Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Heart Rhythm ; 21(5): 622-629, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38280622

RESUMO

BACKGROUND: More than 50% of patients with atrial fibrillation (AF) suffer from sleep disordered breathing (SDB). Obstructive respiratory events contribute to a transient, vagally mediated atrial arrhythmogenic substrate, which is resistant to most available antiarrhythmic drugs. OBJECTIVE: The purpose of this study was to investigate the effect of pharmacologic inhibition of the G-protein-gated acetylcholine-regulated potassium current (IK,ACh) with and without acute autonomic nervous system activation by nicotine in a pig model for obstructive respiratory events. METHODS: In 21 pigs, SDB was simulated by applying an intermittent negative upper airway pressure (INAP). AF inducibility and atrial effective refractory periods (aERPs) were determined before and during INAP by an S1S2 atrial pacing-protocol. Pigs were randomized into 3 groups-group 1: vehicle (n = 4); group 2: XAF-1407 (IK,ACh inhibitor) (n = 7); and group 3: nicotine followed by XAF-1407 (n = 10). RESULTS: In group 1, INAP shortened aERP (ΔaERP -42.6 ms; P = .004) and transiently increased AF inducibility from 0% to 31%. In group 2, XAF-1407 prolonged aERP by 25.2 ms (P = .005) during normal breathing and prevented INAP-induced aERP shortening (ΔaERP -3.6 ms; P = .3) and AF inducibility. In group 3, INAP transiently shortened aERP during nicotine perfusion (ΔaERP -33.6 ms; P = .004) and increased AF inducibility up to 61%, which both were prevented by XAF-1407. CONCLUSION: Simulated obstructive respiratory events transiently shorten aERP and increase AF inducibility, which can be prevented by the IK,ACh-inhibitor XAF-1407. XAF-1407 also prevents these arrhythmogenic changes induced by obstructive respiratory events during nicotine perfusion. Whether IK,ACh channels represent a target for SDB-related AF in humans warrants further study.


Assuntos
Fibrilação Atrial , Modelos Animais de Doenças , Animais , Suínos , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/etiologia , Fibrilação Atrial/prevenção & controle , Átrios do Coração/fisiopatologia , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/metabolismo , Acetilcolina/farmacologia , Nicotina/farmacologia , Síndromes da Apneia do Sono/fisiopatologia , Síndromes da Apneia do Sono/complicações
2.
Echocardiography ; 40(7): 695-702, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37335308

RESUMO

BACKGROUND: Atrial fibrillation (AF) is the most common cardiac arrhythmia following coronary artery bypass grafting (CABG). We hypothesized that measures of left atrial (LA) function would be useful in predicting AF in patients undergoing CABG. METHODS AND RESULTS: In the study, 611 patients were included after CABG. All patients had echocardiograms performed preoperatively and LA functional measurements were assessed. These measurements were LA maximum volume index (LAVmax), LA minimum volume index (LAVmin) and LA emptying fraction (LAEF). The endpoint was AF occurring >14 days after surgery. During the follow-up period of a median of 3.7 years, 52 (9%) developed AF. The mean age was 67 years, 84% were male and the average left ventricle ejection fraction was 50%. Patients who developed AF had a lower CCS class and lower LAEF (40 vs. 45%), otherwise no clinical differences were observed between outcome groups. No functional LA measurements were significant predictors of AF in the whole CABG population. However, in patients with normal-sized LA (n = 532, events: 49), both LAEF and LAVmin were univariable predictors of AF. When the functional measurements were adjusted for the CHADS2 score, both LAVmin (HR = 1.07 [1.01-1.13], p = .014) and LAEF (HR: 1.02 [1.00-1.03], p = .023), remained significant predictors. CONCLUSION: No echocardiographic measurements were significant predictors of AF after CABG. In patients with a normal LA size, LAVmin as well as LAEF were significant predictors of AF.


Assuntos
Apêndice Atrial , Fibrilação Atrial , Humanos , Masculino , Idoso , Feminino , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/etiologia , Fibrilação Atrial/epidemiologia , Fatores de Risco , Átrios do Coração , Ponte de Artéria Coronária/efeitos adversos
3.
Nano Lett ; 22(12): 4758-4764, 2022 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-35679577

RESUMO

Freestanding oxide membranes constitute an intriguing material platform for new functionalities and allow integration of oxide electronics with technologically important platforms such as silicon. Sambri et al. recently reported a method to fabricate freestanding LaAlO3/SrTiO3 (LAO/STO) membranes by spalling of strained heterostructures. Here, we first develop a scheme for the high-yield fabrication of membrane devices on silicon. Second, we show that the membranes exhibit metallic conductivity and a superconducting phase below ∼200 mK. Using anisotropic magnetotransport we extract the superconducting phase coherence length ξ ≈ 36-80 nm and establish an upper bound on the thickness of the superconducting electron gas d ≈ 17-33 nm, thus confirming its two-dimensional character. Finally, we show that the critical current can be modulated using a silicon-based backgate. The ability to form superconducting nanostructures of LAO/STO membranes, with electronic properties similar to those of the bulk counterpart, opens opportunities for integrating oxide nanoelectronics with silicon-based architectures.

4.
5.
J Gen Physiol ; 153(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33533890

RESUMO

The dynein motor protein transports proteins away from the cell membrane along the microtubule network. Recently, we found the microtubule network was important for regulating the membrane abundance of voltage-gated Kv7.4 potassium channels in vascular smooth muscle. Here, we aimed to investigate the influence of dynein on the microtubule-dependent internalization of the Kv7.4 channel. Patch-clamp recordings from HEK293B cells showed Kv7.4 currents were increased after inhibiting dynein function with ciliobrevin D or by coexpressing p50/dynamitin, which specifically interferes with dynein motor function. Mutation of a dynein-binding site in the Kv7.4 C terminus increased the Kv7.4 current and prevented p50 interference. Structured illumination microscopy, proximity ligation assays, and coimmunoprecipitation showed colocalization of Kv7.4 and dynein in mesenteric artery myocytes. Ciliobrevin D enhanced mesenteric artery relaxation to activators of Kv7.2-Kv7.5 channels and increased membrane abundance of Kv7.4 protein in isolated smooth muscle cells and HEK293B cells. Ciliobrevin D failed to enhance the negligible S-1-mediated relaxations after morpholino-mediated knockdown of Kv7.4. Mass spectrometry revealed an interaction of dynein with caveolin-1, confirmed using proximity ligation and coimmunoprecipitation assays, which also provided evidence for interaction of caveolin-1 with Kv7.4, confirming that Kv7.4 channels are localized to caveolae in mesenteric artery myocytes. Lastly, cholesterol depletion reduced the interaction of Kv7.4 with caveolin-1 and dynein while increasing the overall membrane expression of Kv7.4, although it attenuated the Kv7.4 current in oocytes and interfered with the action of ciliobrevin D and channel activators in arterial segments. Overall, this study shows that dynein can traffic Kv7.4 channels in vascular smooth muscle in a mechanism dependent on cholesterol-rich caveolae.


Assuntos
Dineínas , Canais de Potássio KCNQ , Membrana Celular , Músculo Liso Vascular , Miócitos de Músculo Liso
6.
Equine Vet J ; 53(1): 186-193, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32285961

RESUMO

BACKGROUND: Three-dimensional electroanatomical mapping is of potential interest in equine cardiology to identify arrhythmia mechanisms, characterise electroanatomical substrates and guide ablation strategies. OBJECTIVES: To describe three-dimensional electroanatomical mapping in standing horses. STUDY DESIGN: Research methodology, proof of concept study. METHODS: Four Standardbred horses (2 geldings, 2 mares, median age 4.5 [4-9] years, mean bodyweight 485 [440-550] kg) were sedated and placed in stocks. Via the jugular vein, a high-density multipolar grid catheter (Advisor™ HD Grid Mapping Catheter with EnSite VelocityTM, Abbott Medical) was used for endocardial mapping of the right atrium. The P-wave on the surface ECG was used as a timing reference for simultaneous local activation time- and bipolar voltage-mapping. For a positional reference a 10-pole catheter (Abbott Medical) was placed in the caudal vena cava. RESULTS: Endocardial right atrial mapping guided by the three-dimensional mapping system and local electrograms was successfully performed in all four horses. A median of 32719 [25499-65078] points, covering the entire right atrium, were collected. Three-dimensional electroanatomical mapping provided detailed information about activation patterns and electrogram-characteristics of the sinoatrial node, intervenous tubercle and cavotricuspid isthmus. Additionally, transvenous biopsy forceps connected to the mapping system were visualised on screen to guide biopsy collection. MAIN LIMITATIONS: The feasibility of electroanatomical mapping for the left atrium and in larger breeds requires further study. CONCLUSIONS: High-density three-dimensional electroanatomical mapping of the right atrium is feasible in the standing horse.


Assuntos
Ablação por Cateter , Doenças dos Cavalos , Animais , Arritmias Cardíacas/veterinária , Ablação por Cateter/veterinária , Catéteres , Feminino , Átrios do Coração , Cavalos , Masculino
7.
Front Physiol ; 11: 493, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32595514

RESUMO

BACKGROUND: Adenosine leads to atrial action potential (AP) shortening through activation of adenosine 1 receptors (A1-R) and subsequent opening of G-protein-coupled inwardly rectifying K+ channels. Extracellular production of adenosine is drastically increased during stress and ischemia. OBJECTIVE: The aim of this study was to address whether the pharmacological blockade of endogenous production of adenosine and of its signaling prevents atrial fibrillation (AF). METHODS: The role of A1-R activation on atrial action potential duration, refractoriness, and AF vulnerability was investigated in rat isolated beating heart preparations (Langendorff) with an A1-R agonist [2-chloro-N 6-cyclopentyladenosine (CCPA), 50 nM] and antagonist [1-butyl-3-(3-hydroxypropyl)-8-(3-noradamantyl)xanthine (PSB36), 40 nM]. Furthermore, to interfere with the endogenous adenosine release, the ecto-5'-nucleotidase (CD73) inhibitor was applied [5'-(α,ß-methylene) diphosphate sodium salt (AMPCP), 500 µM]. Isolated trabeculae from human right atrial appendages (hRAAs) were used for comparison. RESULTS: As expected, CCPA shortened AP duration at 90% of repolarization (APD90) and effective refractory period (ERP) in rat atria. PSB36 prolonged APD90 and ERP in rat atria, and CD73 inhibition with AMPCP prolonged ERP in rats, confirming that endogenously produced amount of adenosine is sufficiently high to alter atrial electrophysiology. In human atrial appendages, CCPA shortened APD90, while PSB36 prolonged it. Rat hearts treated with CCPA are prone to AF. In contrast, PSB36 and AMPCP prevented AF events and reduced AF duration (vehicle, 11.5 ± 2.6 s; CCPA, 40.6 ± 16.1 s; PSB36, 6.5 ± 3.7 s; AMPCP, 3.0 ± 1.4 s; P < 0.0001). CONCLUSION: A1-R activation by intrinsic adenosine release alters atrial electrophysiology and promotes AF. Inhibition of adenosine pathway protects atria from arrhythmic events.

8.
Nat Commun ; 8(1): 1289, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-29097701

RESUMO

Familial growth hormone deficiency provides an opportunity to identify new genetic causes of short stature. Here we combine linkage analysis with whole-genome resequencing in patients with growth hormone deficiency and maternally inherited gingival fibromatosis. We report that patients from three unrelated families harbor either of two missense mutations, c.347G>T p.(Arg116Leu) or c.1106C>T p.(Pro369Leu), in KCNQ1, a gene previously implicated in the long QT interval syndrome. Kcnq1 is expressed in hypothalamic GHRH neurons and pituitary somatotropes. Co-expressing KCNQ1 with the KCNE2 ß-subunit shows that both KCNQ1 mutants increase current levels in patch clamp analyses and are associated with reduced pituitary hormone secretion from AtT-20 cells. In conclusion, our results reveal a role for the KCNQ1 potassium channel in the regulation of human growth, and show that growth hormone deficiency associated with maternally inherited gingival fibromatosis is an allelic disorder with cardiac arrhythmia syndromes caused by KCNQ1 mutations.


Assuntos
Fibromatose Gengival/genética , Hormônio do Crescimento Humano/deficiência , Canal de Potássio KCNQ1/genética , Mutação de Sentido Incorreto , Adolescente , Hormônio Adrenocorticotrópico/metabolismo , Adulto , Alelos , Substituição de Aminoácidos , Animais , Arritmias Cardíacas/genética , Criança , Pré-Escolar , Feminino , Fibromatose Gengival/metabolismo , Humanos , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/metabolismo , Masculino , Herança Materna/genética , Camundongos , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Mapas de Interação de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Adulto Jovem
9.
Circulation ; 135(18): 1705-1719, 2017 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-28235848

RESUMO

BACKGROUND: Loss-of-function mutations in hERG (encoding the Kv11.1 voltage-gated potassium channel) cause long-QT syndrome type 2 (LQT2) because of prolonged cardiac repolarization. However, Kv11.1 is also present in pancreatic α and ß cells and intestinal L and K cells, secreting glucagon, insulin, and the incretins glucagon-like peptide-1 (GLP-1) and GIP (glucose-dependent insulinotropic polypeptide), respectively. These hormones are crucial for glucose regulation, and long-QT syndrome may cause disturbed glucose regulation. We measured secretion of these hormones and cardiac repolarization in response to glucose ingestion in LQT2 patients with functional mutations in hERG and matched healthy participants, testing the hypothesis that LQT2 patients have increased incretin and ß-cell function and decreased α-cell function, and thus lower glucose levels. METHODS: Eleven patients with LQT2 and 22 sex-, age-, and body mass index-matched control participants underwent a 6-hour 75-g oral glucose tolerance test with ECG recording and blood sampling for measurements of glucose, insulin, C-peptide, glucagon, GLP-1, and GIP. RESULTS: In comparison with matched control participants, LQT2 patients had 56% to 78% increased serum insulin, serum C-peptide, plasma GLP-1, and plasma GIP responses (P=0.03-0.001) and decreased plasma glucose levels after glucose ingestion (P=0.02) with more symptoms of hypoglycemia (P=0.04). Sixty-three percent of LQT2 patients developed hypoglycemic plasma glucose levels (<70 mg/dL) versus 36% control participants (P=0.16), and 18% patients developed serious hypoglycemia (<50 mg/dL) versus none of the controls. LQT2 patients had defective glucagon responses to low glucose, P=0.008. ß-Cell function (Insulin Secretion Sensitivity Index-2) was 2-fold higher in LQT2 patients than in controls (4398 [95% confidence interval, 2259-8562] versus 2156 [1961-3201], P=0.03). Pharmacological Kv11.1 blockade (dofetilide) in rats had similar effect, and small interfering RNA inhibition of hERG in ß and L cells increased insulin and GLP-1 secretion up to 50%. Glucose ingestion caused cardiac repolarization disturbances with increased QTc intervals in both patients and controls, but with a 122% greater increase in QTcF interval in LQT2 patients (P=0.004). CONCLUSIONS: Besides a prolonged cardiac repolarization phase, LQT2 patients display increased GLP-1, GIP, and insulin secretion and defective glucagon secretion, causing decreased plasma glucose and thus increased risk of hypoglycemia. Furthermore, glucose ingestion increased QT interval and aggravated the cardiac repolarization disturbances in LQT2 patients. CLINICAL TRIAL REGISTRATION: URL: http://clinicaltrials.gov. Unique identifier: NCT02775513.


Assuntos
Glicemia/metabolismo , Canal de Potássio ERG1/genética , Sistema de Condução Cardíaco/fisiopatologia , Hipoglicemia/etiologia , Incretinas/metabolismo , Ilhotas Pancreáticas/metabolismo , Síndrome do QT Longo/genética , Mutação , Potenciais de Ação , Adulto , Animais , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Peptídeo C/sangue , Estudos de Casos e Controles , Linhagem Celular Tumoral , Canal de Potássio ERG1/antagonistas & inibidores , Canal de Potássio ERG1/metabolismo , Eletrocardiografia , Feminino , Polipeptídeo Inibidor Gástrico/sangue , Predisposição Genética para Doença , Glucagon/sangue , Peptídeo 1 Semelhante ao Glucagon/sangue , Teste de Tolerância a Glucose , Sistema de Condução Cardíaco/metabolismo , Humanos , Hipoglicemia/sangue , Hipoglicemia/diagnóstico , Hipoglicemia/fisiopatologia , Insulina/sangue , Síndrome do QT Longo/sangue , Síndrome do QT Longo/diagnóstico , Síndrome do QT Longo/fisiopatologia , Masculino , Camundongos , Pessoa de Meia-Idade , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Interferência de RNA , Ratos Wistar , Fatores de Tempo , Transfecção
10.
Scand J Clin Lab Invest ; 75(8): 699-709, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26403377

RESUMO

BACKGROUND: Congenital long QT syndrome (LQTS) is a hereditary cardiac channelopathy characterized by delayed ventricular repolarization, syncope, torsades de pointes and sudden cardiac death. Thirty-three members of five apparently 'unrelated' Danish families carry the KCNH2:c.87C> A; p.F29L founder mutation. METHODS AND RESULTS: Linkage disequilibrium mapping with microsatellites around KCNH2 enabled us to estimate the age of the founder mutation to be approximately 22 generations, corresponding to around 550 years. Neighbouring-Joining analysis disclosed one early and three later nodes. The median QTc time of the carriers was 490 ms (range: 415-589 ms) and no difference was seen between the different branches of the family. The mutation is malignant with a penetrance of 73%. Ten F29L carriers received implantable defibrillators (ICDs) (median age at implant 20 years), and of those four individuals experienced eight appropriate shocks. Patch-clamp analysis in HEK 293 cells, performed at 34°C disclosed a loss-of-function phenotype with fast deactivation, reduced steady-state inactivation current density and a positive voltage shift in inactivation. Western blotting of HEK 293 cells transfected with KCNH2:WT and KCNH2:c.87C> A revealed a reduced fraction of fully glycosylated hERG:p.F29L suggesting that this mutation results in defective trafficking. CONCLUSION: The altered channel gating kinetics in combination with defective trafficking of mutated channels is expected to result in reduced repolarizing current density and, thus, a LQTS phenotype.


Assuntos
Canais de Potássio Éter-A-Go-Go/genética , Síndrome do QT Longo/genética , Dinamarca , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Efeito Fundador , Estudos de Associação Genética , Células HEK293 , Haplótipos , Humanos , Ativação do Canal Iônico , Cinética , Masculino , Potenciais da Membrana , Repetições de Microssatélites , Mutação de Sentido Incorreto , Fenótipo , Polimorfismo de Nucleotídeo Único , Transporte Proteico , Análise de Sequência de DNA
11.
Channels (Austin) ; 9(3): 121-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26043299

RESUMO

The voltage-gated Kv1.5 potassium channel, conducting the ultra-rapid rectifier K(+) current (IKur), is regulated through several pathways. Here we investigate if Kv1.5 surface expression is controlled by the 2 kinases PKC and AMPK, using Xenopus oocytes, MDCK cells and atrial derived HL-1 cells. By confocal microscopy combined with electrophysiology we demonstrate that PKC activation reduces Kv1.5 current, through a decrease in membrane expressed channels. AMPK activation was found to decrease the membrane expression in MDCK cells, but not in HL-1 cells and was furthermore shown to be dependent on co-expression of Nedd4-2 in Xenopus oocytes. These results indicate that Kv1.5 channels are regulated by both kinases, although through different molecular mechanisms in different cell systems.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Canal de Potássio Kv1.5/metabolismo , Proteína Quinase C/metabolismo , Animais , Linhagem Celular Tumoral , Cães , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Feminino , Células Madin Darby de Rim Canino , Ubiquitina-Proteína Ligases Nedd4 , Oócitos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Xenopus , Xenopus laevis
12.
Cardiovasc Res ; 101(1): 175-84, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24148898

RESUMO

AIMS: The purpose of this study was to investigate the functional role of G-protein-coupled inward rectifier potassium (GIRK) channels in the cardiac ventricle. METHODS AND RESULTS: Immunofluorescence experiments demonstrated that GIRK4 was localized in outer sarcolemmas and t-tubules in GIRK1 knockout (KO) mice, whereas GIRK4 labelling was not detected in GIRK4 KO mice. GIRK4 was localized in intercalated discs in rat ventricle, whereas it was expressed in intercalated discs and outer sarcolemmas in rat atrium. GIRK4 was localized in t-tubules and intercalated discs in human ventricular endocardium and epicardium, but absent in mid-myocardium. Electrophysiological recordings in rat ventricular tissue ex vivo showed that the adenosine A1 receptor agonist N6-cyclopentyladenosine (CPA) and acetylcholine (ACh) shortened action potential duration (APD), and that the APD shortening was reversed by either the GIRK channel blocker tertiapin-Q, the adenosine A1 receptor antagonist DPCPX or by the muscarinic M2 receptor antagonist AF-DX 116. Tertiapin-Q prolonged APD in the absence of the exogenous receptor activation. Furthermore, CPA and ACh decreased the effective refractory period and the effect was reversed by either tertiapin-Q, DPCPX or AF-DX 116. Receptor activation also hyperpolarized the resting membrane potential, an effect that was reversed by tertiapin-Q. In contrast, tertiapin-Q depolarized the resting membrane potential in the absence of the exogenous receptor activation. CONCLUSION: Confocal microscopy shows that among species GIRK4 is differentially localized in the cardiac ventricle, and that it is heterogeneously expressed across human ventricular wall. Electrophysiological recordings reveal that GIRK current may contribute significantly to ventricular repolarization and thereby to cardiac electrical stability.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Ventrículos do Coração/metabolismo , Animais , Humanos , Masculino , Potenciais da Membrana , Camundongos , Camundongos Knockout , Potássio/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
13.
Circ Cardiovasc Genet ; 6(5): 452-61, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24021552

RESUMO

BACKGROUND: Mutations in CAV3, coding for caveolin-3, the major constituent scaffolding protein of cardiac caveolae, have been associated with skeletal muscle disease, cardiomyopathy, and most recently long-QT syndrome (LQTS) and sudden infant death syndrome. We examined the occurrence of CAV3 mutations in a large cohort of patients with LQTS. METHODS AND RESULTS: Probands with LQTS (n=167) were screened for mutations in CAV3 using direct DNA sequencing. A single proband (0.6%) was found to be a heterozygous carrier of a previously described missense mutation, caveolin-3:p.T78M. The proband was also a heterozygous carrier of the trafficking-deficient Kv11.1:p.I400N mutation. The caveolin-3:p.T78M mutation was found isolated in 3 family members, none of whom had a prolonged QTc interval. Coimmunoprecipitations of caveolin-3 and the voltage-gated potassium channel subunit (Kv11.1) were performed, and the electrophysiological classification of the Kv11.1 mutant was carried out by patch-clamp technique in human embryonic kidney 293 cells. Furthermore, the T-wave morphology was assessed in mutation carriers, double mutation carriers, and nonmutation carriers by applying a morphology combination score. The morphology combination score was normal for isolated caveolin-3:p.T78M carriers and of LQT2 type in double heterozygotes. CONCLUSIONS: Mutations in CAV3 are rare in LQTS. Furthermore, caveolin-3:p.T78M did not exhibit a LQTS phenotype. Because no association has ever been found between LQTS and isolated CAV3 mutations, we suggest that LQTS9 is considered a provisional entity.


Assuntos
Caveolina 3/genética , Canais de Potássio Éter-A-Go-Go/genética , Síndrome do QT Longo/genética , Adolescente , Adulto , Idoso , Caveolina 3/metabolismo , Criança , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Células HEK293 , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Técnicas de Patch-Clamp , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA
14.
J Cardiovasc Pharmacol ; 62(2): 192-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23609329

RESUMO

G protein-coupled inwardly rectifying K⁺ channels (GIRK) are important in the regulation of heart rate and atrial electrophysiology. GIRK channels are activated by G protein-coupled receptors, including muscarinic M2 receptors and adenosine A1 receptors. The aim of this study was to characterize and compare the electrophysiological effects of acetylcholine (ACh) and adenosine on GIRK channels in rat atria. Action potential duration at 90% repolarization (APD90), effective refractory period (ERP), and resting membrane potential (RMP) were investigated in isolated rat atria by intracellular recordings. Both the adenosine analog N6-cyclopentyladenosine (CPA) and ACh profoundly shortened APD90 and ERP and hyperpolarized the RMP. No additive or synergistic effect of CPA and ACh coapplication was observed. To antagonize GIRK channel activation, the specific inhibitor rTertiapin Q (TTQ) was applied. The coapplication of TTQ reversed the CPA and ACh-induced effects. When TTQ was applied without exogenous receptor activator, both APD90 and ERP were prolonged and RMP was depolarized, confirming a basal activity of the GIRK current. The results reveal that activation of A1 and M2 receptors has a profound and equal effect on the electrophysiology in rat atrium. This effect is to a major extent mediated through GIRK channels. Furthermore, these results support the notion that atrial GIRK currents from healthy hearts have a basal component and additional activation can be mediated via at least 2 different receptor mechanisms.


Assuntos
Arritmias Cardíacas/prevenção & controle , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/agonistas , Coração/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Miocárdio/metabolismo , Receptor Muscarínico M2/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Acetilcolina/metabolismo , Adenosina/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/metabolismo , Função Atrial/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Agonistas do Receptor Purinérgico P1/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor Muscarínico M2/agonistas , Período Refratário Eletrofisiológico/efeitos dos fármacos
15.
Cell Physiol Biochem ; 31(2-3): 366-78, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23485744

RESUMO

BACKGROUND/AIMS: Signalling via CysLT1 is involved in activation of volume sensitive K(+) channels and homologous desensitization of the LTD4 receptor impairs regulatory volume decrease (RVD). The aim is to illustrate the effect of mutation of putative PKC consensus phosphorylation sites in the CysLT1R on desensitization and RVD. METHODS: mCysLT1 contains 4 putative PKC consensus phosphorylation sites, and four mutants were created: Thr151Gly, Thr323Gly, Thr151Gly plus Thr323Gly, and Thr236Gly plus Ser243Gly. Functional mCysLT1 receptor activity after injection of in vitro transcribed cRNA into Xenopus laevis oocytes was visualized as a LTD4-evoked, Ca(2+)-activated Cl(-) currents recorded by two-electrode voltage clamp. RESULTS: Repetitive LTD4 administration (100 nM) desensitized the LTD4-evoked currents in oocytes expressing wild type CysLT1. Single mutations as well as the double mutation Thr236Gly plus Ser243Gly had no or a slight effect on the LTD4 induced desensitization. However, double mutation Thr323Gly plus Thr151Gly prevented the desensitization. As a functional consequence we find that inhibition of PKC accelerates RVD and prevents the inhibitory effect of LTD4-pretreatment on RVD in Ehrlich ascites tumour cells. CONCLUSION: These data indicate that simultaneous PKC-mediated phosphorylation at the 2(nd) inner loop (Thr(151)) and at the C-terminal domain (Thr(323)) leads to mCysLT1 receptor desensitization and abrogates the RVD response following osmotic cell swelling.


Assuntos
Proteína Quinase C/metabolismo , Receptores de Leucotrienos/metabolismo , Animais , Carbazóis/farmacologia , Tamanho Celular , Canais de Cloreto/metabolismo , Leucotrieno D4/farmacologia , Camundongos , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Proteína Quinase C/antagonistas & inibidores , Receptores de Leucotrienos/genética , Transdução de Sinais/efeitos dos fármacos , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
16.
J Cardiovasc Electrophysiol ; 23(10): 1092-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22882672

RESUMO

INTRODUCTION: Sudden unexplained death account for one-third of all sudden natural deaths in the young (1-35 years). Hitherto, the prevalence of genopositive cases has primarily been based on deceased persons referred for postmortem genetic testing. These deaths potentially may represent the worst of cases, thus possibly overestimating the prevalence of potentially disease causing mutations in the 3 major long-QT syndrome (LQTS) genes in the general population. We therefore wanted to investigate the prevalence of mutations in an unselected population of sudden unexplained deaths in a nationwide setting. METHODS: DNA for genetic testing was available for 44 cases of sudden unexplained death in Denmark in the period 2000-2006 (equaling 33% of all cases of sudden unexplained death in the age group). KCNQ1, KCNH2, and SCN5A were sequenced and in vitro electrophysiological studies were performed on novel mutations. RESULTS: In total, 5 of 44 cases (11%) carried a mutation in 1 of the 3 genes corresponding to 11% of all investigated cases (R190W KCNQ1, F29L KCNH2 (2 cases), P297S KCNH2 and P1177L SCN5A). P1177L SCN5A has not been reported before. In vitro electrophysiological studies of P1177L SCN5A revealed an increased sustained current suggesting a LQTS phenotype. CONCLUSION: In a nationwide setting, the genetic investigation of an unselected population of sudden unexplained death cases aged 1-35 years finds a lower than expected number of mutations compared to referred populations previously reported. We therefore conclude that the prevalence of mutations in the 3 major LQTS associated genes may not be as abundant as previously estimated.


Assuntos
Morte Súbita Cardíaca/etiologia , Canais de Potássio Éter-A-Go-Go/genética , Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Adolescente , Adulto , Fatores Etários , Análise de Variância , Autopsia , Criança , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Morte Súbita Cardíaca/epidemiologia , Dinamarca , Canal de Potássio ERG1 , Técnicas Eletrofisiológicas Cardíacas , Canais de Potássio Éter-A-Go-Go/metabolismo , Feminino , Frequência do Gene , Predisposição Genética para Doença , Células HEK293 , Humanos , Lactente , Canal de Potássio KCNQ1/metabolismo , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/mortalidade , Masculino , Potenciais da Membrana , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Técnicas de Patch-Clamp , Linhagem , Fenótipo , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/mortalidade , Transfecção , Adulto Jovem
17.
Heart Rhythm ; 9(3): 440-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22024150

RESUMO

BACKGROUND: KCNQ1 (Kv7.1), together with its KCNE ß subunits, plays a pivotal role both in the repolarization of cardiac tissue and in water and salt transport across epithelial membranes. Nedd4/Nedd4-like (neuronal precursor cell-expressed developmentally downregulated 4) ubiquitin-protein ligases interact with the KCNQ1 potassium channel through a PY motif located in the C terminus of KCNQ1. This interaction induces ubiquitylation of KCNQ1, resulting in a reduced surface density of the channel. It was reported recently that the epithelial sodium channel is regulated by the reverse process-deubiquitylation-mediated by USP2 (ubiquitin-specific protease 2). OBJECTIVE: In this article, we investigated whether deubiquitylation may regulate KCNQ1 channel complexes. METHODS: In this study, we used electrophysiology, biochemistry, and confocal microscopy. RESULTS: Electrophysiological investigations of KCNQ1/KCNE1 proteins coexpressed with USP2-45 or USP2-69 isoforms and Nedd4-2 in Xenopus laevis oocytes and mammalian cells revealed that both USP2 isoforms counter the Nedd4-2-specific downregulation of I(Ks). Biochemical studies showed that the total and surface-expressed KCNQ1 protein was more abundant when coexpressed with USP2 and Nedd4-2 as compared with Nedd4-2 alone. Western blotting revealed partial protection against covalent attachment of ubiquitin moieties on KCNQ1 when USP2 was coexpressed with Nedd4-2. Coimmunoprecipitation assays suggested that USP2 can bind to KCNQ1 independently of the PY motif. Immunocytochemistry confirmed that USP2 restores the membrane localization of KCNQ1. CONCLUSION: These results demonstrate that USP2 can be a potent regulator of KCNQ1 surface density. USP2, which is well expressed in many tissues, may therefore be important in controlling the KCNQ1 channel dynamics in vivo.


Assuntos
Endopeptidases/metabolismo , Ativação do Canal Iônico/fisiologia , Canal de Potássio KCNQ1/metabolismo , Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Potenciais de Ação/fisiologia , Animais , Membrana Celular/fisiologia , Regulação para Baixo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Células Epiteliais/metabolismo , Miocárdio/metabolismo , Ubiquitina-Proteína Ligases Nedd4 , Transporte Proteico/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Proteases Específicas de Ubiquitina , Proteínas de Xenopus , Xenopus laevis
18.
Pflugers Arch ; 456(6): 1137-48, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18504605

RESUMO

I (Kr) is the fast component of the delayed rectifier potassium currents responsible for the repolarization of the cardiac muscle. The molecular correlate underlying the I (Kr) current has been identified as the hERG1 channel. Recently, two splice variants of the hERG1 alpha-subunit, hERG1a and hERG1b, have been shown to be co-expressed in human cardiomyocytes. In this paper, we present the electrophysiological characterization of hERG1a, hERG1b, and co-expressed hERG1a/b channels in a mammalian expression system using the whole-cell patch clamp technique. We also quantified the messenger RNA (mRNA) levels of hERG1a and hERG1b in human cardiac tissue, and based on the expressed ratios, we evaluated the resulting currents in Xenopus laevis oocytes. Compared to hERG1a channels, activation was faster for both hERG1b and hERG1a/b channels. The deactivation kinetics was greatly accelerated in the presence of hERG1b, whereas no difference in the time constant of inactivation was observed. The voltage-dependent recovery from inactivation was also similar. However, the time constant of recovery from inactivation was significantly faster for hERG1b channels compared to hERG1a and hERG1a/b. Quantification of hERG1a and hERG1b mRNA in the human heart showed that hERG1b mRNA constitutes, on average, 19% in the right atrium and 12% in the left ventricle of the total hERG1 mRNA. Expression of the observed ratios of hERG1a to hERG1b in X. laevis oocytes showed that these ratios are indeed sufficient to change the deactivation phenotype markedly. The present work suggests that hERG1b is likely to play a role in the formation of the native I (Kr) current.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , DNA/genética , Interpretação Estatística de Dados , Eletrofisiologia , Humanos , Cinética , Miocárdio/citologia , Miocárdio/metabolismo , Oócitos/metabolismo , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Xenopus laevis
19.
Biochem Biophys Res Commun ; 348(4): 1455-62, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16930557

RESUMO

In order to identify proteins interacting with the cardiac voltage-gated sodium channel Na(v)1.5, we used the last 66 amino acids of the C-terminus of the channel as bait to screen a human cardiac cDNA library. We identified the protein tyrosine phosphatase PTPH1 as an interacting protein. Pull-down experiments confirmed the interaction, and indicated that it depends on the PDZ-domain binding motif of Na(v)1.5. Co-expression experiments in HEK293 cells showed that PTPH1 shifts the Na(v)1.5 availability relationship toward hyperpolarized potentials, whereas an inactive PTPH1 or the tyrosine kinase Fyn does the opposite. The results of this study suggest that tyrosine phosphorylation destabilizes the inactivated state of Na(v)1.5.


Assuntos
Proteínas Musculares/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Canais de Sódio/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Condutividade Elétrica , Humanos , Proteínas Musculares/química , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 3 , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Deleção de Sequência , Canais de Sódio/química , Técnicas do Sistema de Duplo-Híbrido
20.
Am J Physiol Cell Physiol ; 288(3): C692-701, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15548568

RESUMO

The voltage-gated Na(+) channels (Na(v)) form a family composed of 10 genes. The COOH termini of Na(v) contain a cluster of amino acids that are nearly identical among 7 of the 10 members. This COOH-terminal sequence, PPSYDSV, is a PY motif known to bind to WW domains of E3 protein-ubiquitin ligases of the Nedd4 family. We recently reported that cardiac Na(v)1.5 is regulated by Nedd4-2. In this study, we further investigated the molecular determinants of regulation of Na(v) proteins. When expressed in HEK-293 cells and studied using whole cell voltage clamping, the neuronal Na(v)1.2 and Na(v)1.3 were also downregulated by Nedd4-2. Pull-down experiments using fusion proteins bearing the PY motif of Na(v)1.2, Na(v)1.3, and Na(v)1.5 indicated that mouse brain Nedd4-2 binds to the Na(v) PY motif. Using intrinsic tryptophan fluorescence imaging of WW domains, we found that Na(v)1.5 PY motif binds preferentially to the fourth WW domain of Nedd4-2 with a K(d) of approximately 55 muM. We tested the binding properties and the ability to ubiquitinate and downregulate Na(v)1.5 of three Nedd4-like E3s: Nedd4-1, Nedd4-2, and WWP2. Despite the fact that along with Nedd4-2, Nedd4-1 and WWP2 bind to Na(v)1.5 PY motif, only Nedd4-2 robustly ubiquitinated and downregulated Na(v)1.5. Interestingly, coexpression of WWP2 competed with the effect of Nedd4-2. Finally, using brefeldin A, we found that Nedd4-2 accelerated internalization of Na(v)1.5 stably expressed in HEK-293 cells. This study shows that Nedd4-dependent ubiquitination of Na(v) channels may represent a general mechanism regulating the excitability of neurons and myocytes via modulation of channel density at the plasma membrane.


Assuntos
Isoformas de Proteínas/metabolismo , Canais de Sódio/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Linhagem Celular , Regulação para Baixo , Eletrofisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte , Humanos , Ativação do Canal Iônico , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Ubiquitina-Proteína Ligases Nedd4 , Peptídeos/metabolismo , Ligação Proteica , Isoformas de Proteínas/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Canais de Sódio/genética , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA