [Cl-] i Molecular docking simulations further support an interaction between CLCN2 and HSPA8. These findings suggest that CLCN2 in rat heart contains HSPA8 as a unique accessory protein.


Asunto(s)
Potenciales de Acción , Canales de Cloruro/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Miocitos Cardíacos/metabolismo , Venas Pulmonares/citología , Animales , Sitios de Unión , Canales de Cloruro CLC-2 , Células Cultivadas , Canales de Cloruro/química , Células HEK293 , Proteínas del Choque Térmico HSC70/química , Proteínas del Choque Térmico HSC70/genética , Ventrículos Cardíacos/citología , Humanos , Masculino , Simulación del Acoplamiento Molecular , Miocitos Cardíacos/fisiología , Células PC12 , Unión Proteica , Venas Pulmonares/metabolismo , Ratas , Ratas Wistar
6.
Biol Pharm Bull ; 41(1): 20-23, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29311479

RESUMEN

We recently found that 10.5% of sporadic Parkinson's disease (PD) patients lacked one copy of the midnolin (MIDN) gene. In addition, gene knock-down/out of MIDN caused down-regulation of parkin E3 ubiquitin ligase, indicating MIDN to be a novel PD-risk factor or causative gene. In this study, we performed RNA-sequencing and transcriptome analysis of Midn wild-type and knockout cells. Midn positively or negatively regulated the expression of a wide variety of genes, including causative familial PD genes, such as α-synuclein, parkin, and EIF4G1. However, EIF4G1 protein levels were not altered by the reduction of its mRNA by Midn loss, as seen that parkin protein levels were correlated to the mRNA down-regulation. Taken together, these findings indicate that MIDN regulates the expression of a wide variety of genes, including multiple PD-causative genes and is associated with PD onset.


Asunto(s)
Proteínas Nucleares/genética , Enfermedad de Parkinson/genética , ARN Mensajero/genética , Transcriptoma , Animales , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Nucleares/metabolismo , Células PC12 , Enfermedad de Parkinson/metabolismo , Ratas , Ubiquitina-Proteína Ligasas/genética
7.
Int J Mol Sci ; 19(7)2018 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-29996472

RESUMEN

Extracellular signal-regulated kinase 5 (ERK5) regulates diverse physiological responses such as proliferation, differentiation, and gene expression. Previously, we demonstrated that ERK5 is essential for neurite outgrowth and catecholamine biosynthesis in PC12 cells and sympathetic neurons. However, it remains unclear how ERK5 regulates the activity of ion channels, which are important for membrane excitability. Thus, we examined the effect of ERK5 on the ion channel activity in the PC12 cells that overexpress both ERK5 and the constitutively active MEK5 mutant. The gene and protein expression levels of voltage-dependent Ca2+ and K⁺ channels were determined by RT-qPCR or Western blotting. The A-type K⁺ current was recorded using the whole-cell patch clamp method. In these ERK5-activated cells, the gene expression levels of voltage-dependent L- and P/Q-type Ca2+ channels did not alter, but the N-type Ca2+ channel was slightly reduced. In contrast, those of Kv4.2 and Kv4.3, which are components of the A-type current, were significantly enhanced. Unexpectedly, the protein levels of Kv4.2 were not elevated by ERK5 activation, but the phosphorylation levels were increased by ERK5 activation. By electrophysiological analysis, the inactivation time constant of the A-type current was prolonged by ERK5 activation, without changes in the peak current. Taken together, ERK5 inhibits an inactivation of the A-type current by phosphorylation of Kv4.2, which may contribute to the neuronal differentiation process.


Asunto(s)
Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Animales , Catecolaminas/biosíntesis , Diferenciación Celular , Regulación de la Expresión Génica , Potenciales de la Membrana , Neuronas/citología , Neuronas/metabolismo , Células PC12 , Técnicas de Placa-Clamp , Fosforilación , Ratas , Transducción de Señal
8.
Biochem Biophys Res Commun ; 493(1): 409-415, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28882596

RESUMEN

Diabetes mellitus affects ion channel physiology. We have previously reported that acute application of insulin suppresses the KCNQ1/KCNE1 currents that play an important role in terminating ventricular action potential. In this study, we investigated the effect of long-term insulin treatment on KCNQ1/KCNE1 currents using the Xenopus oocyte expression system. Insulin treatment with a duration longer than 6 h had an opposite effect to acute insulin application, that is, it augmented the KCNQ1/KCNE1 currents. Inhibitors of PI3K, wortmannin and LY294002, and a MEK inhibitor, U0126, abolished the potentiating effect of long-term insulin treatment. The long-term treatment with insulin had no effect on KCNQ1 currents indicating an essential role of KCNE1 in the insulin effect, which is similar to the acute insulin effect. Cycloheximide, an inhibitor of protein synthesis, and brefeldin A, an inhibitor of protein transport from endoplasmic reticulum, suppressed the long-term insulin effect. Western blotting analysis combined with these pharmacological data suggest that long-term insulin treatment augments KCNQ1/KCNE1 currents by increasing KCNE1 protein expression.


Asunto(s)
Insulina/administración & dosificación , Activación del Canal Iónico/fisiología , Canal de Potasio KCNQ1/metabolismo , Potenciales de la Membrana/fisiología , Canales de Potasio con Entrada de Voltaje/metabolismo , Potasio/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Oocitos/efectos de los fármacos , Oocitos/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Xenopus laevis
9.
Pflugers Arch ; 466(5): 937-46, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24068254

RESUMEN

Abnormal QT prolongation in diabetic patients has become a clinical problem because it increases the risk of lethal ventricular arrhythmia. In an animal model of type 1 diabetes mellitus, several ion currents, including the slowly activating delayed rectifier potassium current (IKs), are altered. The IKs channel is composed of KCNQ1 and KCNE1 subunits, whose genetic mutations are well known to cause long QT syndrome. Although insulin is known to affect many physiological and pathophysiological events in the heart, acute effects of insulin on cardiac ion channels are poorly understood at present. This study was designed to investigate direct electrophysiological effects of insulin on IKs (KCNQ1/KCNE1) currents. KCNQ1 and KCNE1 were co-expressed in Xenopus oocytes, and whole cell currents were measured by a two-microelectrode voltage-clamp method. Acute application of insulin suppressed the KCNQ1/KCNE1 currents and phosphorylated Akt and extracellular signal-regulated kinase (ERK), the two major downstream effectors, in a concentration-dependent manner. Wortmannin (10(-6) M), a phosphoinositide 3-kinase (PI3K) inhibitor, attenuated the suppression of the currents and phosphorylation of Akt by insulin, whereas U0126 (10(-5) M), a mitogen-activated protein kinase kinase (MEK) inhibitor, had no effect on insulin-induced suppression of the currents. In addition, insulin had little effect on KCNQ1 currents without KCNE1, which indicated an essential role of KCNE1 in the acute suppressive effects of insulin. Mutagenesis studies revealed amino acid residues 111-118 within the distal third C-terminus of KCNE1 as an important region. Insulin has direct electrophysiological effects on IKs currents, which may affect cardiac excitability.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Insulina/farmacología , Canal de Potasio KCNQ1/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Androstadienos/farmacología , Animales , Sitios de Unión , Butadienos/farmacología , Humanos , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/genética , Mutación , Nitrilos/farmacología , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Inhibidores de Proteínas Quinasas/farmacología , Wortmanina , Xenopus
10.
Neurochem Res ; 38(11): 2397-407, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24057400

RESUMEN

The cellular effects of eleven compounds including chalcone glycosides isolated from Brassica rapa L. 'hidabeni' and their synthetic derivatives were studied in rat pheochromocytoma PC12 cells. Of the compounds tested, 4'-O-ß-D-glucopyranosyl-3',4-dimethoxychalcone (A2) significantly increased the levels of the phosphorylated forms of extracellular signal-regulated kinases 1/2 (ERK 1/2), p38 mitogen-activated protein kinase (p38MAPK), and stress-activated protein kinases/Jun amino-terminal kinases (JNK/SAPK), but it did not affect Akt. Nerve growth factor (NGF), a well-known neurotrophic factor, increased the levels of phosphorylated ERK1/2, JNK/SAPK, and Akt but not p38MAPK, which may mediate marked neurite outgrowth. Signals evoked by A2 shared common characteristics with those induced by NGF; therefore, we evaluated the neuritogenic activity of A2 and found it induced only weak neurite outgrowth. However, this effect was enhanced by pre-treatment with a p38MAPK inhibitor, suggesting that the phosphorylation of p38MAPK down-regulated neurite outgrowth. From the results of this study, it was found that A2 in combination with a p38MAPK inhibitor can induce NGF-like effects. Hence, a combination of chalcone glycosides containing A2 and a p38MAPK inhibitor increases the likelihood that chalcone glycosides could be put to practical use in the form of drugs or alternative medicines to maintain neural health.


Asunto(s)
Chalconas/farmacología , Neuritas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Chalconas/síntesis química , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuritas/efectos de los fármacos , Células PC12 , Piridinas/farmacología , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Pharmacol Sci ; 120(4): 264-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23165803

RESUMEN

Understanding the regulation of cardiac ion channels is critical for the prevention of arrhythmia caused by abnormal excitability. Ion channels can be regulated by a change in function (qualitative) and a change in number (quantitative). Functional changes have been extensively investigated for many ion channels including cardiac voltage-dependent potassium channels. By contrast, the regulation of ion channel numbers has not been widely examined, particularly with respect to acute modulation of ion channels. This article briefly summarizes stimulus-induced endocytic regulation of major voltage-dependent potassium channels in the heart. The stimuli known to cause their endocytosis include receptor activation, drugs, and low extracellular [K(+)], following which the potassium channels undergo either clathrin-mediated or caveolin-mediated endocytosis. Receptor-mediated endocytic regulation has been demonstrated for Kv1.2, Kv1.5, KCNQ1 (Kv7.1), and Kv4.3, while drug-induced endocytosis has been demonstrated for Kv1.5 and hERG. Low [K(+)](o)-induced endocytosis might be unique for hERG channels, whose electrophysiological characteristics are known to be under strong influence of [K(+)](o). Although the precise mechanisms have not been elucidated, it is obvious that major cardiac voltage-dependent potassium channels are modulated by endocytosis, which leads to changes in cardiac excitability.


Asunto(s)
Endocitosis/fisiología , Miocardio/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio con Entrada de Voltaje/fisiología , Animales , Antiarrítmicos/farmacología , Arritmias Cardíacas/etiología , Caveolinas/fisiología , Clatrina/fisiología , Perros , Endocitosis/efectos de los fármacos , Humanos , Bloqueadores de los Canales de Potasio/farmacología , Quinidina/farmacología , Receptores Acoplados a Proteínas G/metabolismo
12.
Bioorg Med Chem ; 19(2): 749-53, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21215644

RESUMEN

Three new Lycopodium alkaloids, lyconadins D (1) and E (2), and complanadine E (3), were isolated from the club moss Lycopodium complanatum. Lyconadin D (1) was the first example of fastigiatine-type alkaloid isolated from Lycopodium complanatum. The structures and relative stereochemistry of 1-3 were elucidated on the basis of spectroscopic data. Complanadine E (3) enhanced mRNA expression for NGF.


Asunto(s)
Alcaloides/química , Compuestos Heterocíclicos de 4 o más Anillos/química , Lycopodium/química , Alcaloides/aislamiento & purificación , Línea Celular Tumoral , Compuestos Heterocíclicos de 4 o más Anillos/aislamiento & purificación , Humanos , Espectroscopía de Resonancia Magnética , Conformación Molecular , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , ARN Mensajero/metabolismo
13.
Life Sci ; 269: 119043, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33453240

RESUMEN

AIMS: Azelnidipine, a third-generation dihydropyridine calcium channel blocker (DHP CCB), has a characteristic hypotensive effect that persists even after it has disappeared from the plasma, which is thought to be due to its high hydrophobicity. However, because azelnidipine is unique, it might have other unknown effects on L-type Cav1.2 channels that result in the long-lasting decrease of blood pressure. The aim of this study was to investigate the potential quantitative modification of Cav1.2 by azelnidipine. MAIN METHODS: HEK293 cells were used to express Cav1.2 channels. Immunocytochemical analysis was performed to detect changes in the surface expression of the pore-forming subunit of the Cav1.2 channel, Cav1.2α1c. Western blotting analysis was performed to evaluate changes in expression levels of total Cav1.2α1c and Cavß2c. KEY FINDINGS: The surface expression of Cav1.2α1c was markedly reduced by treatment with azelnidipine, but not with other DHP CCBs (amlodipine and nicardipine). Results obtained with a dynamin inhibitor and an early endosome marker suggested that the reduction of surface Cav1.2α1c was not likely caused by internalization. Azelnidipine reduced the total amount of Cav1.2α1c protein in HEK293 cells and rat pulmonary artery smooth muscle cells. The reduction of Cav1.2α1c was rescued by inhibiting proteasome activity. In contrast, azelnidipine did not affect the amount of auxiliary Cavß2c subunits that function as a chaperone of Cav1.2. SIGNIFICANCE: This study is the first to demonstrate that azelnidipine reduces the expression of Cav1.2α1c, which might partly explain its long-lasting hypotensive effect.


Asunto(s)
Ácido Azetidinocarboxílico/análogos & derivados , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Dihidropiridinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Arteria Pulmonar/metabolismo , Animales , Ácido Azetidinocarboxílico/farmacología , Canales de Calcio Tipo L/química , Células Cultivadas , Células HEK293 , Humanos , Músculo Liso Vascular/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Ratas
14.
Mol Pharmacol ; 77(1): 10-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19858097

RESUMEN

Extracellular signal-regulated kinases (ERKs) or mitogen-activated protein kinases (MAPKs) are involved in cellular proliferation, differentiation, migration, and gene expression. The MAPK family includes ERK1/2, c-Jun NH(2)-terminal kinases 1, 2, and 3, p38MAPK alpha, beta, gamma, and -delta, and ERK5 as conventional MAPKs and ERK3, ERK4 NLK, and ERK7 as atypical MAPKs. Like other MAPKs, ERK5 is activated by variety of stimuli, including growth factors, G-protein-coupled receptor (GPCR) agonists, cytokines, and stress. However, the signaling pathway leading to ERK5 activation is not well understood compared with the other conventional MAPKs. For example, the pharmacological reagents that induce second messenger cAMP and Ca(2+) downstream of GPCRs do not activate ERK5 in neuronal cells. In addition, conflicting results have come from studies examining the involvement of small G-proteins in ERK5 activation by growth factors, and the details of the signaling pathway remain controversial. In addition, the physiological roles of ERK5 in neuronal cells have not been clarified. One reason was the lack of a selective ERK5 pharmacological inhibitor until the novel selective MEK5/ERK5 inhibitors BIX02188 and BIX02189 (Biochem Biophys Res Commun 377:120-125, 2008) reported last year. Another reason is that the use of interfering mutants is limited in neuronal cells because the transfection efficiency is low. Despite these difficulties, recent studies suggest that ERK5 mediates the promotion of neuronal survival and neuronal differentiation in vitro and in vivo. In this review, the signaling pathway leading to ERK5 activation through heterotrimeric and small G-proteins and the physiological roles of ERK5 in neuronal cells are summarized and discussed.


Asunto(s)
Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Transducción de Señal , Proteínas de Unión al GTP/metabolismo , Neuronas/metabolismo
15.
J Pharmacol Exp Ther ; 334(3): 809-19, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20511347

RESUMEN

Lipid rafts, formed by sphingolipids and cholesterol within the membrane bilayer, are believed to have a critical role in signal transduction. P2Y(2) receptors are known to couple with G(q) family G proteins, causing the activation of phospholipase C (PLC) and an increase in intracellular Ca(2+) ([Ca(2+)](i)) levels. In the present study, we investigated the involvement of lipid rafts in P2Y(2) receptor-mediated signaling and cell migration in NG 108-15 cells. When NG 108-15 cell lysates were fractionated by sucrose density gradient centrifugation, Galpha(q/11) and a part of P2Y(2) receptors were distributed in a fraction where the lipid raft markers, cholesterol, flotillin-1, and ganglioside GM1 were abundant. Methyl-beta-cyclodextrin (CD) disrupted not only lipid raft markers but also Galpha(q/11) and P2Y(2) receptors in this fraction. In the presence of CD, P2Y(2) receptor-mediated phosphoinositide hydrolysis and [Ca(2+)](i) elevation were inhibited. It is noteworthy that UTP-induced cell migration was inhibited by CD or the G(q/11)-selective inhibitor YM254890 [(1R)-1-{(3S,6S,9S,12S,18R,21S,22R)-21-acetamido-18-benzyl-3-[(1R)-1-methoxyethyl]-4,9,10,12,16, 22-hexamethyl-15-methylene-2,5,8,11,14,17,-20-heptaoxo-1,19-dioxa-4,7,10,13,16-pentaazacyclodocosan-6-yl}-2-methylpropyl rel-(2S,3R)-2-acetamido-3-hydroxy-4-methylpentanoate]. Moreover CD and YM254890 completely inhibited Rho-A activation. Downstream of Rho-A signaling, stress fiber formation and phosphorylation of cofilin were also inhibited by CD or YM254890. However, UTP-induced phosphorylation of cofilin was not affected by the expression of p115-regulator of G protein signaling, which inhibits the G(12/13) signaling pathway. This implies that UTP-induced Rho-A activation was relatively regulated by the G(q/11) signaling pathway. These results suggest that lipid rafts are critical for P2Y(2) receptor-mediated G(q/11)-PLC-Ca(2+) signaling and this cascade is important for cell migration in NG 108-15 cells.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Microdominios de Membrana/fisiología , Receptores Purinérgicos P2/fisiología , Uridina Trifosfato/farmacología , Citoesqueleto de Actina/fisiología , Factores Despolimerizantes de la Actina/metabolismo , Western Blotting , Línea Celular , Colesterol/metabolismo , Colorantes , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Humanos , L-Lactato Deshidrogenasa/metabolismo , Microdominios de Membrana/efectos de los fármacos , Péptidos Cíclicos/farmacología , Fosfatidilinositoles/metabolismo , Fosforilación , Receptores Purinérgicos P2Y2 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sales de Tetrazolio , Tiazoles , Proteínas de Unión al GTP rho/metabolismo
16.
Cell Signal ; 20(7): 1275-83, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18407464

RESUMEN

Extracellular signal-regulated kinases (ERKs) play important physiological roles in proliferation, differentiation and gene expression. ERK5 is twice the size of ERK1/2, the amino-terminal half contains the kinase domain that shares the homology with ERK1/2 and TEY activation motif, whereas the carboxy-terminal half is unique. In this study, we examined the cross-talk mechanism between G-protein-coupled receptors (GPCRs) and receptor tyrosine kinases, focusing on ERK1/2 and 5. The pretreatment of rat pheochromocytoma cells (PC12) with pertussis toxin (PTX) specifically enhanced epidermal growth factor (EGF)-induced ERK5 phosphorylation. In addition, lysophosphatidic acid (LPA) attenuated the EGF-induced ERK5 phosphorylation in LPA(1) receptor- and G(i/o)-dependent manners. On the other hand, LPA alone activated ERK1/2 via Gbetagamma subunits and Ras and potentiated EGF-induced ERK1/2 phosphorylation at late time points. These results suggest G(i/o) negatively regulates ERK5, while it positively regulates ERK1/2. LPA did not affect cAMP levels after EGF treatment, and the reagents promoting cAMP production such as forskolin and cholera toxin also attenuated the EGF-induced ERK5 phosphorylation, indicating that the inhibitory effect of LPA on ERK5 inhibition via G(i/o) is not due to inhibition of adenylyl cyclase by Galpha(i/o). However, the inhibitory effect of LPA on ERK5 was abolished in PC12 cells stably overexpressing C-terminus of GPCR kinase2 (GRK2), and overexpression of Gbeta(1) and gamma(2) subunits also suppressed ERK5 phosphorylation by EGF. In response to LPA, Gbetagamma subunits interacted with EGF receptor in a time-dependent manner. These results strongly suggest that LPA negatively regulates the EGF-induced ERK5 phosphorylation through Gbetagamma subunits.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades de Proteína/metabolismo , Animales , Colforsina/farmacología , AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Receptores ErbB/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Modelos Biológicos , Células PC12 , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Ratas , Receptores del Ácido Lisofosfatídico/metabolismo , Proteínas ras/metabolismo
17.
Ann Clin Transl Neurol ; 6(11): 2205-2211, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31588691

RESUMEN

OBJECTIVE: Genetic analysis of patients with familial Parkinson's disease (PD) identified many causative genes. However, the majority of PD cases are sporadic, and the mechanisms of onset still remain unclear. Previously, we found that Midnolin (MIDN) is associated with PD in a Yamagata (Japan) cohort study and that MIDN regulates neurite outgrowth and Parkin expression in neuronal cells. In the present study, we aimed to replicate the genetic association between MIDN and PD in a large British population cohort. METHODS: In this replication study, we analyzed the copy number variations and single-nucleotide polymorphisms of the MIDN gene in a large British population on a case-control genome-wide association study dataset including 2,860 controls and 2,168 PD patients. RESULTS: There was significant copy number loss in the MIDN gene with an odds ratio of 4.35 (P < 2.2 × 10-16 ). Furthermore, there were many patients in both the British and Yamagata case groups who have a long spanning deletion. The odds ratio dramatically increased to 22.3 (P = 3.59 × 10-15 ) when a deletion spanning more than 50,000 bp was defined as the copy number loss. There were no significant differences between the controls and study cases for two relatively frequent single-nucleotide polymorphisms (rs3746106 and rs3746107). INTERPRETATION: We showed the strong genetic association of MIDN with PD development in a British population and in a Japanese population, suggesting MIDN is a confirmed and universal genetic risk factor for PD.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Proteínas Nucleares/genética , Enfermedad de Parkinson/genética , Estudios de Casos y Controles , Variaciones en el Número de Copia de ADN/genética , Estudio de Asociación del Genoma Completo , Humanos , Japón , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Reino Unido
18.
Biochem Pharmacol ; 169: 113628, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31491415

RESUMEN

KCNQ1 (Kv7.1 or KvLQT1) plays important physiological roles in various tissues forming potassium channels with KCNE subunits. Among the channels formed by KCNQ1 and KCNE subunits, the best studied is the slow delayed rectifier potassium channel in the heart, the IKs (KCNQ1/KCNE1) channel, which is critical for repolarization of cardiac action potential. The KCNQ1 channel is internalized by Nedd4/Nedd4-like ligase-dependent ubiquitination. It is also reported that phosphorylation of KCNE1 by PKC results in internalization of the KCNQ1/KCNE1 channel. Because we have observed down-regulation of KCNQ1/KCNE1 currents by activation of the α1-adrenergic receptor (α1AR) that activates PKC, this study investigated whether α1AR causes internalization of the KCNQ1 protein. We fused HaloTag to the extracellular region of KCNQ1 (Halo-KCNQ1) and co-expressed it with α1ARs in HEK293 cells. The KCNQ1 protein on the cell surface was selectively labeled with membrane-impermeable HaloTag ligands, and changes in its localization were monitored by confocal fluorescence microscopy. Activation of α1AAR and α1BAR caused marked internalization of KCNQ1, which was not KCNE1-dependent. Internalization of KCNQ1 by α1AR activation was inhibited by disruption of the PY motif or the YXXΦ motif in the C-terminus. Double staining for the receptor and the channel revealed that KCNQ1 internalization was independent of α1AR internalization. Our results suggest that α1AR-mediated direct internalization of KCNQ1 is AP2/clathrin-dependent and may be triggered by ubiquitination of KCNQ1 via the AMP dependent kinase (AMPK)/Nedd4-2 pathway. When phenylephrine was applied to rat neonatal cardiomyocytes transfected with KCNQ1 and α1AR, the KCNQ1 protein was internalized. The internalization of KCNQ1 by α1AR would affect pathophysiology in a variety of tissues expressing KCNQ1, which merits further in vivo study.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Receptores Adrenérgicos alfa 1/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Células HEK293 , Humanos , Miocitos Cardíacos/metabolismo , Proteína Quinasa C/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Transferrina/análisis
19.
Brain Res ; 1184: 57-64, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-17980863

RESUMEN

Since neurotrophic factors are essential for neurons to form neuronal networks and maintain neuronal functions, neurotrophic factor-like substances or inducers of neurotrophic factors can be useful for the treatment of serious neuronal diseases such as Alzheimer's and Parkinson's diseases. In the present study, we examined an effect of 5,19-cyclo-9beta,10xi-androstane-3,17-dione (CAD) on neurotrophic factor synthesis in glial cells and scopolamine-induced impairment of learning in mice. 1321N1 human astrocytoma cells promoted secretion of certain neurotrophic factors in response to CAD with no cytotoxicity, which caused dramatic neurite outgrowth in rat pheochromocytoma (PC12) cells. In fact, CAD significantly enhanced nerve growth factor (NGF) secretion and its gene expression in 1321N1 cells, in a time and concentration-dependent manner. Because second messengers such as cAMP, inositol 1,4,5-trisphosphates and Ca(2+) induce NGF gene expression, we measured activities of adenylyl cyclase and phospholipase C and intracellular Ca(2+) concentration in 1321N1 cells. However, CAD changed neither second messenger levels. CAD enhanced the gene expression of proto-oncogene, c-fos that is one of the components of transcription factor (AP-1). In addition to those above, the in vivo effects of CAD were also examined. Although injection of muscarinic receptor antagonist scopolamine impaired passive avoidance learning in mice, pretreatment with CAD significantly reversed the adverse effect in a dose-dependent manner. Taking these results together, CAD has enormous therapeutic potential for serious neuronal diseases.


Asunto(s)
Astrocitoma/metabolismo , Reacción de Prevención/efectos de los fármacos , Diterpenos/uso terapéutico , Discapacidades para el Aprendizaje/tratamiento farmacológico , Factor de Crecimiento Nervioso/metabolismo , Androsterona/análogos & derivados , Androsterona/farmacología , Androsterona/uso terapéutico , Animales , Astrocitoma/patología , Conducta Animal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , AMP Cíclico/metabolismo , Diterpenos/química , Diterpenos/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Fosfatos de Inositol/metabolismo , Discapacidades para el Aprendizaje/inducido químicamente , Masculino , Ratones , Neuritas/efectos de los fármacos , Células PC12 , Proto-Oncogenes Mas , Ratas , Escopolamina , Sales de Tetrazolio , Tiazoles
20.
Life Sci ; 80(18): 1669-77, 2007 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-17337280

RESUMEN

We examined the effects of cyrneine A, a novel diterpene isolated from the mushroom Sarcodon cyrneus, on morphology of rat pheochromocytoma cells (PC12). In response to cyrneine A, PC12 cells extended their neurites, an effect partially blocked by the extracellular signal-regulated kinase (ERK) kinase inhibitor PD98059, but not by the protein kinase C inhibitor GF109203X, nor the phosphatidylinositol-3-kinase inhibitor wortmannin. Cyrneine A did not activate ERK at any of the time points tested (5-120 min), indicating that only the basal activity of ERK is required for cyrneine A-induced neurite outgrowth. As transcriptional regulation is required for neurite extension, the activity of three major transcription factors was determined. Cyrneine A enhanced activation of the transcription factors activator protein-1 (AP-1) and nuclear factor-kappaB, but not CREB, and this was accompanied by enhanced c-fos expression. Moreover, we determined the role of Rac1, a small GTPase protein of the Rho family that regulates actin dynamics, in cyrneine A-induced neurite outgrowth. Treatment with cyrneine A led to actin translocation and subsequently, to accumulation of F-actin at the tip of neurites. Rac1 activity was increased by cyrneine A and expression of a dominant-negative Rac1 mutant significantly inhibited the cyrneine A-induced extension of neurites. These results suggest that cyrneine A induces neurite outgrowth in a Rac1-dependent mechanism.


Asunto(s)
Actinas/metabolismo , Diterpenos/farmacología , Neuritas/metabolismo , Factores de Transcripción/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Androstadienos/farmacología , Animales , Basidiomycota/química , Diterpenos/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Indoles/farmacología , Maleimidas/farmacología , Mutación , Células PC12 , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Ratas , Wortmanina , Proteína de Unión al GTP rac1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA