Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(3): 102993, 2023 03.
Article in English | MEDLINE | ID: mdl-36758801

ABSTRACT

Heart failure is one of the leading causes of death worldwide. RhoA, a small GTPase, governs actin dynamics in various tissue and cell types, including cardiomyocytes; however, its involvement in cardiac function has not been fully elucidated. Here, we generated cardiomyocyte-specific RhoA conditional knockout (cKO) mice, which demonstrated a significantly shorter lifespan with left ventricular dilation and severely impaired ejection fraction. We found that the cardiac tissues of the cKO mice exhibited structural disorganization with fibrosis and also exhibited enhanced senescence compared with control mice. In addition, we show that cardiomyocyte mitochondria were structurally abnormal in the aged cKO hearts. Clearance of damaged mitochondria by mitophagy was remarkably inhibited in both cKO cardiomyocytes and RhoA-knockdown HL-1 cultured cardiomyocytes. In RhoA-depleted cardiomyocytes, we reveal that the expression of Parkin, an E3 ubiquitin ligase that plays a crucial role in mitophagy, was reduced, and expression of N-Myc, a negative regulator of Parkin, was increased. We further reveal that the RhoA-Rho kinase axis induced N-Myc phosphorylation, which led to N-Myc degradation and Parkin upregulation. Re-expression of Parkin in RhoA-depleted cardiomyocytes restored mitophagy, reduced mitochondrial damage, attenuated cardiomyocyte senescence, and rescued cardiac function both in vitro and in vivo. Finally, we found that patients with idiopathic dilated cardiomyopathy without causal mutations for dilated cardiomyopathy showed reduced cardiac expression of RhoA and Parkin. These results suggest that RhoA promotes Parkin-mediated mitophagy as an indispensable mechanism contributing to cardioprotection in the aging heart.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure , Animals , Mice , Cardiomyopathy, Dilated/metabolism , Heart Failure/metabolism , Mitochondria/metabolism , Mitophagy/genetics , Myocytes, Cardiac/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
FASEB J ; 34(5): 6399-6417, 2020 05.
Article in English | MEDLINE | ID: mdl-32175648

ABSTRACT

Brugada syndrome (BrS) is an inherited channelopathy responsible for almost 20% of sudden cardiac deaths in patients with nonstructural cardiac diseases. Approximately 70% of BrS patients, the causative gene mutation(s) remains unknown. In this study, we used whole exome sequencing to investigate candidate mutations in a family clinically diagnosed with BrS. A heterozygous 1616G>A substitution (R539Q mutation) was identified in the transmembrane protein 168 (TMEM168) gene of symptomatic individuals. Similar to endogenous TMEM168, both TMEM168 wild-type (WT) and mutant proteins that were ectopically induced in HL-1 cells showed nuclear membrane localization. A significant decrease in Na+ current and Nav 1.5 protein expression was observed in HL-1 cardiomyocytes expressing mutant TMEM168. Ventricular tachyarrhythmias and conduction disorders were induced in the heterozygous Tmem168 1616G>A knock-in mice by pharmacological stimulation, but not in WT mice. Na+ current was reduced in ventricular cardiomyocytes isolated from the Tmem168 knock-in heart, and Nav 1.5 expression was also impaired. This impairment was dependent on increased Nedd4-2 binding to Nav 1.5 and subsequent ubiquitination. Collectively, our results show an association between the TMEM168 1616G>A mutation and arrhythmogenesis in a family with BrS.


Subject(s)
Brugada Syndrome/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Mutation , Myocytes, Cardiac/pathology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Adult , Animals , Brugada Syndrome/pathology , Female , Humans , Male , Membrane Proteins/metabolism , Mice , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Pedigree , Young Adult
3.
Circulation ; 139(18): 2157-2169, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30764634

ABSTRACT

BACKGROUND: Bradyarrhythmia is a common clinical manifestation. Although the majority of cases are acquired, genetic analysis of families with bradyarrhythmia has identified a growing number of causative gene mutations. Because the only ultimate treatment for symptomatic bradyarrhythmia has been invasive surgical implantation of a pacemaker, the discovery of novel therapeutic molecular targets is necessary to improve prognosis and quality of life. METHODS: We investigated a family containing 7 individuals with autosomal dominant bradyarrhythmias of sinus node dysfunction, atrial fibrillation with slow ventricular response, and atrioventricular block. To identify the causative mutation, we conducted the family-based whole exome sequencing and genome-wide linkage analysis. We characterized the mutation-related mechanisms based on the pathophysiology in vitro. After generating a transgenic animal model to confirm the human phenotypes of bradyarrhythmia, we also evaluated the efficacy of a newly identified molecular-targeted compound to upregulate heart rate in bradyarrhythmias by using the animal model. RESULTS: We identified one heterozygous mutation, KCNJ3 c.247A>C, p.N83H, as a novel cause of hereditary bradyarrhythmias in this family. KCNJ3 encodes the inwardly rectifying potassium channel Kir3.1, which combines with Kir3.4 (encoded by KCNJ5) to form the acetylcholine-activated potassium channel ( IKACh channel) with specific expression in the atrium. An additional study using a genome cohort of 2185 patients with sporadic atrial fibrillation revealed another 5 rare mutations in KCNJ3 and KCNJ5, suggesting the relevance of both genes to these arrhythmias. Cellular electrophysiological studies revealed that the KCNJ3 p.N83H mutation caused a gain of IKACh channel function by increasing the basal current, even in the absence of m2 muscarinic receptor stimulation. We generated transgenic zebrafish expressing mutant human KCNJ3 in the atrium specifically. It is interesting to note that the selective IKACh channel blocker NIP-151 repressed the increased current and improved bradyarrhythmia phenotypes in the mutant zebrafish. CONCLUSIONS: The IKACh channel is associated with the pathophysiology of bradyarrhythmia and atrial fibrillation, and the mutant IKACh channel ( KCNJ3 p.N83H) can be effectively inhibited by NIP-151, a selective IKACh channel blocker. Thus, the IKACh channel might be considered to be a suitable pharmacological target for patients who have bradyarrhythmia with a gain-of-function mutation in the IKACh channel.


Subject(s)
Atrial Fibrillation , Atrioventricular Block , Bradycardia , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Genetic Diseases, Inborn , Mutation, Missense , Amino Acid Substitution , Animals , Animals, Genetically Modified , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Atrioventricular Block/genetics , Atrioventricular Block/metabolism , Atrioventricular Block/pathology , Atrioventricular Block/physiopathology , Benzopyrans/pharmacology , Bradycardia/genetics , Bradycardia/metabolism , Bradycardia/pathology , Bradycardia/physiopathology , Electrophysiologic Techniques, Cardiac , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/physiopathology , Humans , Male , Xenopus laevis , Zebrafish
4.
Heart Rhythm ; 16(1): 108-116, 2019 01.
Article in English | MEDLINE | ID: mdl-30075281

ABSTRACT

BACKGROUND: In the heart, slow delayed rectifier channels provide outward currents (IKs) for action potential (AP) repolarization in a region- and context-dependent manner. In diseased hearts, chronic elevation of angiotensin II (Ang II) may remodel IKs in a region-dependent manner, contributing to atrial and ventricular arrhythmias of different mechanisms. OBJECTIVE: The purpose of this study was to study whether/how chronic in vivo Ang II administration remodels IKs in atrial and ventricular myocytes. METHODS: We used the guinea pig (GP) model whose myocytes express robust IKs. GPs were implanted with minipumps containing Ang II or vehicle. Treatment continued for 4-6 weeks. We used patch clamp, immunofluorescence/confocal microscopy, and immunoblots to evaluate changes in IKs function and to explore the underlying mechanisms. RESULTS: We confirmed the pathologic state of the heart after chronic Ang II treatment. IKs density was increased in atrial myocytes but decreased in ventricular myocytes in Ang II- vs vehicle-treated animals. The former was correlated with an increase in KCNQ1/KCNE1 colocalization in myocyte periphery, whereas the latter was correlated with a decrease in KCNQ1 protein level. Interestingly, these changes in IKs were not translated into expected alterations in AP duration or plateau voltage, indicating that other currents were involved. In atrial myocytes from Ang II-treated animals, the L-type Ca channel current was increased, contributing to AP plateau elevation and AP duration prolongation. CONCLUSION: IKs is differentially modulated by chronic in vivo Ang II administration between atrial and ventricular myocytes. Other currents remodeled by Ang II treatment also contribute to changes in action potentials.


Subject(s)
Angiotensin II/administration & dosage , Heart Atria/physiopathology , Heart Ventricles/physiopathology , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Tachycardia, Ventricular/drug therapy , Animals , Cells, Cultured , Disease Models, Animal , Guinea Pigs , Heart Atria/metabolism , Heart Atria/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology , Vasoconstrictor Agents/administration & dosage
5.
Oncogene ; 37(40): 5416-5434, 2018 10.
Article in English | MEDLINE | ID: mdl-29867202

ABSTRACT

Tumor metastasis is the most common cause of cancer death. Elucidation of the mechanism of tumor metastasis is therefore important in the development of novel, effective anti-cancer therapies to reduce cancer mortality. Interaction between cancer cells and surrounding stromal cells in the tumor microenvironment is a key factor in tumor metastasis. Using a co-culture assay system with human prostate cancer LNCaP cells and primary human prostate stromal cells, we identified epithelial membrane protein 1 (EMP1) as a gene with elevated expression in the cancer cells. The orthotopic injection of LNCaP cells overexpressing EMP1 (EMP1-LNCaP cells) into the prostate of nude mice induced lymph node and lung metastases, while that of control LNCaP cells did not. EMP1-LNCaP cells had higher cell motility and Rac1 activity than control LNCaP cells. These results were also observed in other lines of cancer cells. We newly identified copine-III as an intracellular binding partner of EMP1. Knockdown of copine-III attenuated the increased cell motility and Rac1 activity in EMP1-LNCaP cells. Reduced cell motility and Rac1 activity following knockdown of copine-III in EMP1-LNCaP cells were recovered by re-expression of wild-type copine-III, but not of a copine-III mutant incapable of interacting with EMP1, suggesting the importance of the EMP1-copine-III interaction. Phosphorylated and activated Src and a Rac guanine nucleotide exchange factor Vav2 were found to be involved in the EMP1-induced enhancement of cell motility and Rac1 activation. Moreover, EMP1 was highly expressed in prostate cancer samples obtained from patients with higher Gleason score. These results demonstrate that upregulation of EMP1 significantly increases cancer cell migration that leads to tumor metastasis, suggesting that EMP1 may play an essential role as a positive regulator of tumor metastasis.


Subject(s)
Cell Movement , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Cell Surface/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line, Tumor , Disease Progression , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction , Stromal Cells/pathology , Up-Regulation
6.
Int J Mol Sci ; 19(4)2018 Apr 16.
Article in English | MEDLINE | ID: mdl-29659486

ABSTRACT

Vascular endothelial growth factors (VEGFs) include five molecules (VEGF-A, -B, -C, -D, and placental growth factor), and have various roles that crucially regulate cellular functions in many kinds of cells and tissues. Intracellular signal transduction induced by VEGFs has been extensively studied and is usually initiated by their binding to two classes of transmembrane receptors: receptor tyrosine kinase VEGF receptors (VEGF receptor-1, -2 and -3) and neuropilins (NRP1 and NRP2). In addition to many established results reported by other research groups, we have previously identified small G proteins, especially Ras homologue gene (Rho) and Ras-related protein (Rap), as important mediators of VEGF-A-stimulated signaling in cancer cells as well as endothelial cells. This review article describes the VEGF-A-induced signaling pathways underlying diverse cellular functions, including cell proliferation, migration, and angiogenesis, and the involvement of Rho, Rap, and their related molecules in these pathways.


Subject(s)
Vascular Endothelial Growth Factor A/metabolism , rap GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Humans , Models, Biological , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Circ J ; 81(12): 1862-1870, 2017 Nov 24.
Article in English | MEDLINE | ID: mdl-28659552

ABSTRACT

BACKGROUND: Pressure overload induces cardiac hypertrophy, which often ends in heart failure. Afadin is an adaptor protein that is ubiquitously expressed and, in the heart, it localizes at intercalated disks. The current study aimed to examine the afadin-mediated cardiac phenotype in mice exposed to different types of pressure overload: transverse aortic constriction (TAC) burden and angiotensin II (Ang II) stimulation.Methods and Results:Conditional knockout mice with selective deletion of afadin (afadin cKO) in cardiomyocytes were generated. TAC-operated and Ang II-infused mice at 4 weeks had a similar degree of pressure overload and cardiac hypertrophy in the heart. In afadin cKO mice, TAC operation caused progressive left ventricular dysfunction and heart failure, while Ang II infusion did not deteriorate cardiac function. Furthermore, TAC operation produced more fibrosis and apoptosis in the heart than Ang II infusion, and the expression of growth differentiation factor 15, which can promote apoptosis, in the afadin cKO heart was higher in TAC-operated mice than Ang II-infused ones. CONCLUSIONS: In the 2 pressure overload models, myocardial afadin is involved in mechanical stress-induced, but not pharmacological Ang II-related, compensated cardiac hypertrophy.


Subject(s)
Cardiomegaly/etiology , Microfilament Proteins/physiology , Pressure/adverse effects , Stress, Mechanical , Angiotensin II/pharmacology , Animals , Apoptosis , Fibrosis , Humans , Mice , Mice, Knockout , Microfilament Proteins/genetics , Myocardium/chemistry
8.
Sci Rep ; 7: 39335, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045017

ABSTRACT

Adhesive intercellular connections at cardiomyocyte intercalated disks (IDs) support contractile force and maintain structural integrity of the heart muscle. Disturbances of the proteins at IDs deteriorate cardiac function and morphology. An adaptor protein afadin, one of the components of adherens junctions, is expressed ubiquitously including IDs. At present, the precise role of afadin in cardiac physiology or disease is unknown. To explore this, we generated conditional knockout (cKO) mice with cardiomyocyte-targeted deletion of afadin. Afadin cKO mice were born according to the expected Mendelian ratio and have no detectable changes in cardiac phenotype. On the other hand, chronic pressure overload induced by transverse aortic constriction (TAC) caused systolic dysfunction, enhanced fibrogenesis and apoptosis in afadin cKO mice. Afadin deletion increased macrophage infiltration and monocyte chemoattractant protein-1 expression, and suppressed transforming growth factor (TGF) ß receptor signaling early after TAC procedure. Afadin also associated with TGFß receptor I at IDs. Pharmacological antagonist of TGFß receptor I (SB431542) augmented mononuclear infiltration and fibrosis in the hearts of TAC-operated control mice. In conclusion, afadin is a critical molecule for cardiac protection against chronic pressure overload. The beneficial effects are likely to be a result from modulation of TGFß receptor signaling pathways by afadin.


Subject(s)
Aorta/physiology , Heart Failure, Systolic/genetics , Intercellular Junctions/metabolism , Macrophages/immunology , Microfilament Proteins/metabolism , Myocardium/pathology , Myocytes, Cardiac/physiology , Animals , Apoptosis/genetics , Cell Movement , Chemokine CCL2/metabolism , Constriction, Pathologic , Fibrosis , Mice , Mice, Knockout , Microfilament Proteins/genetics , Myocardial Contraction , Signal Transduction , Transforming Growth Factor beta/metabolism
9.
Hypertension ; 68(3): 630-41, 2016 09.
Article in English | MEDLINE | ID: mdl-27456521

ABSTRACT

Dipeptidyl peptidase III (DPP III) cleaves dipeptide residues from the N terminus of polypeptides ranging from 3 to 10 amino acids in length and is implicated in pathophysiological processes through the breakdown of certain oligopeptides or their fragments. In this study, we newly identified the biochemical properties of DPP III for angiotensin II (Ang II), which consists of 8 amino acids. DPP III quickly and effectively digested Ang II with Km = 3.7×10(-6) mol/L. In the in vivo experiments, DPP III remarkably reduced blood pressure in Ang II-infused hypertensive mice without alteration of heart rate. DPP III did not affect hemodynamics in noradrenalin-induced hypertensive mice or normotensive mice, suggesting specificity for Ang II. When DPP III was intravenously injected every other day for 4 weeks after Ang II osmotic minipump implantation in mice, Ang II-induced cardiac fibrosis and hypertrophy were significantly attenuated. This DPP III effect was at least similar to that caused by an angiotensin receptor blocker candesartan. Furthermore, administration of DPP III dramatically reduced the increase in urine albumin excretion and kidney injury and inflammation markers caused by Ang II infusion. Both DPP III and candesartan administration showed slight additive inhibition in the albumin excretion. These results reveal a novel potential use of DPP III in the treatment of hypertension and its protective effects on hypertension-sensitive organs, such as the heart and kidneys.


Subject(s)
Angiotensin II/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/pharmacology , Hemodynamics/drug effects , Hypertension/drug therapy , Analysis of Variance , Animals , Blood Pressure/drug effects , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Disease Models, Animal , Glomerular Filtration Rate/drug effects , Heart Rate/drug effects , Hypertension/enzymology , Mice , Mice, Inbred ICR , Random Allocation , Role , Sensitivity and Specificity , Treatment Outcome
10.
Biomed Res Int ; 2015: 314178, 2015.
Article in English | MEDLINE | ID: mdl-25883953

ABSTRACT

Vasculature is present in all tissues and therefore is indispensable for development, biology, and pathology of multicellular organisms. Endothelial cells guarantee proper function of the vessels and are the original component in angiogenesis. Morphogenesis of the vascular system utilizes processes like cell adhesion, motility, proliferation, and survival that are closely related to the dynamics of actin filaments and actin-tethered adhesion complexes. Here we review involvement of actin cytoskeleton-associated junctional molecules of endothelial cells in angiogenesis and lymphangiogenesis. Particularly, we focus on F-actin binding protein afadin, an adaptor protein involved in broad range of signaling mechanisms. Afadin mediates the pathways of vascular endothelial growth factor- (VEGF-) and sphingosine 1-phosphate-triggered angiogenesis and is essential for embryonic development of lymph vessels in mice. We propose that targeting actin-tethered junctional molecules, including afadin, may present a new approach to angiogenic therapy that in combination with today used medications like VEGF inhibitors will benefit against development of pathological angiogenesis.


Subject(s)
Actins/physiology , Gene Expression Regulation , Lymphangiogenesis , Microfilament Proteins/physiology , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Catenins/metabolism , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Humans , Integrins/metabolism , Lymphatic Vessels/pathology , Lysophospholipids/metabolism , Mice , Mice, Knockout , Neovascularization, Physiologic , Sphingosine/analogs & derivatives , Sphingosine/metabolism , beta Catenin/metabolism , Delta Catenin
11.
Hypertens Res ; 38(4): 244-51, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25427682

ABSTRACT

Zn-α2-glycoprotein (ZAG) (molecular weight=41 kDa) is one component in the α2 fraction of human plasma, and is reported to be associated with several diseases, such as cancers and metabolic syndromes. ZAG is also considered to be an important modulator of lipid metabolism. However, little is known about the correlation of serum ZAG levels with indicators of metabolic syndrome. Serum ZAG concentrations analyzed by enzyme-linked immunoassay were positively correlated with systolic and diastolic blood pressure in 326 subjects (236 males and 90 females) aged 17-79 years who had an annual health examination. By luciferase reporter and electrophoretic mobility shift assays, the core promoter region to regulate the ZAG gene expression was found to exist between -110 and -101. The transcription factor Sp1 interacted with this region, and Sp1 knockdown experiments showed that Sp1 critically regulated ZAG expression. Furthermore, ZAG increased the active form of RhoA, which was determined by pull-down assay. Increased serum ZAG concentrations induced, at least partly, by Sp1 may cause an increase in vascular tone through the activation of RhoA and contribute to elevated blood pressure.


Subject(s)
Blood Pressure/physiology , Seminal Plasma Proteins/blood , Adolescent , Adult , Aged , Animals , Blood Pressure/genetics , Cell Line , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Hypertension/genetics , Hypertension/physiopathology , Male , Metabolic Syndrome/blood , Middle Aged , Muscle Tonus/genetics , Promoter Regions, Genetic/genetics , Rats , Seminal Plasma Proteins/biosynthesis , Seminal Plasma Proteins/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/physiology , Young Adult , Zn-Alpha-2-Glycoprotein , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
12.
J Biol Chem ; 288(49): 35358-71, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24142691

ABSTRACT

Our goals are to simultaneously determine the three-dimensional distribution patterns of KCNQ1 and KCNE1 in cardiac myocytes and to study the mechanism and functional implications for variations in KCNQ1/KCNE1 colocalization in myocytes. We monitored the distribution patterns of KCNQ1, KCNE1, and markers for subcellular compartments/organelles using immunofluorescence/confocal microscopy and confirmed the findings in ventricular myocytes by directly observing fluorescently tagged KCNQ1-GFP and KCNE1-dsRed expressed in these cells. We also monitored the effects of stress on KCNQ1-GFP and endoplasmic reticulum (ER) remodeling during live cell imaging. The data showed that 1) KCNE1 maintained a stable cell surface localization, whereas KCNQ1 exhibited variations in the cytosolic compartment (striations versus vesicles) and the degree of presence on the cell surface; 2) the degree of cell surface KCNQ1/KCNE1 colocalization was positively correlated with slow delayed rectifier (IKs) current density; 3) KCNQ1 and calnexin (an ER marker) shared a cytosolic compartment; and 4) in response to stress ([Ca(2+)]i elevation, oxidative overload, or AT1R stimulation), KCNQ1 exited the cytosolic compartment and trafficked to the cell periphery in vesicles. This was accompanied by partial ER fragmentation. We conclude that the cellular milieu regulates KCNQ1 distribution in cardiac myocytes and that stressful conditions can increase IKs by inducing KCNQ1 movement to the cell surface. This represents a hitherto unrecognized mechanism by which IKs fulfills its function as a repolarization reserve in ventricular myocytes.


Subject(s)
Calcium/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , KCNQ1 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Animals , COS Cells , Calnexin/metabolism , Cell Membrane/metabolism , Chlorocebus aethiops , Cytosol/metabolism , Fluorescence Resonance Energy Transfer , Guinea Pigs , Heart Atria/cytology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , KCNQ1 Potassium Channel/genetics , Oxidative Stress , Potassium Channels, Voltage-Gated/genetics , Protein Transport , Rats , Receptor, Angiotensin, Type 1/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
PLoS One ; 8(6): e68134, 2013.
Article in English | MEDLINE | ID: mdl-23840823

ABSTRACT

Afadin is an intracellular binding partner of nectins, cell-cell adhesion molecules, and plays important roles in the formation of cell-cell junctions. Afadin-knockout mice show early embryonic lethality, therefore little is known about the function of afadin during organ development. In this study, we generated mice lacking afadin expression in endothelial cells, and found that the majority of these mice were embryonically lethal as a result of severe subcutaneous edema. Defects in the lymphatic vessels of the skin were observed, although the morphology in the blood vessels was almost normal. Severe disruption of VE-cadherin-mediated cell-cell junctions occurred only in lymphatic endothelial cells, but not in blood endothelial cells. Knockout of afadin did not affect the differentiation and proliferation of lymphatic endothelial cells. Using in vitro assays with blood and lymphatic microvascular endothelial cells (BMVECs and LMVECs, respectively), knockdown of afadin caused elongated cell shapes and disruption of cell-cell junctions among LMVECs, but not BMVECs. In afadin-knockdown LMVECs, enhanced F-actin bundles at the cell periphery and reduced VE-cadherin immunostaining were found, and activation of RhoA was strongly increased compared with that in afadin-knockdown BMVECs. Conversely, inhibition of RhoA activation in afadin-knockdown LMVECs restored the cell morphology. These results indicate that afadin has different effects on blood and lymphatic endothelial cells by controlling the levels of RhoA activation, which may critically regulate the lymphangiogenesis of mouse embryos.


Subject(s)
Embryo, Mammalian/cytology , Embryonic Development , Endothelium, Vascular/cytology , Lymphangiogenesis/physiology , Microfilament Proteins/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Cell Adhesion/physiology , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/metabolism , Endothelium, Vascular/metabolism , Humans , Intercellular Junctions/physiology , Mice , Mice, Knockout , Mice, Transgenic
14.
J Gen Physiol ; 140(6): 653-69, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23183700

ABSTRACT

KCNE1 associates with KCNQ1 to increase its current amplitude and slow the activation gating process, creating the slow delayed rectifier channel that functions as a "repolarization reserve" in human heart. The transmembrane domain (TMD) of KCNE1 plays a key role in modulating KCNQ1 pore conductance and gating kinetics, and the extracellular juxtamembrane (EJM) region plays a modulatory role by interacting with the extracellular surface of KCNQ1. KCNE2 is also expressed in human heart and can associate with KCNQ1 to suppress its current amplitude and slow the deactivation gating process. KCNE1 and KCNE2 share the transmembrane topology and a high degree of sequence homology in TMD and surrounding regions. The structural basis for their distinctly different effects on KCNQ1 is not clear. To address this question, we apply cysteine (Cys) scanning mutagenesis to TMDs and EJMs of KCNE1 and KCNE2. We analyze the patterns of functional perturbation to identify high impact positions, and probe disulfide formation between engineered Cys side chains on KCNE subunits and native Cys on KCNQ1. We also use methanethiosulfonate reagents to probe the relationship between EJMs of KCNE subunits and KCNQ1. Our data suggest that the TMDs of both KCNE subunits are at about the same location but interact differently with KCNQ1. In particular, the much closer contact of KCNE2 TMD with KCNQ1, relative to that of KCNE1, is expected to impact the allosteric modulation of KCNQ1 pore conductance and may explain their differential effects on the KCNQ1 current amplitude. KCNE1 and KCNE2 also differ in the relationship between their EJMs and KCNQ1. Although the EJM of KCNE1 makes intimate contacts with KCNQ1, there appears to be a crevice between KCNQ1 and KCNE2. This putative crevice may perturb the electrical field around the voltage-sensing domain of KCNQ1, contributing to the differential effects of KCNE2 versus KCNE1 on KCNQ1 gating kinetics.


Subject(s)
Ion Channel Gating/physiology , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Cysteine/genetics , Cysteine/metabolism , Ion Channel Gating/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis , Oocytes/metabolism
15.
Am J Physiol Heart Circ Physiol ; 302(4): H910-22, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22180649

ABSTRACT

KCNE2 functions as an auxiliary subunit in voltage-gated K and HCN channels in the heart. Genetic variations in KCNE2 have been linked to long QT syndrome. The underlying mechanisms are not entirely clear. One of the issues is whether KCNE2 protein is expressed in ventricles. We use adenovirus-mediated genetic manipulations of adult cardiac myocytes to validate two antibodies (termed Ab1 and Ab2) for their ability to detect native KCNE2 in the heart. Ab1 faithfully detects native KCNE2 proteins in spontaneously hypertensive rat and guinea pig hearts. In both cases, KCNE2 protein is more abundant in ventricles than in atria. In both ventricular and atrial myocytes, KCNE2 protein is preferentially distributed on the cell surface. Ab1 can detect a prominent KCNE2 band in human ventricular muscle from nonfailing hearts. The band intensity is much fainter in atria and in failing ventricles. Ab2 specifically detects S98 phosphorylated KCNE2. Through exploring the functional significance of S98 phosphorylation, we uncover a novel mechanism by which KCNE2 modulates the human ether-a-go-go related gene (hERG) current amplitude: by accelerating hERG protein degradation and thus reducing the hERG protein level on the cell surface. S98 phosphorylation appears to be required for this modulation, so that S98 dephosphorylation leads to an increase in hERG/rapid delayed rectifier current amplitude. Our data confirm that KCNE2 protein is expressed in the ventricles of human and animal models. Furthermore, KCNE2 can modulate its partner channel function not only by altering channel conductance and/or gating kinetics, but also by affecting protein stability.


Subject(s)
Heart Atria/metabolism , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Proteolysis , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dogs , Female , Guinea Pigs , Heart Atria/cytology , Heart Ventricles/cytology , Humans , Male , Models, Animal , Molecular Sequence Data , Myocytes, Cardiac/cytology , Patch-Clamp Techniques , Phosphorylation , Potassium Channels, Voltage-Gated/analysis , Rats , Rats, Inbred SHR , Shal Potassium Channels , Transcriptional Regulator ERG
16.
Circ Arrhythm Electrophysiol ; 4(3): 352-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21493962

ABSTRACT

BACKGROUND: Brugada syndrome (BrS) has a significantly higher incidence among the male sex. Among genes coding ion channels and their modulatory proteins, KCNE5 (KCNE1L) is located in the X chromosome and encodes an auxiliary ß-subunit for K channels. KCNE5 has been shown to modify the transient outward current (I(to)), which plays a key role in determining the repolarization process in the myocardium. This study investigated whether KCNE5 mutations could be responsible for BrS and other idiopathic ventricular fibrillation (IVF). METHODS AND RESULTS: In 205 Japanese patients with BrS or IVF who tested negative for SCN5A mutation, we conducted a genetic screen for KCNE5 variants. We identified 2 novel KCNE5 variants: p.Y81H in 3 probands and p.[D92E;E93X] in 1 proband from 4 unrelated families. Y81H was identified in 1 man and 2 women; D92E;E93X was found in a 59-year-old man. All probands received implantable cardioverter-defibrillators. Functional consequences of the KCNE5 variants were determined through biophysical assay using cotransfection with KCND3 or KCNQ1. In the experiments with KCND3, which encodes Kv4.3, I(to) was significantly increased for both KCNE5 variants compared to wild type. In contrast, there were no significant changes in current properties reconstructed by KCNQ1+ wild type KCNE5 and the 2 variants. With the simulation model, both variants demonstrated notch-and-dome or loss-of-dome patterns. CONCLUSIONS: KCNE5 modulates I(to), and its novel variants appeared to cause IVF, especially BrS, in male patients through gain-of-function effects on I(to). Screening for KCNE5 variants is relevant for BrS or IVF.


Subject(s)
Brugada Syndrome/genetics , DNA/genetics , Mutation , Myocardium/metabolism , Potassium Channels, Voltage-Gated/genetics , Ventricular Fibrillation/genetics , Adult , Brugada Syndrome/metabolism , Brugada Syndrome/physiopathology , Cell Line , Computer Simulation , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Myocardium/pathology , Patch-Clamp Techniques , Pedigree , Polymerase Chain Reaction , Potassium Channels, Voltage-Gated/metabolism , Ventricular Fibrillation/metabolism , Ventricular Fibrillation/physiopathology
17.
J Membr Biol ; 235(2): 73-87, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20490473

ABSTRACT

HL-1 is the adult murine cardiac cell line that can be passaged repeatedly in vitro without losing differentiated phenotype. The present study was designed to characterize the rapidly activating delayed rectifier potassium current, I (Kr), endogenously expressed in HL-1 cells using the whole-cell patch-clamp technique. In the presence of nisoldipine, depolarizing voltage steps applied from a holding potential of -50 mV evoked the time-dependent outward current, followed by slowly decaying outward tail current upon return to the holding potential. The amplitude of the current increased with depolarizations up to 0 mV but then progressively decreased with further depolarizations. The time-dependent outward current as well as the tail current were highly sensitive to block by E-4031 and dofetilide (IC(50) of 21.1 and 15.1 nM, respectively) and almost totally abolished by micromolar concentrations of each drug, suggesting that most of the outward current in HL-1 cells was attributable to I (Kr). The magnitude of I (Kr) available from HL-1 cells (18.1 +/- 1.5 pA pF(-1)) was sufficient for reliable measurements of various gating parameters. RT-PCR and Western blot analysis revealed the expression of alternatively spliced forms of mouse ether-a-go-go-related genes (mERG1), the full-length mERG1a and the N-terminally truncated mERG1b isoforms. Knockdown of mERG1 transcripts with small interfering RNA (siRNA) dramatically reduced I (Kr) amplitude, confirming the molecular link of mERG1 and I (Kr) in HL-1 cells. These findings demonstrate that HL-1 cells possess I (Kr) with properties comparable to those in native cardiac I (Kr) and provide an experimental model suitable for studies of I (Kr) channels.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Myocytes, Cardiac/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Blotting, Western , Cell Line , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Mice , Myocytes, Cardiac/drug effects , Nisoldipine/pharmacology , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium/metabolism , Pyridines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
18.
Heart Rhythm ; 6(7): 1038-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19419905

ABSTRACT

BACKGROUND: KCNH2 gene mutations disrupting rapid component of I(K) (I(Kr)) underlie type 2 congenital long QT syndrome (LQT2). Startled auditory stimuli are specific symptomatic triggers in LQT2, thus suggesting fast arrhythmogenic mechanism. OBJECTIVE: We investigated acute alpha(1A)- and cyclic adenosine monophosphate (cAMP)-related beta-adrenergic modulation of I(Kr) in HL-1 cardiomyocytes, wild type (WT)- and 2 LQT2-associated mutant Kv11.1 channels (Y43D- and K595E-Kv11.1) reconstituted in Chinese hamster ovary (CHO) cells. METHODS: I(Kr) and Kv11.1 currents were recorded using the whole-cell patch-clamp technique and confocal microscopy of HL-1 cardiomyocytes transfected with green fluorescent protein (GFP)-tagged pleckstrin homology domain of phospholipase C-delta(1) visualized fluctuations of membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)) content. RESULTS: In HL-1 cardiomyocytes expressing human alpha(1A)-adrenoceptor, superfusion with phenylephrine significantly reduced I(Kr) amplitude, shifted current activation to more positive potentials, and accelerated kinetics of deactivation. Confocal images showed a decline of membrane PIP(2) content during phenylephrine exposure. Simultaneous application of adenylyl cyclase activator forskolin and phosphodiesterase inhibitor 3-isobutyl-1-methylxantine (IBMX) shifted I(Kr) activation to more negative potentials and decreased tail current amplitudes after depolarizations between +10 and +50 mV. In CHO cells, alpha(1A)-adrenoceptor activation downregulated WT-Kv11.1 channels and forskolin/IBMX produced a dual effect. Expressed alone, the Y43D-Kv11.1 or K595E-Kv11.1 channel had no measurable function. However, co-expression of WT-Kv11.1 and each mutant protein evoked currents with loss-of-function alterations but identical to WT-Kv11.1 alpha(1A)- and forskolin/IBMX-induced regulation. CONCLUSION: Acute adrenergic regulation of at least 2 Kv11.1 mutant channels is preserved as in WT-Kv11.1 and native I(Kr). Suppression of alpha(1A)-adrenoceptor-related transduction might have therapeutic implications in some cases of LQT2.


Subject(s)
Long QT Syndrome/physiopathology , Myocytes, Cardiac/physiology , Potassium Channels, Voltage-Gated/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Adult , Animals , Cells, Cultured , Cricetinae , Disease Models, Animal , Electrophysiologic Techniques, Cardiac , Female , Humans , Long QT Syndrome/congenital , Long QT Syndrome/genetics , Microscopy, Confocal , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/genetics
19.
Pflugers Arch ; 458(5): 837-49, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19404674

ABSTRACT

A repolarizing conduction in the heart augmented by hyposmotic or mechanically induced membrane stretch is the slow component of delayed rectifier K(+) current (I (Ks)). I (Ks) upregulation is recognized as a factor promoting appearance of atrial fibrillation (AF) since gain-of-function mutations of the channel genes have been detected in congenital AF. Mechanical stretch activates angiotensin II type 1 (AT(1)) receptor in the absence of its physiological ligand angiotensin II. We investigated the functional role of AT(1) receptor in I (Ks) enhancement in hyposmotically challenged guinea pig atrial myocytes using the whole-cell patch-clamp method. In atrial myocytes exposed to hyposmotic solution with osmolality decreased to 70% of the physiological level, I (Ks) was enhanced by 84.1%, the duration of action potential at 90% repolarization (APD(90)) was decreased by 16.8%, and resting membrane potential was depolarized (+4.9 mV). The hyposmotic-induced effects on I (Ks) and APD(90) were significantly attenuated by specific AT(1) receptor antagonist candesartan (1 and 5 muM). Pretreatment of atrial myocytes with protein tyrosine kinase inhibitors tyrphostin A23 and A25 suppressed but the presence of tyrosine phosphatase inhibitor orthovanadate augmented hyposmotic stimulation of I (Ks). The above results implicate AT(1) receptor and tyrosine kinases in the hyposmotic modulation of atrial I (Ks) and suggest acute antiarrhythmic properties of AT(1) antagonists in the settings of stretch-related atrial tachyarrhythmias.


Subject(s)
Electrophysiological Phenomena/physiology , Heart Atria/metabolism , Potassium/metabolism , Receptor, Angiotensin, Type 1/physiology , Action Potentials/drug effects , Action Potentials/physiology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Cell Shape/drug effects , Cell Shape/physiology , Electrophysiological Phenomena/drug effects , Enzyme Inhibitors/pharmacology , Guinea Pigs , Heart Atria/cytology , Hypotonic Solutions/pharmacology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Osmolar Concentration , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Receptor, Angiotensin, Type 1/immunology , Signal Transduction/drug effects , Signal Transduction/physiology , Tetrazoles/pharmacology
20.
Hum Mutat ; 30(4): 557-63, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19306396

ABSTRACT

Long QT syndrome (LQTS) is an inherited disease involving mutations in the genes encoding a number of cardiac ion channels and a membrane adaptor protein. Among the genes that are responsible for LQTS, KCNE1 and KCNE2 are members of the KCNE family of genes, and function as ancillary subunits of Kv channels. The third KCNE gene, KCNE3, is expressed in cardiac myocytes and interacts with KCNQ1 to change the channel properties. However, KCNE3 has never been linked to LQTS. To investigate the association between KCNE3 and LQTS, we conducted a genetic screening of KCNE3 mutations and single nucleotide polymorphisms (SNPs) in 485 Japanese LQTS probands using DHPLC-WAVE system and direct sequencing. Consequently, we identified two KCNE3 missense mutations, located in the N- and C-terminal domains. The functional effects of these mutations were examined by heterologous expression systems using CHO cells stably expressing KCNQ1. One mutation, p.R99lambdaH was identified in a 76-year-old woman who suffered torsades de pointes (TdP) after administration of disopyramide. Another mutation, p.T4A was identified in a 16-year-old boy and 67-year-old woman. Although the boy carried another KCNH2 mutation, he was asymptomatic. On the other hand, the woman suffered from hypokalemia-induced TdP. In a series of electrophysiological analyses, the KCNQ1(Q1)+KCNE3(E3)-R99lambdaH channel significantly reduced outward current compared to Q1+E3-WT, though the current density of the Q1+E3-T4A channel displayed no statistical significance. This is the first report of KCNE3 mutations associated with LQTS. Screening for variants in the KCNE3 gene is of clinical importance for LQTS patients.


Subject(s)
Long QT Syndrome/genetics , Mutation, Missense , Potassium Channels, Voltage-Gated/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Female , Humans , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Long QT Syndrome/physiopathology , Male , Membrane Potentials , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/physiology , Protein Binding , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...