Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 112(40): 12528-33, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26392562

RESUMEN

Nav channels are essential for metazoan membrane depolarization, and Nav channel dysfunction is directly linked with epilepsy, ataxia, pain, arrhythmia, myotonia, and irritable bowel syndrome. Human Nav channelopathies are primarily caused by variants that directly affect Nav channel permeability or gating. However, a new class of human Nav channelopathies has emerged based on channel variants that alter regulation by intracellular signaling or cytoskeletal proteins. Fibroblast growth factor homologous factors (FHFs) are a family of intracellular signaling proteins linked with Nav channel regulation in neurons and myocytes. However, to date, there is surprisingly little evidence linking Nav channel gene variants with FHFs and human disease. Here, we provide, to our knowledge, the first evidence that mutations in SCN5A (encodes primary cardiac Nav channel Nav1.5) that alter FHF binding result in human cardiovascular disease. We describe a five*generation kindred with a history of atrial and ventricular arrhythmias, cardiac arrest, and sudden cardiac death. Affected family members harbor a novel SCN5A variant resulting in p.H1849R. p.H1849R is localized in the central binding core on Nav1.5 for FHFs. Consistent with these data, Nav1.5 p.H1849R affected interaction with FHFs. Further, electrophysiological analysis identified Nav1.5 p.H1849R as a gain-of-function for INa by altering steady-state inactivation and slowing the rate of Nav1.5 inactivation. In line with these data and consistent with human cardiac phenotypes, myocytes expressing Nav1.5 p.H1849R displayed prolonged action potential duration and arrhythmogenic afterdepolarizations. Together, these findings identify a previously unexplored mechanism for human Nav channelopathy based on altered Nav1.5 association with FHF proteins.


Asunto(s)
Arritmias Cardíacas/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.5/genética , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Canalopatías/genética , Canalopatías/metabolismo , Canalopatías/fisiopatología , Salud de la Familia , Femenino , Predisposición Genética a la Enfermedad/genética , Células HEK293 , Humanos , Immunoblotting , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Linaje , Unión Proteica
2.
J Biol Chem ; 290(19): 12210-21, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25825486

RESUMEN

Proper trafficking of membrane-bound ion channels and transporters is requisite for normal cardiac function. Endosome-based protein trafficking of membrane-bound ion channels and transporters in the heart is poorly understood, particularly in vivo. In fact, for select cardiac cell types such as atrial myocytes, virtually nothing is known regarding endosomal transport. We previously linked the C-terminal Eps15 homology domain-containing protein 3 (EHD3) with endosome-based protein trafficking in ventricular cardiomyocytes. Here we sought to define the roles and membrane protein targets for EHD3 in atria. We identify the voltage-gated T-type Ca(2+) channels (CaV3.1, CaV3.2) as substrates for EHD3-dependent trafficking in atria. Mice selectively lacking EHD3 in heart display reduced expression and targeting of both Cav3.1 and CaV3.2 in the atria. Furthermore, functional experiments identify a significant loss of T-type-mediated Ca(2+) current in EHD3-deficient atrial myocytes. Moreover, EHD3 associates with both CaV3.1 and CaV3.2 in co-immunoprecipitation experiments. T-type Ca(2+) channel function is critical for proper electrical conduction through the atria. Consistent with these roles, EHD3-deficient mice demonstrate heart rate variability, sinus pause, and atrioventricular conduction block. In summary, our findings identify CaV3.1 and CaV3.2 as substrates for EHD3-dependent protein trafficking in heart, provide in vivo data on endosome-based trafficking pathways in atria, and implicate EHD3 as a key player in the regulation of atrial myocyte excitability and cardiac conduction.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Endosomas/metabolismo , Atrios Cardíacos/metabolismo , Alelos , Animales , Calcio/química , Canales de Calcio Tipo T/genética , Enfermedades Cardiovasculares/metabolismo , Electrocardiografía , Regulación de la Expresión Génica , Frecuencia Cardíaca , Ventrículos Cardíacos/citología , Ratones , Ratones Noqueados , Células Musculares/citología , Mutación , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato
3.
Circulation ; 131(8): 695-708, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25632041

RESUMEN

BACKGROUND: The cardiac cytoskeleton plays key roles in maintaining myocyte structural integrity in health and disease. In fact, human mutations in cardiac cytoskeletal elements are tightly linked to cardiac pathologies, including myopathies, aortopathies, and dystrophies. Conversely, the link between cytoskeletal protein dysfunction and cardiac electric activity is not well understood and often overlooked in the cardiac arrhythmia field. METHODS AND RESULTS: Here, we uncover a new mechanism for the regulation of cardiac membrane excitability. We report that ßII spectrin, an actin-associated molecule, is essential for the posttranslational targeting and localization of critical membrane proteins in heart. ßII spectrin recruits ankyrin-B to the cardiac dyad, and a novel human mutation in the ankyrin-B gene disrupts the ankyrin-B/ßII spectrin interaction, leading to severe human arrhythmia phenotypes. Mice lacking cardiac ßII spectrin display lethal arrhythmias, aberrant electric and calcium handling phenotypes, and abnormal expression/localization of cardiac membrane proteins. Mechanistically, ßII spectrin regulates the localization of cytoskeletal and plasma membrane/sarcoplasmic reticulum protein complexes, including the Na/Ca exchanger, ryanodine receptor 2, ankyrin-B, actin, and αII spectrin. Finally, we observe accelerated heart failure phenotypes in ßII spectrin-deficient mice. CONCLUSIONS: Our findings identify ßII spectrin as critical for normal myocyte electric activity, link this molecule to human disease, and provide new insight into the mechanisms underlying cardiac myocyte biology.


Asunto(s)
Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Citoesqueleto/fisiología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Espectrina/fisiología , Secuencia de Aminoácidos , Animales , Ancirinas/genética , Ancirinas/fisiología , Arritmias Cardíacas/genética , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/fisiología , Microtúbulos/fisiología , Datos de Secuencia Molecular , Mutación/genética , Fenotipo , Espectrina/análisis , Espectrina/química
4.
Am J Physiol Heart Circ Physiol ; 310(11): H1583-91, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27106045

RESUMEN

ß2-Spectrin is critical for integrating membrane and cytoskeletal domains in excitable and nonexcitable cells. The role of ß2-spectrin for vertebrate function is illustrated by dysfunction of ß2-spectrin-based pathways in disease. Recently, defects in ß2-spectrin association with protein partner ankyrin-B were identified in congenital forms of human arrhythmia. However, the role of ß2-spectrin in common forms of acquired heart failure and arrhythmia is unknown. We report that ß2-spectrin protein levels are significantly altered in human cardiovascular disease as well as in large and small animal cardiovascular disease models. Specifically, ß2-spectrin levels were decreased in atrial samples of patients with atrial fibrillation compared with tissue from patients in sinus rhythm. Furthermore, compared with left ventricular samples from nonfailing hearts, ß2-spectrin levels were significantly decreased in left ventricle of ischemic- and nonischemic heart failure patients. Left ventricle samples of canine and murine heart failure models confirm reduced ß2-spectrin protein levels. Mechanistically, we identify that ß2-spectrin levels are tightly regulated by posttranslational mechanisms, namely Ca(2+)- and calpain-dependent proteases. Furthermore, consistent with this data, we observed Ca(2+)- and calpain-dependent loss of ß2-spectrin downstream effector proteins, including ankyrin-B in heart. In summary, our findings illustrate that ß2-spectrin and downstream molecules are regulated in multiple forms of cardiovascular disease via Ca(2+)- and calpain-dependent proteolysis.


Asunto(s)
Fibrilación Atrial/metabolismo , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Espectrina/metabolismo , Adulto , Anciano , Animales , Ancirinas/metabolismo , Fibrilación Atrial/fisiopatología , Calcio/metabolismo , Calpaína/metabolismo , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Perros , Regulación hacia Abajo , Femenino , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteolisis , Transducción de Señal , Volumen Sistólico , Función Ventricular Izquierda
5.
Proc Natl Acad Sci U S A ; 110(43): 17576-81, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24101510

RESUMEN

Identified over a dozen years ago in the brain and pancreatic islet, ßIV-spectrin is critical for the local organization of protein complexes throughout the nervous system. ßIV-Spectrin targets ion channels and adapter proteins to axon initial segments and nodes of Ranvier in neurons, and ßIV-spectrin dysfunction underlies ataxia and early death in mice. Despite advances in ßIV-spectrin research in the nervous system, its role in pancreatic islet biology is unknown. Here, we report that ßIV-spectrin serves as a multifunctional structural and signaling platform in the pancreatic islet. We report that ßIV-spectrin directly associates with and targets the calcium/calmodulin-dependent protein kinase II (CaMKII) in pancreatic islets. In parallel, ßIV-spectrin targets ankyrin-B and the ATP-sensitive potassium channel. Consistent with these findings, ßIV-spectrin mutant mice lacking CaMKII- or ankyrin-binding motifs display selective loss of expression and targeting of key protein components, including CaMKIIδ. ßIV-Spectrin-targeted CaMKII directly phosphorylates the inwardly-rectifying potassium channel, Kir6.2 (alpha subunit of KATP channel complex), and we identify the specific residue, Kir6.2 T224, responsible for CaMKII-dependent regulation of KATP channel function. CaMKII-dependent phosphorylation alters channel regulation resulting in KATP channel inhibition, a cellular phenotype consistent with aberrant insulin regulation. Finally, we demonstrate aberrant KATP channel phosphorylation in ßIV-spectrin mutant mice. In summary, our findings establish a broader role for ßIV-spectrin in regulation of cell membrane excitability in the pancreatic islet, define the pathway for CaMKII local control in pancreatic beta cells, and identify the mechanism for CaMKII-dependent regulation of KATP channels.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Secretoras de Insulina/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Espectrina/metabolismo , Animales , Ancirinas/metabolismo , Sitios de Unión/genética , Células COS , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas , Chlorocebus aethiops , Immunoblotting , Inmunohistoquímica , Masculino , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Mutación , Fosforilación , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/fisiología , Unión Proteica , Espectrina/genética
6.
J Biol Chem ; 289(8): 5285-95, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24394417

RESUMEN

N-type and P/Q-type calcium channels are documented players in the regulation of synaptic function; however, the mechanisms underlying their expression and cellular targeting are poorly understood. Ankyrin polypeptides are essential for normal integral membrane protein expression in a number of cell types, including neurons, cardiomyocytes, epithelia, secretory cells, and erythrocytes. Ankyrin dysfunction has been linked to defects in integral protein expression, abnormal cellular function, and disease. Here, we demonstrate that ankyrin-B associates with Cav2.1 and Cav2.2 in cortex, cerebellum, and brain stem. Additionally, using in vitro and in vivo techniques, we demonstrate that ankyrin-B, via its membrane-binding domain, associates with a highly conserved motif in the DII/III loop domain of Cav2.1 and Cav2.2. Further, we demonstrate that this domain is necessary for proper targeting of Cav2.1 and Cav2.2 in a heterologous system. Finally, we demonstrate that mutation of a single conserved tyrosine residue in the ankyrin-binding motif of both Cav2.1 (Y797E) and Cav2.2 (Y788E) results in loss of association with ankyrin-B in vitro and in vivo. Collectively, our findings identify an interaction between ankyrin-B and both Cav2.1 and Cav2.2 at the amino acid level that is necessary for proper Cav2.1 and Cav2.2 targeting in vivo.


Asunto(s)
Ancirinas/metabolismo , Canales de Calcio Tipo N/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Encéfalo , Canales de Calcio Tipo N/química , Secuencia Conservada , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Unión Proteica , Células de Purkinje/metabolismo , Ratas , Tirosina/metabolismo
7.
Biochim Biophys Acta ; 1838(2): 723-30, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23732236

RESUMEN

The past twenty years have revealed the existence of numerous ion channel mutations resulting in human pathology. Ion channels provide the basis of diverse cellular functions, ranging from hormone secretion, excitation-contraction coupling, cell signaling, immune response, and trans-epithelial transport. Therefore, the regulation of biophysical properties of channels is vital in human physiology. Only within the last decade has the role of non-ion channel components come to light in regard to ion channel spatial, temporal, and biophysical regulation in physiology. A growing number of auxiliary components have been determined to play elemental roles in excitable cell physiology, with dysfunction resulting in disorders and related manifestations. This review focuses on the broad implications of such dysfunction, focusing on disease-causing mutations that alter interactions between ion channels and auxiliary ion channel components in a diverse set of human excitable cell disease. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé


Asunto(s)
Canalopatías/patología , Canales Iónicos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Animales , Transporte Biológico , Canalopatías/metabolismo , Humanos
8.
Am J Physiol Gastrointest Liver Physiol ; 309(6): G506-12, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26185330

RESUMEN

Human jejunum smooth muscle cells (SMCs) and interstitial cells of Cajal (ICCs) express the SCN5A-encoded voltage-gated, mechanosensitive sodium channel NaV1.5. NaV1.5 contributes to small bowel excitability, and NaV1.5 inhibitor ranolazine produces constipation by an unknown mechanism. We aimed to determine the presence and molecular identity of Na(+) current in the human colon smooth muscle and to examine the effects of ranolazine on Na(+) current, mechanosensitivity, and smooth muscle contractility. Inward currents were recorded by whole cell voltage clamp from freshly dissociated human colon SMCs at rest and with shear stress. SCN5A mRNA and NaV1.5 protein were examined by RT-PCR and Western blots, respectively. Ascending human colon strip contractility was examined in a muscle bath preparation. SCN5A mRNA and NaV1.5 protein were identified in human colon circular muscle. Freshly dissociated human colon SMCs had Na(+) currents (-1.36 ± 0.36 pA/pF), shear stress increased Na(+) peaks by 17.8 ± 1.8% and accelerated the time to peak activation by 0.7 ± 0.3 ms. Ranolazine (50 µM) blocked peak Na(+) current by 43.2 ± 9.3% and inhibited shear sensitivity by 25.2 ± 3.2%. In human ascending colon strips, ranolazine decreased resting tension (31%), reduced the frequency of spontaneous events (68%), and decreased the response to smooth muscle electrical field stimulation (61%). In conclusion, SCN5A-encoded NaV1.5 is found in human colonic circular smooth muscle. Ranolazine blocks both peak amplitude and mechanosensitivity of Na(+) current in human colon SMCs and decreases contractility of human colon muscle strips. Our data provide a likely mechanistic explanation for constipation induced by ranolazine.


Asunto(s)
Colon/metabolismo , Miocitos del Músculo Liso/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Ranolazina/farmacología , Colon/efectos de los fármacos , Colon Ascendente/efectos de los fármacos , Colon Ascendente/metabolismo , Estreñimiento/genética , Células HEK293 , Humanos , Contracción Muscular/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Estimulación Física
9.
J Biol Chem ; 288(3): 1568-81, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23223335

RESUMEN

Cardiac ATP-sensitive potassium (K(ATP)) channels are key sensors and effectors of the metabolic status of cardiomyocytes. Alteration in their expression impacts their effectiveness in maintaining cellular energy homeostasis and resistance to injury. We sought to determine how activation of calcium/calmodulin-dependent protein kinase II (CaMKII), a central regulator of calcium signaling, translates into reduced membrane expression and current capacity of cardiac K(ATP) channels. We used real-time monitoring of K(ATP) channel current density, immunohistochemistry, and biotinylation studies in isolated hearts and cardiomyocytes from wild-type and transgenic mice as well as HEK cells expressing wild-type and mutant K(ATP) channel subunits to track the dynamics of K(ATP) channel surface expression. Results showed that activation of CaMKII triggered dynamin-dependent internalization of K(ATP) channels. This process required phosphorylation of threonine at 180 and 224 and an intact (330)YSKF(333) endocytosis motif of the K(ATP) channel Kir6.2 pore-forming subunit. A molecular model of the µ2 subunit of the endocytosis adaptor protein, AP2, complexed with Kir6.2 predicted that µ2 docks by interaction with (330)YSKF(333) and Thr-180 on one and Thr-224 on the adjacent Kir6.2 subunit. Phosphorylation of Thr-180 and Thr-224 would favor interactions with the corresponding arginine- and lysine-rich loops on µ2. We concluded that calcium-dependent activation of CaMKII results in phosphorylation of Kir6.2, which promotes endocytosis of cardiac K(ATP) channel subunits. This mechanism couples the surface expression of cardiac K(ATP) channels with calcium signaling and reveals new targets to improve cardiac energy efficiency and stress resistance.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Regulación de la Expresión Génica , Miocitos Cardíacos/enzimología , Canales de Potasio de Rectificación Interna/metabolismo , Complejo 2 de Proteína Adaptadora/química , Complejo 2 de Proteína Adaptadora/metabolismo , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/química , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Dinaminas/genética , Dinaminas/metabolismo , Endocitosis , Activación Enzimática , Células HEK293 , Humanos , Transporte Iónico , Ratones , Ratones Transgénicos , Modelos Moleculares , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Fosforilación , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Transducción de Señal , Treonina/metabolismo
10.
Curr Top Membr ; 72: 121-58, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24210429

RESUMEN

The vertebrate cardiac myocyte has evolved a highly organized cellular membrane architecture and cell-cell contacts in order to effectively transmit precisely timed and homogeneous depolarizing waves without failure (>2 billion times/human life span). Two unique specialized membrane domains, the intercalated disc and the transverse tubule (T-tubule), function to ensure the rapid and coordinated propagation of the action potential throughout the heart. Based on their critical roles in structure, signaling, and electric inter- and intracellular communication, it is not surprising that dysfunction in these membrane structures is associated with aberrant vertebrate physiology, resulting in potentially fatal congenital and acquired disease. This chapter will review the fundamental components of cardiomyocyte intercalated disc and transverse-tubule membranes with a focus on linking dysfunction in these membranes with human cardiovascular disease.


Asunto(s)
Membrana Celular/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Cadherinas/química , Cadherinas/genética , Cadherinas/metabolismo , Cateninas/química , Cateninas/genética , Cateninas/metabolismo , Membrana Celular/química , Conexinas/química , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Humanos , Canales Iónicos/química , Canales Iónicos/metabolismo , Miocitos Cardíacos/citología , Vinculina/química , Vinculina/genética , Vinculina/metabolismo
11.
Circ Res ; 107(1): 84-95, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20489164

RESUMEN

RATIONALE: Cardiac membrane excitability is tightly regulated by an integrated network of membrane-associated ion channels, transporters, receptors, and signaling molecules. Membrane protein dynamics in health and disease are maintained by a complex ensemble of intracellular targeting, scaffolding, recycling, and degradation pathways. Surprisingly, despite decades of research linking dysfunction in membrane protein trafficking with human cardiovascular disease, essentially nothing is known regarding the molecular identity or function of these intracellular targeting pathways in excitable cardiomyocytes. OBJECTIVE: We sought to discover novel pathways for membrane protein targeting in primary cardiomyocytes. METHODS AND RESULTS: We report the initial characterization of a large family of membrane trafficking proteins in human heart. We used a tissue-wide screen for novel ankyrin-associated trafficking proteins and identified 4 members of a unique Eps15 homology (EH) domain-containing protein family (EHD1, EHD2, EHD3, EHD4) that serve critical roles in endosome-based membrane protein targeting in other cell types. We show that EHD1-4 directly associate with ankyrin, provide the first information on the expression and localization of these molecules in primary cardiomyocytes, and demonstrate that EHD1-4 are coexpressed with ankyrin-B in the myocyte perinuclear region. Notably, the expression of multiple EHD proteins is increased in animal models lacking ankyrin-B, and EHD3-deficient cardiomyocytes display aberrant ankyrin-B localization and selective loss of Na/Ca exchanger expression and function. Finally, we report significant modulation of EHD expression following myocardial infarction, suggesting that these proteins may play a key role in regulating membrane excitability in normal and diseased heart. CONCLUSIONS: Our findings identify and characterize a new class of cardiac trafficking proteins, define the first group of proteins associated with the ankyrin-based targeting network, and identify potential new targets to modulate membrane excitability in disease. Notably, these data provide the first link between EHD proteins and a human disease model.


Asunto(s)
Proteínas Portadoras/fisiología , Membrana Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Transporte Vesicular/fisiología , Proteínas Portadoras/metabolismo , Membrana Celular/química , Membrana Celular/genética , Proteínas de Unión al ADN/fisiología , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Familia de Multigenes/fisiología , Proteínas Nucleares/fisiología , Estructura Terciaria de Proteína/genética , Transporte de Proteínas/genética , Proteínas de Transporte Vesicular/metabolismo
12.
Proc Natl Acad Sci U S A ; 106(39): 16669-74, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19805355

RESUMEN

The coordinated sorting of ion channels to specific plasma membrane domains is necessary for excitable cell physiology. K(ATP) channels, assembled from pore-forming (Kir6.x) and regulatory sulfonylurea receptor subunits, are critical electrical transducers of the metabolic state of excitable tissues, including skeletal and smooth muscle, heart, brain, kidney, and pancreas. Here we show that the C-terminal domain of Kir6.2 contains a motif conferring membrane targeting in primary excitable cells. Kir6.2 lacking this motif displays aberrant channel targeting due to loss of association with the membrane adapter ankyrin-B (AnkB). Moreover, we demonstrate that this Kir6.2 C-terminal AnkB-binding motif (ABM) serves a dual role in K(ATP) channel trafficking and membrane metabolic regulation and dysfunction in these pathways results in human excitable cell disease. Thus, the K(ATP) channel ABM serves as a previously unrecognized bifunctional touch-point for grading K(ATP) channel gating and membrane targeting and may play a fundamental role in controlling excitable cell metabolic regulation.


Asunto(s)
Secuencias de Aminoácidos , Membrana Celular/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Ancirinas/genética , Ancirinas/metabolismo , Sitios de Unión , Células Cultivadas , Femenino , Humanos , Masculino , Ratones , Canales de Potasio de Rectificación Interna/genética , Transfección
13.
Cells ; 11(4)2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35203314

RESUMEN

Zonula occludens-1 (ZO-1) is an intracellular scaffolding protein that orchestrates the anchoring of membrane proteins to the cytoskeleton in epithelial and specialized tissue including the heart. There is clear evidence to support the central role of intracellular auxiliary proteins in arrhythmogenesis and previous studies have found altered ZO-1 expression associated with atrioventricular conduction abnormalities. Here, using human cardiac tissues, we identified all three isoforms of ZO-1, canonical (Transcript Variant 1, TV1), CRA_e (Transcript Variant 4, TV4), and an additionally expressed (Transcript Variant 3, TV3) in non-failing myocardium. To investigate the role of ZO-1 on ventricular arrhythmogenesis, we generated a haploinsufficient ZO-1 mouse model (ZO-1+/-). ZO-1+/- mice exhibited dysregulated connexin-43 protein expression and localization at the intercalated disc. While ZO-1+/- mice did not display abnormal cardiac function at baseline, adrenergic challenge resulted in rhythm abnormalities, including premature ventricular contractions and bigeminy. At baseline, ventricular myocytes from the ZO-1+/- mice displayed prolonged action potential duration and spontaneous depolarizations, with ZO-1+/- cells displaying frequent unsolicited (non-paced) diastolic depolarizations leading to spontaneous activity with multiple early afterdepolarizations (EADs). Mechanistically, ZO-1 deficient myocytes displayed a reduction in sodium current density (INa) and an increased sensitivity to isoproterenol stimulation. Further, ZO-1 deficient myocytes displayed remodeling in ICa current, likely a compensatory change. Taken together, our data suggest that ZO-1 deficiency results in myocardial substrate susceptible to triggered arrhythmias.


Asunto(s)
Miocardio , Uniones Estrechas , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Ratones , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Sodio/metabolismo , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
14.
J Biol Chem ; 285(37): 28723-30, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20610380

RESUMEN

Ankyrin polypeptides are critical for normal membrane protein expression in diverse cell types, including neurons, myocytes, epithelia, and erythrocytes. Ankyrin dysfunction results in defects in membrane expression of ankyrin-binding partners (including ion channels, transporters, and cell adhesion molecules), resulting in aberrant cellular function and disease. Here, we identify a new role for ankyrin-B in cardiac cell biology. We demonstrate that cardiac sarcolemmal K(ATP) channels directly associate with ankyrin-B in heart via the K(ATP) channel alpha-subunit Kir6.2. We demonstrate that primary myocytes lacking ankyrin-B display defects in Kir6.2 protein expression, membrane expression, and function. Moreover, we demonstrate a secondary role for ankyrin-B in regulating K(ATP) channel gating. Finally, we demonstrate that ankyrin-B forms a membrane complex with K(ATP) channels and the cardiac Na/K-ATPase, a second key membrane transporter involved in the cardiac ischemia response. Collectively, our new findings define a new role for cardiac ankyrin polypeptides in regulation of ion channel membrane expression in heart.


Asunto(s)
Ancirinas/metabolismo , Membrana Celular/metabolismo , Regulación de la Expresión Génica , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Canales de Potasio de Rectificación Interna/sangre , Animales , Ancirinas/genética , Línea Celular , Membrana Celular/genética , Humanos , Activación del Canal Iónico/fisiología , Ratones , Ratones Noqueados , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/genética , Sarcolema/metabolismo
15.
Drug Discov Today Dis Models ; 6(3): 63-69, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20689672

RESUMEN

Proper regulation of cardiac ion channel activity is critical for cellular ion homeostasis and myocyte electrical activity. Recent work has demonstrated that cardiac ion channels are not isolated pores in plasmid membranes, but rather exist within macromolecular signaling complexes. Moreover, within these macro-complexes resides the machinery to finely tune ion channel expression, activity, and signaling. While it is widely-accepted that mutations in ion channel pore-forming genes underlie a number of cardiac arrhythmias, current research is now focusing on the roles of auxiliary subunits in the development of arrhythmia syndromes.

16.
J Clin Invest ; 129(8): 3171-3184, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31264976

RESUMEN

Arrhythmogenic cardiomyopathy (ACM) is an inherited arrhythmia syndrome characterized by severe structural and electrical cardiac phenotypes, including myocardial fibrofatty replacement and sudden cardiac death. Clinical management of ACM is largely palliative, owing to an absence of therapies that target its underlying pathophysiology, which stems partially from our limited insight into the condition. Following identification of deceased ACM probands possessing ANK2 rare variants and evidence of ankyrin-B loss of function on cardiac tissue analysis, an ANK2 mouse model was found to develop dramatic structural abnormalities reflective of human ACM, including biventricular dilation, reduced ejection fraction, cardiac fibrosis, and premature death. Desmosomal structure and function appeared preserved in diseased human and murine specimens in the presence of markedly abnormal ß-catenin expression and patterning, leading to identification of a previously unknown interaction between ankyrin-B and ß-catenin. A pharmacological activator of the WNT/ß-catenin pathway, SB-216763, successfully prevented and partially reversed the murine ACM phenotypes. Our findings introduce what we believe to be a new pathway for ACM, a role of ankyrin-B in cardiac structure and signaling, a molecular link between ankyrin-B and ß-catenin, and evidence for targeted activation of the WNT/ß-catenin pathway as a potential treatment for this disease.


Asunto(s)
Ancirinas , Displasia Ventricular Derecha Arritmogénica , Miocardio , Vía de Señalización Wnt , Animales , Ancirinas/genética , Ancirinas/metabolismo , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/metabolismo , Displasia Ventricular Derecha Arritmogénica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Indoles/farmacología , Masculino , Maleimidas/farmacología , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , beta Catenina/genética , beta Catenina/metabolismo
17.
J Cell Biochem ; 104(4): 1244-53, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18275062

RESUMEN

Inositol 1,4,5-trisphosphate (InsP(3)) receptors are calcium-release channels found in the endoplasmic/sarcoplasmic reticulum (ER/SR) membrane of diverse cell types. InsP(3) receptors release Ca(2+) from ER/SR lumenal stores in response to InsP(3) generated from various stimuli. The complex spatial and temporal patterns of InsP(3) receptor-mediated Ca(2+) release regulate many cellular processes, ranging from gene transcription to memory. Ankyrins are adaptor proteins implicated in the targeting of ion channels and transporters to specialized membrane domains. Multiple independent studies have documented in vitro and in vivo interactions between ankyrin polypeptides and the InsP(3) receptor. Moreover, loss of ankyrin-B leads to loss of InsP(3) receptor membrane expression and stability in cardiomyocytes. Despite extensive biochemical and functional data, the validity of in vivo ankyrin-InsP(3) receptor interactions remains controversial. This controversy is based on inconsistencies between a previously identified ankyrin-binding region on the InsP(3) receptor and InsP(3) receptor topology data that demonstrate the inaccessibility of this lumenal binding site on the InsP(3) receptor to cytosolic ankyrin polypeptides. Here we use two methods to revisit the requirements on InsP(3) receptor for ankyrin binding. We demonstrate that ankyrin-B interacts with the cytoplasmic N-terminal domain of InsP(3) receptor. In summary, our findings demonstrate that the ankyrin-binding site is located on the cytoplasmic face of the InsP(3) receptor, thus validating the feasibility of in vivo ankyrin-InsP(3) receptor interactions.


Asunto(s)
Ancirinas/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ancirinas/química , Sitios de Unión , Citoplasma , Humanos , Receptores de Inositol 1,4,5-Trifosfato/química , Miocitos Cardíacos , Unión Proteica , Transporte de Proteínas , Técnicas del Sistema de Dos Híbridos
18.
Life Sci ; 196: 118-126, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29366750

RESUMEN

AIMS: Right ventricular failure (RVF) varies significantly from the more common left ventricular failure (LVF). This study was undertaken to determine potential molecular pathways that are important in human right ventricular (RV) function and may mediate RVF. MATERIALS AND METHODS: We analyzed mRNA of human non-failing LV and RV samples and RVF samples from patients with pulmonary arterial hypertension (PAH), and post-LVAD implantation. We then performed transcript analysis to determine differential expression of genes in the human heart samples. Immunoblot quantification was performed followed by analysis of non-failing and failing phenotypes. KEY FINDINGS: Inflammatory pathways were more commonly dysregulated in RV tissue (both non-failing and failing phenotypes). In non-failing human RV tissue we found important differences in expression of FIGF, TRAPPAC, and CTGF suggesting that regulation of normal RV and LV function are not the same. In failing RV tissue, FBN2, CTGF, SMOC2, and TRAPP6AC were differentially expressed, and are potential targets for further study. SIGNIFICANCE: This work provides some of the first analyses of the molecular heterogeneity between human RV and LV tissue, as well as key differences in human disease (RVF secondary to pulmonary hypertension and LVAD mediated RVF). Our transcriptional data indicated that inflammatory pathways may be more important in RV tissue, and changes in FIGF and CTGF supported this hypothesis. In PAH RV failure samples, upregulation of FBN2 and CTGF further reinforced the potential significance that altered remodeling and inflammation play in normal RV function and failure.


Asunto(s)
Insuficiencia Cardíaca/genética , Disfunción Ventricular Derecha/genética , Adulto , Anciano , Biomarcadores/análisis , Hipertensión Pulmonar Primaria Familiar/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Derecha/patología , Función Ventricular Derecha
19.
Circ Cardiovasc Genet ; 10(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28196901

RESUMEN

BACKGROUND: Long QT syndrome confers susceptibility to ventricular arrhythmia, predisposing to syncope, seizures, and sudden death. While rare globally, long QT syndrome is ≈15× more common in First Nations of Northern British Columbia largely because of a known mutation in KCNQ1. However, 2 large multigenerational families were affected, but negative for the known mutation. METHODS AND RESULTS: Long QT syndrome panel testing was carried out in the index case of each family, and clinical information was collected. Cascade genotyping was performed. Biochemical and myocyte-based assays were performed to evaluate the identified gene variant for loss-of-function activity. Index cases in these 2 families harbored a novel ANK2 c.1937C>T variant (p.S646F). An additional 16 carriers were identified, including 2 with structural heart disease: one with cardiomyopathy resulting in sudden death and the other with congenital heart disease. For all carriers of this variant, the average QTc was 475 ms (±40). Although ankyrin-B p.S646F is appropriately folded and expressed in bacteria, the mutant polypeptide displays reduced expression in cultured H9c2 cells and aberrant localization in primary cardiomyocytes. Furthermore, myocytes expressing ankyrin-B p.S646F lack normal membrane targeting of the ankyrin-binding partner, the Na/Ca exchanger. Thus, ankyrin-B p.S646F is a loss-of-function variant. CONCLUSIONS: We identify the first disease-causing ANK2 variant localized to the membrane-binding domain resulting in reduced ankyrin-B expression and abnormal localization. Further study is warranted on the potential association of this variant with structural heart disease given the role of ANK2 in targeting and stabilization of key structural and signaling molecules in cardiac cells.


Asunto(s)
Ancirinas/genética , Arritmias Cardíacas/genética , Variación Genética , Indígenas Norteamericanos/genética , Síndrome de QT Prolongado/genética , Adolescente , Adulto , Anciano , Animales , Ancirinas/química , Ancirinas/metabolismo , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etnología , Arritmias Cardíacas/metabolismo , Colombia Británica/epidemiología , Línea Celular , Niño , Preescolar , Electrocardiografía , Femenino , Predisposición Genética a la Enfermedad , Humanos , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/etnología , Síndrome de QT Prolongado/metabolismo , Masculino , Ratones Noqueados , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Ratas , Intercambiador de Sodio-Calcio/metabolismo , Relación Estructura-Actividad , Transfección
20.
Heart Rhythm ; 13(9): 1932-40, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27298202

RESUMEN

BACKGROUND: Human ANK2 (ankyrin-B) loss-of-function variants are directly linked with arrhythmia phenotypes. However, in atypical non-ion channel arrhythmia genes such as ANK2 that lack the same degree of robust structure/function and clinical data, it may be more difficult to assign variant disease risk based simply on variant location, minor allele frequency, and/or predictive structural algorithms. The human ankyrin-B p.L1622I variant found in arrhythmia probands displays significant diversity in minor allele frequency across populations. OBJECTIVE: The objective of this study was to directly test the in vivo impact of ankyrin-B p.L1622I on cardiac electrical phenotypes and arrhythmia risk using a new animal model. METHODS: We tested arrhythmia phenotypes in a new "knock-in" animal model harboring the human ankyrin-B p.L1622I variant. RESULTS: Ankyrin-B p.L1622I displays reduced posttranslational expression in vivo, resulting in reduced cardiac ankyrin-B expression and reduced association with binding-partner Na/Ca exchanger. Ankyrin-B(L1622I/L1622I) mice display changes in heart rate, atrioventricular and intraventricular conduction, and alterations in repolarization. Furthermore, ankyrin-B(L1622I/L1622I) mice display catecholamine-dependent arrhythmias. At the cellular level, ankyrin-B(L1622I/L1622I) myocytes display increased action potential duration and severe arrhythmogenic afterdepolarizations that provide a mechanistic rationale for the arrhythmias. CONCLUSION: Our findings support in vivo arrhythmogenic phenotypes of an ANK2 variant with unusual frequency in select populations. On the basis of our findings and current clinical data, we support classification of p.L1622I as a "mild" loss-of-function variant that may confer arrhythmia susceptibility in the context of secondary risk factors including environment, medication, and/or additional genetic variation.


Asunto(s)
Ancirinas/genética , Arritmias Cardíacas/genética , Potenciales de Acción/genética , Animales , Arritmias Cardíacas/etnología , Arritmias Cardíacas/fisiopatología , Población Negra/genética , Modelos Animales de Enfermedad , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad/etnología , Variación Genética , Humanos , Mutación con Pérdida de Función , Ratones , Ratones Endogámicos C57BL , Fenotipo , Medición de Riesgo/etnología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA