Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Biol Pharm Bull ; 46(3): 379-381, 2023.
Article in English | MEDLINE | ID: mdl-36858566

ABSTRACT

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.


Subject(s)
Nuclear Proteins , Parkinson Disease , Humans , Cohort Studies , Parkinson Disease/genetics , Risk Factors , Nuclear Proteins/genetics , Japan
2.
Int J Mol Sci ; 23(15)2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35955582

ABSTRACT

(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.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , MicroRNAs , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , MicroRNAs/metabolism
3.
J Pharmacol Exp Ther ; 381(2): 68-78, 2022 05.
Article in English | MEDLINE | ID: mdl-35241633

ABSTRACT

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.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Humans , Insulin/pharmacology , Nuclear Proteins , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Risk Factors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Transcription, Genetic
4.
J Pharmacol Sci ; 148(3): 326-330, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35177212

ABSTRACT

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.


Subject(s)
Aniline Compounds/pharmacology , Corpus Striatum/cytology , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Indoles/pharmacology , Methantheline/adverse effects , Microglia/drug effects , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/physiology , Aniline Compounds/therapeutic use , Animals , Calcium-Binding Proteins/metabolism , Corpus Striatum/metabolism , Indoles/therapeutic use , Mice , Microfilament Proteins/metabolism , Microglia/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Psychomotor Agitation , Substance-Related Disorders/prevention & control
5.
Life Sci ; 269: 119043, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33453240

ABSTRACT

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.


Subject(s)
Azetidinecarboxylic Acid/analogs & derivatives , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Dihydropyridines/pharmacology , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Animals , Azetidinecarboxylic Acid/pharmacology , Calcium Channels, L-Type/chemistry , Cells, Cultured , HEK293 Cells , Humans , Muscle, Smooth, Vascular/drug effects , Pulmonary Artery/drug effects , Rats
7.
Ann Clin Transl Neurol ; 6(11): 2205-2211, 2019 11.
Article in English | MEDLINE | ID: mdl-31588691

ABSTRACT

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.


Subject(s)
Genetic Predisposition to Disease/genetics , Nuclear Proteins/genetics , Parkinson Disease/genetics , Case-Control Studies , DNA Copy Number Variations/genetics , Genome-Wide Association Study , Humans , Japan , Polymorphism, Single Nucleotide/genetics , Risk Factors , United Kingdom
8.
Biochem Pharmacol ; 169: 113628, 2019 11.
Article in English | MEDLINE | ID: mdl-31491415

ABSTRACT

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.


Subject(s)
KCNQ1 Potassium Channel/metabolism , Receptors, Adrenergic, alpha-1/physiology , AMP-Activated Protein Kinases/physiology , Animals , HEK293 Cells , Humans , Myocytes, Cardiac/metabolism , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Receptors, Transferrin/analysis
9.
J Biol Chem ; 294(44): 16049-16061, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31506297

ABSTRACT

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.


Subject(s)
Action Potentials , Chloride Channels/metabolism , HSC70 Heat-Shock Proteins/metabolism , Myocytes, Cardiac/metabolism , Pulmonary Veins/cytology , Animals , Binding Sites , CLC-2 Chloride Channels , Cells, Cultured , Chloride Channels/chemistry , HEK293 Cells , HSC70 Heat-Shock Proteins/chemistry , HSC70 Heat-Shock Proteins/genetics , Heart Ventricles/cytology , Humans , Male , Molecular Docking Simulation , Myocytes, Cardiac/physiology , PC12 Cells , Protein Binding , Pulmonary Veins/metabolism , Rats , Rats, Wistar
10.
Int J Mol Sci ; 19(7)2018 Jul 10.
Article in English | MEDLINE | ID: mdl-29996472

ABSTRACT

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.


Subject(s)
Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Animals , Catecholamines/biosynthesis , Cell Differentiation , Gene Expression Regulation , Membrane Potentials , Neurons/cytology , Neurons/metabolism , PC12 Cells , Patch-Clamp Techniques , Phosphorylation , Rats , Signal Transduction
11.
Biol Pharm Bull ; 41(1): 20-23, 2018.
Article in English | MEDLINE | ID: mdl-29311479

ABSTRACT

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.


Subject(s)
Nuclear Proteins/genetics , Parkinson Disease/genetics , RNA, Messenger/genetics , Transcriptome , Animals , Gene Expression Profiling , Gene Knockdown Techniques , Nuclear Proteins/metabolism , PC12 Cells , Parkinson Disease/metabolism , Rats , Ubiquitin-Protein Ligases/genetics
12.
Biochem Biophys Res Commun ; 493(1): 409-415, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28882596

ABSTRACT

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.


Subject(s)
Insulin/administration & dosage , Ion Channel Gating/physiology , KCNQ1 Potassium Channel/metabolism , Membrane Potentials/physiology , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Oocytes/drug effects , Oocytes/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Xenopus laevis
13.
Sci Rep ; 7(1): 5885, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724963

ABSTRACT

Midnolin (MIDN) was first discovered in embryonic stem cells, but its physiological and pathological roles are, to date, poorly understood. In the present study, we therefore examined the role of MIDN in detail. We found that in PC12 cells, a model of neuronal cells, MIDN localized primarily to the nucleus and intracellular membranes. Nerve growth factor promoted MIDN gene expression, which was attenuated by specific inhibitors of extracellular signal-regulated kinases 1/2 and 5. MIDN-deficient PC12 cells created using CRISPR/Cas9 technology displayed significantly impaired neurite outgrowth. Interestingly, a genetic approach revealed that 10.5% of patients with sporadic Parkinson's disease (PD) had a lower MIDN gene copy number whereas no copy number variation was observed in healthy people, suggesting that MIDN is involved in PD pathogenesis. Furthermore, the expression of parkin, a major causative gene in PD, was significantly reduced by CRISPR/Cas9 knockout and siRNA knockdown of MIDN. Activating transcription factor 4 (ATF4) was also down-regulated, which binds to the cAMP response element (CRE) in the parkin core promoter region. The activity of CRE was reduced following MIDN loss. Overall, our data suggests that MIDN promotes the expression of parkin E3 ubiquitin ligase, and that MIDN loss can trigger PD-related pathogenic mechanisms.


Subject(s)
Nuclear Proteins/metabolism , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/metabolism , Aged , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Dosage , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Models, Biological , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , Nuclear Proteins/genetics , PC12 Cells , Rats , Signal Transduction
14.
PLoS One ; 12(3): e0173628, 2017.
Article in English | MEDLINE | ID: mdl-28282426

ABSTRACT

Licochalcones extracted from Glycyrrhiza inflata are known to have a variety of biological properties such as anti-inflammatory, anti-bacterial, and anti-tumor activities, but their action on platelet aggregation has not yet been reported. Therefore, in this study we investigated the effects of licochalcones on platelet aggregation. Collagen and U46619, a thromboxane A2 receptor agonist, caused rabbit platelet aggregation, which was reversed by pretreatment with licochalcones A, C and D in concentration-dependent manners. Among these compounds, licochalcone A caused the most potent inhibitory effect on collagen-induced platelet aggregation. However, the licochalcones showed marginal inhibitory effects on thrombin or ADP-induced platelet aggregation. In addition to rabbit platelets, licochalcone A attenuated collagen-induced aggregation in human platelets. Because licochalcone A also inhibited arachidonic acid-induced platelet aggregation and production of thromboxane A2 induced by collagen in intact platelets, we further examined the direct interaction of licochalcone A with cyclooxygenase (COX)-1. As expected, licochalcone A caused an inhibitory effect on both COX-1 and COX-2 in vitro. Regarding the effect of licochalcone A on COX-1 enzyme reaction kinetics, although licochalcone A showed a stronger inhibition of prostaglandin E2 synthesis induced by lower concentrations of arachidonic acid, Vmax values in the presence or absence of licochalcone A were comparable, suggesting that it competes with arachidonic acid at the same binding site on COX-1. These results suggest that licochalcones inhibit collagen-induced platelet aggregation accompanied by inhibition of COX-1 activity.


Subject(s)
Blood Platelets/enzymology , Chalcones , Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors , Glycyrrhiza/chemistry , Platelet Aggregation/drug effects , Animals , Chalcones/chemistry , Chalcones/isolation & purification , Chalcones/pharmacology , Collagen/pharmacology , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/isolation & purification , Cyclooxygenase Inhibitors/pharmacology , Male , Rabbits
15.
Intern Med ; 55(24): 3611-3621, 2016.
Article in English | MEDLINE | ID: mdl-27980262

ABSTRACT

Composite pheochromocytoma (cPC) is extremely rare, arising in the adrenal medulla as a mixture of PC and other tumors of neural origin. We herein report on a case of adrenal incidentaloma post-operatively diagnosed as cPC with ganglioneuroblastoma (GNBL). The PC component had 7 points on the PASS, a Ki-67 index of 5.1%, a focal absence of sustentacular cells, and no genetic aberrations in succinate dehydrogenase subunit B. The GNBL component exhibited no N-myc amplification. Tumor cells of both components were stained positively for extracellular signal-regulated kinase 5 and ankyrin repeat domain 1. The aberrant activation of growth signaling may play a role in the marginal malignancy of cPC.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Adrenal Medulla/metabolism , Ganglioneuroblastoma/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Pheochromocytoma/metabolism , Repressor Proteins/metabolism , Adrenal Gland Neoplasms/pathology , Adrenal Gland Neoplasms/surgery , Adrenal Medulla/diagnostic imaging , Adrenal Medulla/pathology , Adrenalectomy , Female , Follow-Up Studies , Ganglioneuroblastoma/pathology , Ganglioneuroblastoma/surgery , Humans , Incidental Findings , Middle Aged , Pheochromocytoma/pathology , Pheochromocytoma/surgery , Signal Transduction , Succinate Dehydrogenase/genetics , Treatment Outcome
16.
Cell Signal ; 28(3): 177-189, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26739108

ABSTRACT

Extracellular signal-regulated kinases (ERKs) play important roles in proliferation, differentiation and gene expression. In our previous study, we demonstrated that both ERK5 and ERK1/2 were responsible for neurite outgrowth and tyrosine hydroxylase (TH) expression in rat pheochromocytoma cells (PC12) (J Biol Chem 284, 23,564-23,573, 2009). However, the functional differences between ERK5 and ERK1/2 signaling in neural differentiation remain unclear. In the present study, we show that ERK5, but not ERK1/2 regulates TH levels in rat sympathetic neurons. Furthermore, microarray analysis performed in PC12 cells using ERK5 and ERK1/2-specific inhibitors, identified ankyrin repeat domain 1 (ankrd1) as an ERK5-dependent and ERK1/2-independent gene. Here, we report a novel role of the ERK5/ankrd1 signaling in regulating TH levels and catecholamine biosynthesis. Ankrd1 mRNA was induced by nerve growth factor in time- and concentration-dependent manners. TH levels were reduced by ankrd1 knockdown with no changes in the mRNA levels, suggesting that ankrd1 was involved in stabilization of TH protein. Interestingly, ubiquitination of TH was enhanced and catecholamine biosynthesis was reduced by ankrd1 knockdown. Finally, we examined the relationship of ERK5 to TH levels in human adrenal pheochromocytomas. Whereas TH levels were correlated with ERK5 levels in normal adrenal medullas, ERK5 was down-regulated and TH was up-regulated in pheochromocytomas, indicating that TH levels are regulated by alternative mechanisms in tumors. Taken together, ERK5 signaling is required for catecholamine biosynthesis during neural differentiation, in part to induce ankrd1, and to maintain appropriate TH levels. This pathway is disrupted in pathological conditions.


Subject(s)
Catecholamines/biosynthesis , Mitogen-Activated Protein Kinase 7/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Adolescent , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Catecholamines/analysis , Chromatography, High Pressure Liquid , Down-Regulation/drug effects , Female , Humans , Male , Microscopy, Fluorescence , Middle Aged , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/genetics , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Nerve Growth Factor/pharmacology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , PC12 Cells , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction/drug effects , Tandem Mass Spectrometry , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/drug effects
17.
PLoS One ; 10(2): e0117914, 2015.
Article in English | MEDLINE | ID: mdl-25689862

ABSTRACT

Extracellular signal-regulated kinases (ERKs) play critical roles in numerous cellular processes, including proliferation and differentiation. ERK5 contains a kinase domain at the N-terminal, and the unique extended C-terminal includes multiple autophosphorylation sites that enhance ERK5-dependent transcription. However, the impact of phosphorylation at the various sites remain unclear. In this study, we examined the role of phosphorylation at the ERK5 C-terminal. We found that a constitutively active MEK5 mutant phosphorylated ERK5 at the TEY motif, resulting in the sequential autophosphorylation of multiple C-terminal residues, including Thr732 and Ser769/773/775. However, when ERK1/2 was selectively activated by an oncogenic RAS mutant, ERK5 phosphorylation at Thr732 was induced without affecting the phosphorylation status at TEY or Ser769/773/775. The Thr732 phosphorylation was U0126-sensitive and was observed in a kinase-dead mutant of ERK5 as well, suggesting that ERK1/2 can phosphorylate ERK5 at Thr732. This phosphorylation was also promoted by epidermal growth factor and nerve growth factor in HEK293 and PC12 cells, respectively. The ERK5-T732A mutant was localized in the cytosol under basal conditions. In contrast, ERK5 phosphorylated at Thr732 via the RAS-ERK1/2 pathway and ERK5-T732E, which mimics the phosphorylated form, were localized in both the nucleus and cytosol. Finally, ER-32A and U0126 blocked ERK5-dependent MEF2C transcriptional activity. Based on these findings, we propose a novel cross-talk mechanism in which ERK1/2, following activation by growth factor stimulation, phosphorylates ERK5 at Thr732. This phosphorylation event is responsible for ERK5 nuclear localization and ERK5-dependent transcription.


Subject(s)
Cell Nucleus/metabolism , Mitogen-Activated Protein Kinase 7/chemistry , Mitogen-Activated Protein Kinase 7/metabolism , Threonine/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus , Animals , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PC12 Cells , Phosphorylation , Rats
19.
Pflugers Arch ; 466(5): 937-46, 2014 May.
Article in English | MEDLINE | ID: mdl-24068254

ABSTRACT

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.


Subject(s)
Action Potentials/drug effects , Insulin/pharmacology , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Androstadienes/pharmacology , Animals , Binding Sites , Butadienes/pharmacology , Humans , KCNQ1 Potassium Channel/chemistry , KCNQ1 Potassium Channel/genetics , Mutation , Nitriles/pharmacology , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/genetics , Protein Kinase Inhibitors/pharmacology , Wortmannin , Xenopus
20.
J Cell Biol ; 202(6): 901-16, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-24019534

ABSTRACT

Reorganization of the actin cytoskeleton is responsible for dynamic regulation of endothelial cell (EC) barrier function. Circumferential actin bundles (CAB) promote formation of linear adherens junctions (AJs) and tightening of EC junctions, whereas formation of radial stress fibers (RSF) connected to punctate AJs occurs during junction remodeling. The small GTPase Rap1 induces CAB formation to potentiate EC junctions; however, the mechanism underlying Rap1-induced CAB formation remains unknown. Here, we show that myotonic dystrophy kinase-related CDC42-binding kinase (MRCK)-mediated activation of non-muscle myosin II (NM-II) at cell-cell contacts is essential for Rap1-induced CAB formation. Our data suggest that Rap1 induces FGD5-dependent Cdc42 activation at cell-cell junctions to locally activate the NM-II through MRCK, thereby inducing CAB formation. We further reveal that Rap1 suppresses the NM-II activity stimulated by the Rho-ROCK pathway, leading to dissolution of RSF. These findings imply that Rap1 potentiates EC junctions by spatially controlling NM-II activity through activation of the Cdc42-MRCK pathway and suppression of the Rho-ROCK pathway.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Myosin Type II/metabolism , Telomere-Binding Proteins/metabolism , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Human Umbilical Vein Endothelial Cells/cytology , Humans , Immunoenzyme Techniques , Myosin Type II/genetics , Myotonin-Protein Kinase , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , Shelterin Complex , Telomere-Binding Proteins/antagonists & inhibitors , Telomere-Binding Proteins/genetics , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL