Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(2): 287-302, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35105987

RESUMEN

The volume-regulated anion channel (VRAC) is formed by LRRC8 proteins and is responsible for the regulatory volume decrease (RVD) after hypotonic cell swelling. Besides chloride, VRAC transports other molecules, for example, immunomodulatory cyclic dinucleotides (CDNs) including 2'3'cGAMP. Here, we identify LRRC8C as a critical component of VRAC in T cells, where its deletion abolishes VRAC currents and RVD. T cells of Lrrc8c-/- mice have increased cell cycle progression, proliferation, survival, Ca2+ influx and cytokine production-a phenotype associated with downmodulation of p53 signaling. Mechanistically, LRRC8C mediates the transport of 2'3'cGAMP in T cells, resulting in STING and p53 activation. Inhibition of STING recapitulates the phenotype of LRRC8C-deficient T cells, whereas overexpression of p53 inhibits their enhanced T cell function. Lrrc8c-/- mice have exacerbated T cell-dependent immune responses, including immunity to influenza A virus infection and experimental autoimmune encephalomyelitis. Our results identify cGAMP uptake through LRRC8C and STING-p53 signaling as a new inhibitory signaling pathway in T cells and adaptive immunity.


Asunto(s)
Aniones/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Canales Iónicos/metabolismo , Proteínas de la Membrana/metabolismo , Linfocitos T/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Calcio/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Nucleótidos Cíclicos/metabolismo , Transducción de Señal/fisiología
2.
Am J Physiol Cell Physiol ; 324(5): C1017-C1027, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36878847

RESUMEN

Sirtuins are NAD+-dependent deacetylases with beneficial roles in conditions relevant to human health, including metabolic disease, type II diabetes, obesity, cancer, aging, neurodegenerative diseases, and cardiac ischemia. Since ATP-sensitive K+ (KATP) channels have cardioprotective roles, we investigated whether they are regulated by sirtuins. Nicotinamide mononucleotide (NMN) was used to increase cytosolic NAD+ levels and to activate sirtuins in cell lines, isolated rat and mouse cardiomyocytes or insulin-secreting INS-1 cells. KATP channels were studied with patch clamping, biochemistry techniques, and antibody uptake experiments. NMN led to an increase in intracellular NAD+ levels and an increase in the KATP channel current, without significant changes in the unitary current amplitude or open probability. An increased surface expression was confirmed using surface biotinylation approaches. The rate of KATP channel internalization was diminished by NMN, which may be a partial explanation for the increased surface expression. We show that NMN acts via sirtuins since the increased KATP channel surface expression was prevented by blockers of SIRT1 and SIRT2 (Ex527 and AGK2) and mimicked by SIRT1 activation (SRT1720). The pathophysiological relevance of this finding was studied using a cardioprotection assay with isolated ventricular myocytes, in which NMN protected against simulated ischemia or hypoxia in a KATP channel-dependent manner. Overall, our data draw a link between intracellular NAD+, sirtuin activation, KATP channel surface expression, and cardiac protection against ischemic damage.


Asunto(s)
Diabetes Mellitus Tipo 2 , Sirtuinas , Ratas , Ratones , Humanos , Animales , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo , NAD/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Miocitos Cardíacos/metabolismo , Adenosina Trifosfato/metabolismo , Canales KATP/genética , Canales KATP/metabolismo
3.
Physiol Rev ; 96(1): 177-252, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26660852

RESUMEN

KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Canales KATP/metabolismo , Transducción de Señal , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/fisiopatología , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Canales KATP/antagonistas & inhibidores , Canales KATP/química , Canales KATP/genética , Masculino , Potenciales de la Membrana , Ratones Transgénicos , Fenotipo , Bloqueadores de los Canales de Potasio/farmacología , Conformación Proteica , Subunidades de Proteína , Transporte de Proteínas , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
4.
Proc Natl Acad Sci U S A ; 117(19): 10593-10602, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32332165

RESUMEN

A physiological role for long-chain acyl-CoA esters to activate ATP-sensitive K+ (KATP) channels is well established. Circulating palmitate is transported into cells and converted to palmitoyl-CoA, which is a substrate for palmitoylation. We found that palmitoyl-CoA, but not palmitic acid, activated the channel when applied acutely. We have altered the palmitoylation state by preincubating cells with micromolar concentrations of palmitic acid or by inhibiting protein thioesterases. With acyl-biotin exchange assays we found that Kir6.2, but not sulfonylurea receptor (SUR)1 or SUR2, was palmitoylated. These interventions increased the KATP channel mean patch current, increased the open time, and decreased the apparent sensitivity to ATP without affecting surface expression. Similar data were obtained in transfected cells, rat insulin-secreting INS-1 cells, and isolated cardiac myocytes. Kir6.2ΔC36, expressed without SUR, was also positively regulated by palmitoylation. Mutagenesis of Kir6.2 Cys166 prevented these effects. Clinical variants in KCNJ11 that affect Cys166 had a similar gain-of-function phenotype, but was more pronounced. Molecular modeling studies suggested that palmitoyl-C166 and selected large hydrophobic mutations make direct hydrophobic contact with Kir6.2-bound PIP2 Patch-clamp studies confirmed that palmitoylation of Kir6.2 at Cys166 enhanced the PIP2 sensitivity of the channel. Physiological relevance is suggested since palmitoylation blunted the regulation of KATP channels by α1-adrenoreceptor stimulation. The Cys166 residue is conserved in some other Kir family members (Kir6.1 and Kir3, but not Kir2), which are also subject to regulated palmitoylation, suggesting a general mechanism to control the open state of certain Kir channels.


Asunto(s)
Canales KATP/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Acilcoenzima A/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Cisteína/metabolismo , Células HEK293 , Humanos , Canales KATP/genética , Lipoilación/fisiología , Mutagénesis/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp/métodos , Canales de Potasio/metabolismo , Canales de Potasio de Rectificación Interna/fisiología , Cultivo Primario de Células , Ratas , Receptores de Sulfonilureas/genética
5.
Am J Physiol Cell Physiol ; 322(6): C1230-C1247, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35508187

RESUMEN

Sarcolemmal/plasmalemmal ATP-sensitive K+ (KATP) channels have key roles in many cell types and tissues. Hundreds of studies have described how the KATP channel activity and ATP sensitivity can be regulated by changes in the cellular metabolic state, by receptor signaling pathways and by pharmacological interventions. These alterations in channel activity directly translate to alterations in cell or tissue function, that can range from modulating secretory responses, such as insulin release from pancreatic ß-cells or neurotransmitters from neurons, to modulating contractile behavior of smooth muscle or cardiac cells to elicit alterations in blood flow or cardiac contractility. It is increasingly becoming apparent, however, that KATP channels are regulated beyond changes in their activity. Recent studies have highlighted that KATP channel surface expression is a tightly regulated process with similar implications in health and disease. The surface expression of KATP channels is finely balanced by several trafficking steps including synthesis, assembly, anterograde trafficking, membrane anchoring, endocytosis, endocytic recycling, and degradation. This review aims to summarize the physiological and pathophysiological implications of KATP channel trafficking and mechanisms that regulate KATP channel trafficking. A better understanding of this topic has potential to identify new approaches to develop therapeutically useful drugs to treat KATP channel-related diseases.


Asunto(s)
Células Secretoras de Insulina , Adenosina Trifosfato/metabolismo , Transporte Biológico , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Canales KATP/genética , Canales KATP/metabolismo , Transporte de Proteínas
6.
J Mol Cell Cardiol ; 144: 1-11, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32339567

RESUMEN

BACKGROUND: Genetic variants in SCN5A can result in channelopathies such as the long QT syndrome type 3 (LQT3), but the therapeutic response to Na+ channel blockers can vary. We previously reported a case of an infant with malignant LQT3 and a missense Q1475P SCN5A variant, who was effectively treated with phenytoin, but only partially with mexiletine. Here, we functionally characterized this variant and investigated possible mechanisms for the differential drug actions. METHODS: Wild-type or mutant Nav1.5 cDNAs were examined in transfected HEK293 cells with patch clamping and biochemical assays. We used computational modeling to provide insights into altered channel kinetics and to predict effects on the action potential. RESULTS: The Q1475P variant in Nav1.5 reduced the current density and channel surface expression, characteristic of a trafficking defect. The variant also led to positive shifts in the voltage dependence of steady-state activation and inactivation, faster inactivation and recovery from inactivation, and increased the "late" Na+ current. Simulations of Nav1.5 gating with a 9-state Markov model suggested that transitions from inactivated to closed states were accelerated in Q1475P channels, leading to accumulation of channels in non-inactivated closed states. Simulations with a human ventricular myocyte model predicted action potential prolongation with Q1475P, compared with wild type, channels. Patch clamp data showed that mexiletine and phenytoin similarly rescued some of the gating defects. Chronic incubation with mexiletine, but not phenytoin, rescued the Nav1.5-Q1475P trafficking defect, thus increasing mutant channel expression. CONCLUSIONS: The gain-of-function effects of Nav1.5-Q1475P predominate to cause a malignant long QT phenotype. Phenytoin partially corrects the gating defect without restoring surface expression of the mutant channel, whereas mexiletine restores surface expression of the mutant channel, which may explain the lack of efficacy of mexiletine when compared to phenytoin. Our data makes a case for experimental studies before embarking on a one-for-all therapy of arrhythmias.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/etiología , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Síndrome de QT Prolongado/etiología , Fenitoína/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Potenciales de Acción/efectos de los fármacos , Sustitución de Aminoácidos , Antiarrítmicos/farmacología , Trastorno del Sistema de Conducción Cardíaco/diagnóstico , Trastorno del Sistema de Conducción Cardíaco/tratamiento farmacológico , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Células Cultivadas , Mutación con Ganancia de Función , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/metabolismo , Modelos Biológicos , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Fenitoína/uso terapéutico
7.
FASEB J ; 32(3): 1613-1625, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29133341

RESUMEN

ATP-sensitive K+ (KATP) channels uniquely link cellular energy metabolism to membrane excitability and are expressed in diverse cell types that range from the endocrine pancreas to neurons and smooth, skeletal, and cardiac muscle. A decrease in the surface expression of KATP channels has been linked to various disorders, including dysregulated insulin secretion, abnormal blood pressure, and impaired resistance to cardiac injury. In contrast, up-regulation of KATP channel surface expression may be protective, for example, by mediating the beneficial effect of ischemic preconditioning. Molecular mechanisms that regulate KATP channel trafficking are poorly understood. Here, we used cellular assays with immunofluorescence, surface biotinylation, and patch clamping to demonstrate that Eps15 homology domain-containing protein 2 (EHD2) is a novel positive regulator of KATP channel trafficking to increase surface KATP channel density. EHD2 had no effect on cardiac Na+ channels (Nav1.5). The effect is specific to EHD2 as other members of the EHD family-EHD1, EHD3, and EHD4-had no effect on KATP channel surface expression. EHD2 did not directly affect KATP channel properties as unitary conductance and ATP sensitivity were unchanged. Instead, we observed that the mechanism by which EHD2 increases surface expression is by stabilizing KATP channel-containing caveolar structures, which results in a reduced rate of endocytosis. EHD2 also regulated KATP channel trafficking in isolated cardiomyocytes, which validated the physiologic relevance of these observations. Pathophysiologically, EHD2 may be cardioprotective as a dominant-negative EHD2 mutant sensitized cardiomyocytes to ischemic damage. Our findings highlight EHD2 as a potential pharmacologic target in the treatment of diseases with KATP channel trafficking defects.-Yang, H. Q., Jana, K., Rindler, M. J., Coetzee, W. A. The trafficking protein, EHD2, positively regulates cardiac sarcolemmal KATP channel surface expression: role in cardioprotection.


Asunto(s)
Proteínas Portadoras/metabolismo , Canales KATP/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Animales , Células COS , Proteínas Portadoras/genética , Chlorocebus aethiops , Células HEK293 , Humanos , Canales KATP/genética , Ratones , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Miocardio/patología , Miocitos Cardíacos/patología , Transporte de Proteínas , Ratas , Sarcolema/genética
8.
Pacing Clin Electrophysiol ; 42(2): 275-282, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30578647

RESUMEN

The HCN4 gene encodes a subunit of the hyperpolarization-activated cyclic nucleotide-gated channel, type 4 that is essential for the proper generation of pacemaker potentials in the sinoatrial node. The HCN4 gene is often present in targeted genetic testing panels for various cardiac conduction system disorders and there are several reports of HCN4 variants associated with conduction disorders. Here, we report the in vitro functional characterization of four rare variants of uncertain significance (VUS) in HCN4, identified through testing a cohort of 296 sudden unexpected natural deaths. The variants are all missense alterations, leading to single amino acid changes: p.E66Q in the N-terminus, p.D546N in the C-linker domain, and both p.S935Y and p.R1044Q in the C-terminus distal to the CNBD. We also identified a likely benign variant, p. P1063T, which has a high minor allele frequency in the gnomAD, which is utilized here as a negative control. Three of the HCN4 VUS (p.E66Q, p.S935Y, and p.R1044Q) had electrophysiological characteristics similar to the wild-type channel, suggesting that these variants are benign. In contrast, the p.D546N variant in the C-linker domain exhibited a larger current density, slower activation, and was unresponsive to cyclic adenosine monophosphate (cAMP) compared to wild-type. With functional assays, we reclassified three rare HCN4 VUS to likely benign variants, eliminating the necessity for costly and time-consuming further study. Our studies also provide a new lead to investigate how a VUS located in the C-linker connecting the pore to the cAMP binding domain may affect the channel open state probability and cAMP response.


Asunto(s)
Muerte Súbita Cardíaca , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/clasificación , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Proteínas Musculares/clasificación , Proteínas Musculares/genética , Canales de Potasio/clasificación , Canales de Potasio/genética , Células Cultivadas , Fenómenos Electrofisiológicos , Variación Genética , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Proteínas Musculares/fisiología , Canales de Potasio/fisiología
9.
Forensic Sci Med Pathol ; 15(3): 437-444, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30547356

RESUMEN

Determining the cause of unexplained death in all age groups, including infants, is a priority in forensic medicine. The triple risk model proposed for sudden infant death syndrome involves the intersection of three risks: (1) a critical developmental period in homeostatic control (2), exogenous stressors, and (3) a vulnerable infant. Even though sex and age factor into some forms of inherited arrhythmogenic deaths in young individuals and adults, more appropriate a dual-risk disease model for adults involves exogenous stressors and a vulnerable individual. The vulnerability aspect clearly has a genetic component as underscored by a number of recent large-scale and high-throughput genetic testing studies performed in attempt to define the causes of sudden unexplained death. These studies often focus on 'cardiac' and channelopathy genes. Genetic testing often identify lists of rare or ultra-rare nonsynonymous variants, classified according to the ACMG guidelines as 'pathogenic' or 'likely pathogenic', which may form the basis of diagnostic decisions and/or family counseling. However, computer algorithms used to categorize gene variants are not completely accurate and these variants are often not functionally tested to determine their pathogenicity. Due to conflicting computational predictions, a large number of variants are labeled as 'variants of uncertain significance' or VUS. Functional testing of these VUS can greatly assist to reclassify these VUS as 'likely benign' or 'likely pathogenic'. However, functional testing has its limits and by itself cannot be used to determine cause of death. Going forward, computer algorithms must be improved to take account of variants across multiple genes and efforts must be expanded to obtain clinical, familial and segregation data. Forensic genetic testing needs to be held to the same rigorous standards as defined by the NIH Clinical Genome Resource Consortium, where functional evaluation of a channelopathy variant is only one (but important) aspect of the overall picture.


Asunto(s)
Canalopatías/genética , Muerte Súbita/etiología , Variación Genética , Muerte Súbita del Lactante/genética , Genética Forense , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Lactante
10.
Pacing Clin Electrophysiol ; 40(6): 703-712, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28370132

RESUMEN

BACKGROUND: Two genetic variants in SCN5A, encoding the Nav1.5 Na+ channel α-subunit, were found in a 5-month-old girl who died suddenly in her sleep. The first variant is a missense mutation, resulting in an amino acid change (Q1832E), which has been described (but not characterized) in a patient with Brugada syndrome. The second is a nonsense mutation that produces a premature stop codon and a C-terminal truncation (R1944Δ). METHODS AND RESULTS: To investigate their functional relevance with patch clamp experiments in transfected HEK-293 cells. The Q1832E mutation drastically reduced Nav1.5 current density. The R1944Δ C-terminal truncation had negligible effects on Nav1.5 current density. Neither of the mutations affected the voltage dependence of steady activation and inactivation or influenced the late Na+ current or the recovery from inactivation. Biochemical and immunofluorescent approaches demonstrated that the Q1832E mutation caused severe trafficking defects. Polymerase chain reaction cloning and sequencing the victim's genomic DNA allowed us to determine that the two variants were in trans. We investigated the functional consequences by coexpressing Nav1.5(Q1832E) and Nav1.5(R1944Δ), which led to a significantly reduced current amplitude relative to wild-type. CONCLUSIONS: These sudden infant death syndrome (SIDS)-related variants caused a severely dysfunctional Nav1.5 channel, which was mainly due to trafficking defects caused by the Q1832E mutation. The decreased current density is likely to be a major contributing factor to arrhythmogenesis in Brugada syndrome and the sudden death of this SIDS victim.


Asunto(s)
Activación del Canal Iónico/genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Polimorfismo de Nucleótido Simple/genética , Sodio/metabolismo , Muerte Súbita del Lactante/genética , Muerte Súbita Cardíaca , Femenino , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Células HEK293 , Humanos , Incidencia , Lactante , Mutación/genética , Factores de Riesgo , Tasa de Supervivencia
11.
Am J Physiol Heart Circ Physiol ; 310(11): H1558-66, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27037371

RESUMEN

Myocardial ischemia remains the primary cause of morbidity and mortality in the United States. Ischemic preconditioning (IPC) is a powerful form of endogenous protection against myocardial infarction. We studied alterations in KATP channels surface density as a potential mechanism of the protection of IPC. Using cardiac-specific knockout of Kir6.2 subunits, we demonstrated an essential role for sarcolemmal KATP channels in the infarct-limiting effect of IPC in the mouse heart. With biochemical membrane fractionation, we demonstrated that sarcolemmal KATP channel subunits are distributed both to the sarcolemma and intracellular endosomal compartments. Global ischemia causes a loss of sarcolemmal KATP channel subunit distribution and internalization to endosomal compartments. Ischemia-induced internalization of KATP channels was prevented by CaMKII inhibition. KATP channel subcellular redistribution was also observed with immunohistochemistry. Ischemic preconditioning before the index ischemia reduced not only the infarct size but also prevented KATP channel internalization. Furthermore, not only did adenosine mimic IPC by preventing infarct size, but it also prevented ischemia-induced KATP channel internalization via a PKC-mediated pathway. We show that preventing endocytosis with dynasore reduced both KATP channel internalization and strongly mitigated infarct development. Our data demonstrate that plasticity of KATP channel surface expression must be considered as a potentially important mechanism of the protective effects of IPC and adenosine.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Sarcolema/metabolismo , Adenosina/farmacología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Endocitosis , Endosomas/metabolismo , Hidrazonas/farmacología , Preparación de Corazón Aislado , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Canales de Potasio de Rectificación Interna/deficiencia , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Sarcolema/efectos de los fármacos , Factores de Tiempo
12.
Biochem Biophys Res Commun ; 471(1): 129-34, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26828268

RESUMEN

Sarcolemmal ATP-sensitive potassium (KATP) channels control skeletal muscle energy use through their ability to adjust membrane excitability and related cell functions in accordance with cellular metabolic status. Mice with disrupted skeletal muscle KATP channels exhibit reduced adipocyte size and increased fatty acid release into the circulation. As yet, the molecular mechanisms underlying this link between skeletal muscle KATP channel function and adipose mobilization have not been established. Here, we demonstrate that skeletal muscle-specific disruption of KATP channel function in transgenic (TG) mice promotes production and secretion of musclin. Musclin is a myokine with high homology to atrial natriuretic peptide (ANP) that enhances ANP signaling by competing for elimination. Augmented musclin production in TG mice is driven by a molecular cascade resulting in enhanced acetylation and nuclear exclusion of the transcription factor forkhead box O1 (FOXO1) - an inhibitor of transcription of the musclin encoding gene. Musclin production/secretion in TG is paired with increased mobilization of fatty acids and a clear trend toward increased circulating ANP, an activator of lipolysis. These data establish KATP channel-dependent musclin production as a potential mechanistic link coupling "local" skeletal muscle energy consumption with mobilization of bodily resources from fat. Understanding such mechanisms is an important step toward designing interventions to manage metabolic disorders including those related to excess body fat and associated co-morbidities.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales KATP/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Factores de Transcripción/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos
13.
Proc Natl Acad Sci U S A ; 109(3): E154-63, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22203979

RESUMEN

TBX3 is critical for human development: mutations in TBX3 cause congenital anomalies in patients with ulnar-mammary syndrome. Data from mice and humans suggest multiple roles for Tbx3 in development and function of the cardiac conduction system. The mechanisms underlying the functional development, maturation, and maintenance of the conduction system are not well understood. We tested the requirements for Tbx3 in these processes. We generated a unique series of Tbx3 hypomorphic and conditional mouse mutants with varying levels and locations of Tbx3 activity within the heart, and developed techniques for evaluating in vivo embryonic conduction system function. Disruption of Tbx3 function in different regions of the developing heart causes discrete phenotypes and lethal arrhythmias: sinus pauses and bradycardia indicate sinoatrial node dysfunction, whereas preexcitation and atrioventricular block reveal abnormalities in the atrioventricular junction. Surviving Tbx3 mutants are at increased risk for sudden death. Arrhythmias induced by knockdown of Tbx3 in adults reveal its requirement for conduction system homeostasis. Arrhythmias in Tbx3-deficient embryos are accompanied by disrupted expression of multiple ion channels despite preserved expression of previously described conduction system markers. These findings indicate that Tbx3 is required for the conduction system to establish and maintain its correct molecular identity and functional properties. In conclusion, Tbx3 is required for the functional development, maturation, and homeostasis of the conduction system in a highly dosage-sensitive manner. TBX3 and its regulatory targets merit investigation as candidates for human arrhythmias.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Dosificación de Gen , Sistema de Conducción Cardíaco/fisiopatología , Homeostasis/genética , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Alelos , Animales , Animales Recién Nacidos , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/diagnóstico por imagen , Arritmias Cardíacas/patología , Bloqueo Atrioventricular/complicaciones , Bloqueo Atrioventricular/diagnóstico por imagen , Bloqueo Atrioventricular/patología , Bloqueo Atrioventricular/fisiopatología , Nodo Atrioventricular/patología , Nodo Atrioventricular/fisiopatología , Conexina 43/metabolismo , Electrocardiografía , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Sistema de Conducción Cardíaco/anomalías , Sistema de Conducción Cardíaco/diagnóstico por imagen , Sistema de Conducción Cardíaco/patología , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Mutación/genética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recombinación Genética/genética , Análisis de Supervivencia , Proteínas de Dominio T Box/metabolismo , Ultrasonografía
14.
Proteomics ; 13(2): 368-78, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23197389

RESUMEN

ATP-sensitive K(+) (K(ATP)) channels are expressed ubiquitously, but have diverse roles in various organs and cells. Their diversity can partly be explained by distinct tissue-specific compositions of four copies of the pore-forming inward rectifier potassium channel subunits (Kir6.1 and/or Kir6.2) and four regulatory sulfonylurea receptor subunits (SUR1 and/or SUR2). Channel function and/or subcellular localization also can be modified by the proteins with which they transiently or permanently interact to generate even more diversity. We performed a quantitative proteomic analysis of K(ATP) channel complexes in the heart, endothelium, insulin-secreting min6 cells (pancreatic ß-cell like), and the hypothalamus to identify proteins with which they interact in different tissues. Glycolysis is an overrepresented pathway in identified proteins of the heart, min6 cells, and the endothelium. Proteins with other energy metabolic functions were identified in the hypothalamic samples. These data suggest that the metabolo-electrical coupling conferred by K(ATP) channels is conferred partly by proteins with which they interact. A large number of identified cytoskeletal and trafficking proteins suggests endocytic recycling may help control K(ATP) channel surface density and/or subcellular localization. Overall, our data demonstrate that K(ATP) channels in different tissues may assemble with proteins having common functions, but that tissue-specific complex organization also occurs.


Asunto(s)
Canales KATP/química , Canales KATP/metabolismo , Proteómica/métodos , Transportadoras de Casetes de Unión a ATP , Animales , Endotelio/química , Endotelio/metabolismo , Células Secretoras de Insulina/química , Células Secretoras de Insulina/metabolismo , Canales KATP/análisis , Ratones , Miocardio/química , Miocardio/metabolismo , Especificidad de Órganos , Canales de Potasio de Rectificación Interna , Receptores de Droga , Receptores de Sulfonilureas
15.
J Biol Chem ; 287(49): 41258-67, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23066018

RESUMEN

Ventricular ATP-sensitive potassium (K(ATP)) channels link intracellular energy metabolism to membrane excitability and contractility. Our recent proteomics experiments identified plakoglobin and plakophilin-2 (PKP2) as putative K(ATP) channel-associated proteins. We investigated whether the association of K(ATP) channel subunits with junctional proteins translates to heterogeneous subcellular distribution within a cardiac myocyte. Co-immunoprecipitation experiments confirmed physical interaction between K(ATP) channels and PKP2 and plakoglobin in rat heart. Immunolocalization experiments demonstrated that K(ATP) channel subunits (Kir6.2 and SUR2A) are expressed at a higher density at the intercalated disk in mouse and rat hearts, where they co-localized with PKP2 and plakoglobin. Super-resolution microscopy demonstrate that K(ATP) channels are clustered within nanometer distances from junctional proteins. The local K(ATP) channel density, recorded in excised inside-out patches, was larger at the cell end when compared with local currents recorded from the cell center. The K(ATP) channel unitary conductance, block by MgATP and activation by MgADP, did not differ between these two locations. Whole cell K(ATP) channel current density (activated by metabolic inhibition) was ∼40% smaller in myocytes from mice haploinsufficient for PKP2. Experiments with excised patches demonstrated that the regional heterogeneity of K(ATP) channels was absent in the PKP2 deficient mice, but the K(ATP) channel unitary conductance and nucleotide sensitivities remained unaltered. Our data demonstrate heterogeneity of K(ATP) channel distribution within a cardiac myocyte. The higher K(ATP) channel density at the intercalated disk implies a possible role at the intercellular junctions during cardiac ischemia.


Asunto(s)
Canales KATP/química , Miocitos Cardíacos/citología , Adenosina Difosfato/química , Adenosina Trifosfato/química , Animales , Membrana Celular/metabolismo , Desmosomas/metabolismo , Masculino , Ratones , Microscopía/métodos , Isquemia Miocárdica/patología , Placofilinas/metabolismo , Ratas , Ratas Sprague-Dawley , gamma Catenina/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 304(9): H1231-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23436329

RESUMEN

Cardiac metabolism remains altered for an extended period of time after myocardial infarction. Studies have shown fibroblasts from normal hearts express KATP channels in culture. It is unknown whether fibroblasts from infarcted hearts express KATP channels and whether these channels contribute to scar and border zone electrophysiology. KATP channel subunit expression levels were determined in fibroblasts isolated from normal hearts (Fb), and scar (sMI-Fb) and remote (rMI-Fb) regions of left anterior descending coronary artery (LAD) ligated rat hearts. Whole cell KATP current density was determined with patch clamp. Action potential duration (APD) was measured with optical mapping in myocyte-only cultures and heterocellular cultures with fibroblasts with and without 100 µmol/l pinacidil. Whole heart optical mapping was used to assess KATP channel activity following LAD ligation. Pinacidil activated a potassium current (35.4 ± 7.5 pA/pF at 50 mV) in sMI-Fb that was inhibited with 10 µmol/l glibenclamide. Kir6.2 and SUR2 transcript levels were elevated in sMI-Fb. Treatment with Kir6.2 short interfering RNA decreased KATP currents (87%) in sMI-Fb. Treatment with pinacidil decreased APD (26%) in co-cultures with sMI-Fb. APD values were prolonged in LAD ligated hearts after perfusion with glibenclamide. KATP channels are present in fibroblasts from the scar and border zones of infarcted hearts. Activation of fibroblast KATP channels could modulate the electrophysiological substrate beyond the acute ischemic event. Targeting fibroblast KATP channels could represent a novel therapeutic approach to modify border zone electrophysiology after cardiac injury.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Fibroblastos/fisiología , Canales KATP/fisiología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Potenciales de Acción/fisiología , Animales , Fibroblastos/metabolismo , Gliburida/farmacología , Ventrículos Cardíacos/citología , Canales KATP/agonistas , Canales KATP/antagonistas & inhibidores , Canales KATP/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Pinacidilo/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Ratas , Ratas Wistar , Receptores de Droga/genética , Receptores de Droga/metabolismo , Receptores de Sulfonilureas , Transcripción Genética , Imagen de Colorante Sensible al Voltaje
17.
Circ Res ; 109(3): 291-5, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21680895

RESUMEN

RATIONALE: Flecainide prevents arrhythmias in catecholaminergic polymorphic ventricular tachycardia, but the antiarrhythmic mechanism remains unresolved. It is possible for flecainide to directly affect the cardiac ryanodine receptor (RyR2); however, an extracellular site of action is suggested because of the hydrophilic nature of flecainide. OBJECTIVE: To investigate the mechanism for the antiarrhythmic action of flecainide in a RyR2(R4496C+/-) knock-in mouse model of catecholaminergic polymorphic ventricular tachycardia. METHODS AND RESULTS: Flecainide prevented catecholamine-induced sustained ventricular tachycardia in RyR2(R4496C+/-) mice. Cellular studies were performed with isolated RyR2(R4496C+/-) myocytes. Isoproterenol caused the appearance of spontaneous Ca(2+) transients, which were unaffected by flecainide (6 µmol/L). Flecainide did not affect Ca(2+) transient amplitude, decay, or sarcoplasmic reticulum Ca(2+) content. Moreover, it did not affect the frequency of spontaneous Ca(2+) sparks in permeabilized myocytes. In contrast, flecainide effectively prevented triggered activity induced by isoproterenol. The threshold for action potential induction was increased significantly (P<0.01), which suggests a primary extracellular antiarrhythmic effect mediated by Na(+) channel blockade. CONCLUSIONS: Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in RyR2(R4496C+/-) mice; however, at variance with previous reports, we observed minimal effects on intracellular Ca(2+) homeostasis. Our data suggest that the antiarrhythmic activity of the drug is caused by reduction of Na(+) channel availability and by an increase in the threshold for triggered activity.


Asunto(s)
Antiarrítmicos/farmacología , Flecainida/farmacología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Taquicardia Ventricular/tratamiento farmacológico , Taquicardia Ventricular/prevención & control , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Modelos Animales de Enfermedad , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/fisiología , Técnicas de Sustitución del Gen , Isoproterenol/farmacología , Ratones , Ratones Mutantes , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/fisiología , Canales de Sodio/fisiología , Simpatomiméticos/farmacología , Taquicardia Ventricular/genética
18.
Front Pharmacol ; 14: 1197257, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37408765

RESUMEN

Background: KATP channels have diverse roles, including regulation of insulin secretion and blood flow, and protection against biological stress responses and are excellent therapeutic targets. Different subclasses of KATP channels exist in various tissue types due to the unique assemblies of specific pore-forming (Kir6.x) and accessory (SURx) subunits. The majority of pharmacological openers and blockers act by binding to SURx and are poorly selective against the various KATP channel subclasses. Methods and Results: We used 3D models of the Kir6.2/SUR homotetramers based on existing cryo-EM structures of channels in both the open and closed states to identify a potential agonist binding pocket in a functionally critical area of the channel. Computational docking screens of this pocket with the Chembridge Core chemical library of 492,000 drug-like compounds yielded 15 top-ranked "hits", which were tested for activity against KATP channels using patch clamping and thallium (Tl+) flux assays with a Kir6.2/SUR2A HEK-293 stable cell line. Several of the compounds increased Tl+ fluxes. One of them (CL-705G) opened Kir6.2/SUR2A channels with a similar potency as pinacidil (EC50 of 9 µM and 11 µM, respectively). Remarkably, compound CL-705G had no or minimal effects on other Kir channels, including Kir6.1/SUR2B, Kir2.1, or Kir3.1/Kir3.4 channels, or Na+ currents of TE671 medulloblastoma cells. CL-705G activated Kir6.2Δ36 in the presence of SUR2A, but not when expressed by itself. CL-705G activated Kir6.2/SUR2A channels even after PIP2 depletion. The compound has cardioprotective effects in a cellular model of pharmacological preconditioning. It also partially rescued activity of the gating-defective Kir6.2-R301C mutant that is associated with congenital hyperinsulinism. Conclusion: CL-705G is a new Kir6.2 opener with little cross-reactivity with other channels tested, including the structurally similar Kir6.1. This, to our knowledge, is the first Kir-specific channel opener.

19.
J Mol Cell Cardiol ; 52(3): 596-607, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22245446

RESUMEN

Since ion channels move electrical charge during their activity, they have traditionally been studied using electrophysiological approaches. This was sometimes combined with mathematical models, for example with the description of the ionic mechanisms underlying the initiation and propagation of action potentials in the squid giant axon by Hodgkin and Huxley. The methods for studying ion channels also have strong roots in protein chemistry (limited proteolysis, the use of antibodies, etc.). The advent of the molecular cloning and the identification of genes coding for specific ion channel subunits in the late 1980s introduced a multitude of new techniques with which to study ion channels and the field has been rapidly expanding ever since (e.g. antibody development against specific peptide sequences, mutagenesis, the use of gene targeting in animal models, determination of their protein structures) and new methods are still in development. This review focuses on techniques commonly employed to examine ion channel function in an electrophysiological laboratory. The focus is on the K(ATP) channel, but many of the techniques described are also used to study other ion channels.


Asunto(s)
Canales KATP/metabolismo , Miocardio/metabolismo , Animales , Técnicas Electrofisiológicas Cardíacas/métodos , Expresión Génica , Marcación de Gen/métodos , Humanos , Canales KATP/química , Canales KATP/genética , Ratones , Ratones Transgénicos , Complejos Multiproteicos/metabolismo , Procesamiento Proteico-Postraduccional , Subunidades de Proteína , Transporte de Proteínas
20.
J Mol Cell Cardiol ; 52(2): 410-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21888913

RESUMEN

AMPK is an important sensor of cellular energy levels. The aim of these studies was to investigate whether cardiac K(ATP) channels, which couple cellular energy metabolism to membrane excitability, are regulated by AMPK activity. We investigated effects of AMPK on rat ventricular K(ATP) channels using electrophysiological and biochemical approaches. Whole-cell K(ATP) channel current was activated by metabolic inhibition; this occurred more rapidly in the presence of AICAR (an AMPK activator). AICAR had no effects on K(ATP) channel activity recorded in the inside-out patch clamp configuration, but ZMP (the intracellular intermediate of AICAR) strongly activated K(ATP) channels. An AMPK-mediated effect is demonstrated by the finding that ZMP had no effect on K(ATP) channels in the presence of Compound C (an AMPK inhibitor). Recombinant AMPK activated Kir6.2/SUR2A channels in a manner that was dependent on the AMP concentration, whereas heat-inactivated AMPK was without effect. Using mass-spectrometry and co-immunoprecipitation approaches, we demonstrate that the AMPK α-subunit physically associates with K(ATP) channel subunits. Our data demonstrate that the cardiac K(ATP) channel function is directly regulated by AMPK activation. During metabolic stress, a small change in cellular AMP that activates AMPK can be a potential trigger for K(ATP) channel opening. This article is part of a Special Issue entitled "Local Signaling in Myocytes".


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético/fisiología , Canales KATP/metabolismo , Adenosina Monofosfato/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Células COS , Chlorocebus aethiops , Canales KATP/agonistas , Canales KATP/genética , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Transducción de Señal , Estrés Fisiológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA