Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(36): e2206708119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36044551

RESUMEN

The sinoatrial node (SAN), the leading pacemaker region, generates electrical impulses that propagate throughout the heart. SAN dysfunction with bradyarrhythmia is well documented in heart failure (HF). However, the underlying mechanisms are not completely understood. Mitochondria are critical to cellular processes that determine the life or death of the cell. The release of Ca2+ from the ryanodine receptors 2 (RyR2) on the sarcoplasmic reticulum (SR) at mitochondria-SR microdomains serves as the critical communication to match energy production to meet metabolic demands. Therefore, we tested the hypothesis that alterations in the mitochondria-SR connectomics contribute to SAN dysfunction in HF. We took advantage of a mouse model of chronic pressure overload-induced HF by transverse aortic constriction (TAC) and a SAN-specific CRISPR-Cas9-mediated knockdown of mitofusin-2 (Mfn2), the mitochondria-SR tethering GTPase protein. TAC mice exhibited impaired cardiac function with HF, cardiac fibrosis, and profound SAN dysfunction. Ultrastructural imaging using electron microscope (EM) tomography revealed abnormal mitochondrial structure with increased mitochondria-SR distance. The expression of Mfn2 was significantly down-regulated and showed reduced colocalization with RyR2 in HF SAN cells. Indeed, SAN-specific Mfn2 knockdown led to alterations in the mitochondria-SR microdomains and SAN dysfunction. Finally, disruptions in the mitochondria-SR microdomains resulted in abnormal mitochondrial Ca2+ handling, alterations in localized protein kinase A (PKA) activity, and impaired mitochondrial function in HF SAN cells. The current study provides insights into the role of mitochondria-SR microdomains in SAN automaticity and possible therapeutic targets for SAN dysfunction in HF patients.


Asunto(s)
Conectoma , Insuficiencia Cardíaca , Mitocondrias Cardíacas , Retículo Sarcoplasmático , Síndrome del Seno Enfermo , Nodo Sinoatrial , Animales , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ratones , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/patología , Síndrome del Seno Enfermo/patología , Síndrome del Seno Enfermo/fisiopatología , Nodo Sinoatrial/fisiopatología
2.
Cardiovasc Drugs Ther ; 37(1): 25-37, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34499283

RESUMEN

PURPOSE: Nonsteroidal anti-inflammatory drugs (NSAIDs) are among one of the most commonly prescribed medications for pain and inflammation. Diclofenac (DIC) is a commonly prescribed NSAID that is known to increase the risk of cardiovascular diseases. However, the mechanisms underlying its cardiotoxic effects remain largely unknown. In this study, we tested the hypothesis that chronic exposure to DIC increases oxidative stress, which ultimately impairs cardiovascular function. METHODS AND RESULTS: Mice were treated with DIC for 4 weeks and subsequently subjected to in vivo and in vitro functional assessments. Chronic DIC exposure resulted in not only systolic but also diastolic dysfunction. DIC treatment, however, did not alter blood pressure or electrocardiographic recordings. Importantly, treatment with DIC significantly increased inflammatory cytokines and chemokines as well as cardiac fibroblast activation and proliferation. There was increased reactive oxygen species (ROS) production in cardiomyocytes from DIC-treated mice, which may contribute to the more depolarized mitochondrial membrane potential and reduced energy production, leading to a significant decrease in sarcoplasmic reticulum (SR) Ca2+ load, Ca2+ transients, and sarcomere shortening. Using unbiased metabolomic analyses, we demonstrated significant alterations in oxylipin profiles towards inflammatory features in chronic DIC treatment. CONCLUSIONS: Together, chronic treatment with DIC resulted in severe cardiotoxicity, which was mediated, in part, by an increase in mitochondrial oxidative stress.


Asunto(s)
Diclofenaco , Cardiopatías , Ratones , Animales , Diclofenaco/toxicidad , Diclofenaco/metabolismo , Mediadores de Inflamación/metabolismo , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Cardiotoxicidad , Miocitos Cardíacos , Antiinflamatorios no Esteroideos/toxicidad
3.
J Biol Chem ; 290(8): 4663-4676, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25538241

RESUMEN

Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/fisiología , Miocitos Cardíacos/metabolismo , Transcripción Genética/fisiología , Transporte Activo de Núcleo Celular/fisiología , Animales , Canales de Calcio Tipo L/genética , Miosinas Cardíacas/biosíntesis , Miosinas Cardíacas/genética , Núcleo Celular/genética , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Humanos , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Estructura Terciaria de Proteína
4.
Circ Res ; 112(12): 1567-76, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23609114

RESUMEN

RATIONALE: Adenylyl cyclase (AC) represents one of the principal molecules in the ß-adrenergic receptor signaling pathway, responsible for the conversion of ATP to the second messenger, cAMP. AC types 5 (ACV) and 6 (ACVI) are the 2 main isoforms in the heart. Although highly homologous in sequence, these 2 proteins play different roles during the development of heart failure. Caveolin-3 is a scaffolding protein, integrating many intracellular signaling molecules in specialized areas called caveolae. In cardiomyocytes, caveolin is located predominantly along invaginations of the cell membrane known as t-tubules. OBJECTIVE: We take advantage of ACV and ACVI knockout mouse models to test the hypothesis that there is distinct compartmentalization of these isoforms in ventricular myocytes. METHODS AND RESULTS: We demonstrate that ACV and ACVI isoforms exhibit distinct subcellular localization. The ACVI isoform is localized in the plasma membrane outside the t-tubular region and is responsible for ß1-adrenergic receptor signaling-mediated enhancement of the L-type Ca(2+) current (ICa,L) in ventricular myocytes. In contrast, the ACV isoform is localized mainly in the t-tubular region where its influence on ICa,L is restricted by phosphodiesterase. We further demonstrate that the interaction between caveolin-3 with ACV and phosphodiesterase is responsible for the compartmentalization of ACV signaling. CONCLUSIONS: Our results provide new insights into the compartmentalization of the 2 AC isoforms in the regulation of ICa,L in ventricular myocytes. Because caveolae are found in most mammalian cells, the mechanism of ß- adrenergic receptor and AC compartmentalization may also be important for ß-adrenergic receptor signaling in other cell types.


Asunto(s)
Adenilil Ciclasas/metabolismo , Canales de Calcio Tipo L/metabolismo , Ventrículos Cardíacos/enzimología , Miocitos Cardíacos/enzimología , Adenilil Ciclasas/deficiencia , Adenilil Ciclasas/genética , Agonistas Adrenérgicos beta/farmacología , Secuencia de Aminoácidos , Animales , Canales de Calcio Tipo L/efectos de los fármacos , Caveolina 3/metabolismo , Membrana Celular/enzimología , Simulación por Computador , Técnica del Anticuerpo Fluorescente , Ventrículos Cardíacos/efectos de los fármacos , Isoenzimas , Isoproterenol/farmacología , Potenciales de la Membrana , Ratones , Ratones Noqueados , Microscopía Confocal , Datos de Secuencia Molecular , Miocitos Cardíacos/efectos de los fármacos , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 109(35): 14158-63, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891308

RESUMEN

Triclosan (TCS), a high-production-volume chemical used as a bactericide in personal care products, is a priority pollutant of growing concern to human and environmental health. TCS is capable of altering the activity of type 1 ryanodine receptor (RyR1), but its potential to influence physiological excitation-contraction coupling (ECC) and muscle function has not been investigated. Here, we report that TCS impairs ECC of both cardiac and skeletal muscle in vitro and in vivo. TCS acutely depresses hemodynamics and grip strength in mice at doses ≥12.5 mg/kg i.p., and a concentration ≥0.52 µM in water compromises swimming performance in larval fathead minnow. In isolated ventricular cardiomyocytes, skeletal myotubes, and adult flexor digitorum brevis fibers TCS depresses electrically evoked ECC within ∼10-20 min. In myotubes, nanomolar to low micromolar TCS initially potentiates electrically evoked Ca(2+) transients followed by complete failure of ECC, independent of Ca(2+) store depletion or block of RyR1 channels. TCS also completely blocks excitation-coupled Ca(2+) entry. Voltage clamp experiments showed that TCS partially inhibits L-type Ca(2+) currents of cardiac and skeletal muscle, and [(3)H]PN200 binding to skeletal membranes is noncompetitively inhibited by TCS in the same concentration range that enhances [(3)H]ryanodine binding. TCS potently impairs orthograde and retrograde signaling between L-type Ca(2+) and RyR channels in skeletal muscle, and L-type Ca(2+) entry in cardiac muscle, revealing a mechanism by which TCS weakens cardiac and skeletal muscle contractility in a manner that may negatively impact muscle health, especially in susceptible populations.


Asunto(s)
Antiinfecciosos Locales/toxicidad , Calcio/metabolismo , Insuficiencia Cardíaca/inducido químicamente , Contracción Muscular/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Triclosán/toxicidad , Factores de Edad , Animales , Animales Recién Nacidos , Canales de Calcio Tipo L/metabolismo , Cyprinidae , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Contracción Muscular/fisiología , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocardio/citología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Contaminantes del Agua/toxicidad
6.
J Mol Cell Cardiol ; 52(1): 264-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22062954

RESUMEN

Cardiac progenitor cells (CPCs) are multipotent cells that may offer tremendous potentials for the regeneration of injured myocardium. To expand the limited number of CPCs for effective clinical regeneration of myocardium, it is important to understand their proliferative potentials. Single-cell based assays were utilized to purify c-kit(pos) CPCs from human and mouse hearts. MicroRNA profiling identified eight differentially expressed microRNAs in CPCs from neonatal and adult hearts. Notably, the predicted protein targets were predominantly involved in cellular proliferation-related pathways. To directly test this phenotypic prediction, the developmental variance in the proliferation of CPCs was tested. Ki67 protein expression and DNA kinetics were tested in human and mouse in vivo CPCs, and doubling times were tested in primary culture of mouse CPCs. The human embryonic and mouse neonatal CPCs showed a six-fold increase in Ki67 expressing cells, a two-fold increase in the number of cells in S/G2-M phases of cell cycle, and a seven-fold increase in the doubling time in culture when compared to the corresponding adult CPCs. The over-expression of miR-17-92 increased the proliferation in adult CPCs in vivo by two-fold. In addition, the level of retinoblastoma-like 2 (Rbl2/p130) protein was two-fold higher in adult compared to neonatal-mouse CPCs. In conclusion, we demonstrate a differentially regulated cohort of microRNAs that predicts differences in cellular proliferation in CPCs during postnatal development and target microRNAs that are involved in this transition. Our study provides new insights that may enhance the utilization of adult CPCs for regenerative therapy of the injured myocardium.


Asunto(s)
Perfilación de la Expresión Génica , MicroARNs/genética , Mioblastos Cardíacos/metabolismo , Animales , Ciclo Celular/fisiología , Proliferación Celular , Separación Celular , Células Cultivadas , Análisis por Conglomerados , Humanos , Cinética , Ratones , MicroARNs/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteína p130 Similar a la del Retinoblastoma/metabolismo
7.
Circ Res ; 107(7): 851-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20689065

RESUMEN

RATIONALE: Ca(2+)-activated K(+) channels are present in a wide variety of cells. We have previously reported the presence of small conductance Ca(2+)-activated K(+) (SK or K(Ca)) channels in human and mouse cardiac myocytes that contribute functionally toward the shape and duration of cardiac action potentials. Three isoforms of SK channel subunits (SK1, SK2, and SK3) are found to be expressed. Moreover, there is differential expression with more abundant SK channels in the atria and pacemaking tissues compared with the ventricles. SK channels are proposed to be assembled as tetramers similar to other K(+) channels, but the molecular determinants driving their subunit interaction and assembly are not defined in cardiac tissues. OBJECTIVE: To investigate the heteromultimeric formation and the domain necessary for the assembly of 3 SK channel subunits (SK1, SK2, and SK3) into complexes in human and mouse hearts. METHODS AND RESULTS: Here, we provide evidence to support the formation of heteromultimeric complexes among different SK channel subunits in native cardiac tissues. SK1, SK2, and SK3 subunits contain coiled-coil domains (CCDs) in the C termini. In vitro interaction assay supports the direct interaction between CCDs of the channel subunits. Moreover, specific inhibitory peptides derived from CCDs block the Ca(2+)-activated K(+) current in atrial myocytes, which is important for cardiac repolarization. CONCLUSIONS: The data provide evidence for the formation of heteromultimeric complexes among different SK channel subunits in atrial myocytes. Because SK channels are predominantly expressed in atrial myocytes, specific ligands of the different isoforms of SK channel subunits may offer a unique therapeutic opportunity to directly modify atrial cells without interfering with ventricular myocytes.


Asunto(s)
Potenciales de Acción/fisiología , Miocitos Cardíacos/fisiología , Potasio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Secuencia de Aminoácidos , Animales , Células Cultivadas , Atrios Cardíacos/citología , Humanos , Ratones , Datos de Secuencia Molecular , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Multimerización de Proteína , Estructura Terciaria de Proteína , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/química , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Transfección
8.
Proc Natl Acad Sci U S A ; 106(43): 18402-7, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19815520

RESUMEN

The importance of proper ion channel trafficking is underpinned by a number of channel-linked genetic diseases whose defect is associated with failure to reach the cell surface. Conceptually, it is reasonable to suggest that the function of ion channels depends critically on the precise subcellular localization and the number of channel proteins on the cell surface membrane, which is determined jointly by the secretory and endocytic pathways. Yet the precise mechanisms of the entire ion channel trafficking pathway remain unknown. Here, we directly demonstrate that proper membrane localization of a small-conductance Ca(2+)-activated K(+) channel (SK2 or K(Ca)2.2) is dependent on its interacting protein, alpha-actinin2, a major F-actin crosslinking protein. SK2 channel localization on the cell-surface membrane is dynamically regulated, and one of the critical steps includes the process of cytoskeletal anchoring of SK2 channel by its interacting protein, alpha-actinin2, as well as endocytic recycling via early endosome back to the cell membrane. Consequently, alteration of these components of SK2 channel recycling results in profound changes in channel surface expression. The importance of our findings may transcend the area of K(+) channels, given that similar cytoskeletal interaction and anchoring may be critical for the membrane localization of other ion channels in neurons and other excitable cells.


Asunto(s)
Actinina/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Actinina/genética , Actinas/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Células Cultivadas , Motivos EF Hand , Endosomas/metabolismo , Humanos , Ratones , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Unión Proteica , Transporte de Proteínas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/química
9.
STAR Protoc ; 3(2): 101301, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35463464

RESUMEN

Intracellular pH (pHi) plays critical roles in the regulation of cardiac function. Methods and techniques for cardiac pHi measurement have continued to evolve since early 1960s. Fluorescent microscopy is the most recently developed technique with several advantages over other techniques including higher spatial and temporal resolutions, and feasibility for contracting cell measurement. Here, we describe detailed methods for mouse cardiomyocyte isolation, and simultaneous measurement and quantification of pHi and sarcomere length in contracting cardiomyocytes. For complete details on the use and execution of this protocol, please refer to Lyu et al. (2022).


Asunto(s)
Miocitos Cardíacos , Animales , Concentración de Iones de Hidrógeno , Ratones , Miocitos Cardíacos/fisiología
10.
Cardiovasc Res ; 118(1): 267-281, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33125066

RESUMEN

AIMS: One of the hallmarks of myocardial infarction (MI) is excessive inflammation. During an inflammatory insult, damaged endothelial cells shed their glycocalyx, a carbohydrate-rich layer on the cell surface which provides a regulatory interface to immune cell adhesion. Selectin-mediated neutrophilia occurs as a result of endothelial injury and inflammation. We recently designed a novel selectin-targeting glycocalyx mimetic (termed DS-IkL) capable of binding inflamed endothelial cells. This study examines the capacity of DS-IkL to limit neutrophil binding and platelet activation on inflamed endothelial cells, as well as the cardioprotective effects of DS-IkL after acute myocardial infarction. METHODS AND RESULTS: In vitro, DS-IkL diminished neutrophil interactions with both recombinant selectin and inflamed endothelial cells, and limited platelet activation on inflamed endothelial cells. Our data demonstrated that DS-IkL localized to regions of vascular inflammation in vivo after 45 min of left anterior descending coronary artery ligation-induced MI. Further, findings from this study show DS-IkL treatment had short- and long-term cardioprotective effects after ischaemia/reperfusion of the left anterior descending coronary artery. Mice treated with DS-IkL immediately after ischaemia/reperfusion and 24 h later exhibited reduced neutrophil extravasation, macrophage accumulation, fibroblast and endothelial cell proliferation, and fibrosis compared to saline controls. CONCLUSIONS: Our findings suggest that DS-IkL has great therapeutic potential after MI by limiting reperfusion injury induced by the immune response.


Asunto(s)
Antiinflamatorios/farmacología , Selectina E/metabolismo , Células Endoteliales/efectos de los fármacos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Activación Neutrófila/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Animales , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Fibrosis , Humanos , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/inmunología , Miocardio/metabolismo , Miocardio/patología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Activación Plaquetaria/efectos de los fármacos , Transducción de Señal
11.
iScience ; 25(1): 103624, 2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35005560

RESUMEN

The mammalian heart beats incessantly with rhythmic mechanical activities generating acids that need to be buffered to maintain a stable intracellular pH (pHi) for normal cardiac function. Even though spatial pHi non-uniformity in cardiomyocytes has been documented, it remains unknown how pHi is regulated to match the dynamic cardiac contractions. Here, we demonstrated beat-to-beat intracellular acidification, termed pHi transients, in synchrony with cardiomyocyte contractions. The pHi transients are regulated by pacing rate, Cl-/HCO3 - transporters, pHi buffering capacity, and ß-adrenergic signaling. Mitochondrial electron-transport chain inhibition attenuates the pHi transients, implicating mitochondrial activity in sculpting the pHi regulation. The pHi transients provide dynamic alterations of H+ transport required for ATP synthesis, and a decrease in pHi may serve as a negative feedback to cardiac contractions. Current findings dovetail with the prevailing three known dynamic systems, namely electrical, Ca2+, and mechanical systems, and may reveal broader features of pHi handling in excitable cells.

12.
Heart Rhythm ; 19(2): 281-292, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34634443

RESUMEN

BACKGROUND: Long QT syndrome (LQTS) is a hereditary disease that predisposes patients to life-threatening cardiac arrhythmias and sudden cardiac death. Our previous study of the human ether-à-go-go related gene (hERG)-encoded K+ channel (Kv11.1) supports an association between hERG and RING finger protein 207 (RNF207) variants in aggravating the onset and severity of LQTS, specifically T613M hERG (hERGT613M) and RNF207 frameshift (RNF207G603fs) mutations. However, the underlying mechanistic underpinning remains unknown. OBJECTIVE: The purpose of the present study was to test the role of RNF207 in the function of hERG-encoded K+ channel subunits. METHODS: Whole-cell patch-clamp experiments were performed in human embryonic kidney (HEK 293) cells and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) together with immunofluorescent confocal and high resolution microscopy, auto-ubiquitinylation assays, and co-immunoprecipitation experiments to test the functional interactions between hERG and RNF207. RESULTS: Here, we demonstrated that RNF207 serves as an E3 ubiquitin ligase and targets misfolded hERGT613M proteins for degradation. RNF207G603fs exhibits decreased activity and hinders the normal degradation pathway; this increases the levels of hERGT613M subunits and their dominant-negative effect on the wild-type subunits, ultimately resulting in decreased current density. Similar findings are shown for hERGA614V, a known dominant-negative mutant subunit. Finally, the presence of RNF207G603fs with hERGT613M results in significantly prolonged action potential durations and reduced hERG current in human-induced pluripotent stem cell-derived cardiomyocytes. CONCLUSION: Our study establishes RNF207 as an interacting protein serving as a ubiquitin ligase for hERG-encoded K+ channel subunits. Normal function of RNF207 is critical for the quality control of hERG subunits and consequently cardiac repolarization. Moreover, our study provides evidence for protein quality control as a new paradigm in life-threatening cardiac arrhythmias in patients with LQTS.


Asunto(s)
Canal de Potasio ERG1/genética , Síndrome de QT Prolongado/genética , Ubiquitina-Proteína Ligasas/genética , Células HEK293/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp
13.
JCI Insight ; 7(22)2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36509290

RESUMEN

Sinoatrial node (SAN) cells are the heart's primary pacemaker. Their activity is tightly regulated by ß-adrenergic receptor (ß-AR) signaling. Adenylyl cyclase (AC) is a key enzyme in the ß-AR pathway that catalyzes the production of cAMP. There are current gaps in our knowledge regarding the dominant AC isoforms and the specific roles of Ca2+-activated ACs in the SAN. The current study tests the hypothesis that distinct AC isoforms are preferentially expressed in the SAN and compartmentalize within microdomains to orchestrate heart rate regulation during ß-AR signaling. In contrast to atrial and ventricular myocytes, SAN cells express a diverse repertoire of ACs, with ACI as the predominant Ca2+-activated isoform. Although ACI-KO (ACI-/-) mice exhibit normal cardiac systolic or diastolic function, they experience SAN dysfunction. Similarly, SAN-specific CRISPR/Cas9-mediated gene silencing of ACI results in sinus node dysfunction. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated 4 (HCN4) channels form functional microdomains almost exclusively with ACI, while ryanodine receptor and L-type Ca2+ channels likely compartmentalize with ACI and other AC isoforms. In contrast, there were no significant differences in T-type Ca2+ and Na+ currents at baseline or after ß-AR stimulation between WT and ACI-/- SAN cells. Due to its central characteristic feature as a Ca2+-activated isoform, ACI plays a unique role in sustaining the rise of local cAMP and heart rates during ß-AR stimulation. The findings provide insights into the critical roles of the Ca2+-activated isoform of AC in sustaining SAN automaticity that is distinct from contractile cardiomyocytes.


Asunto(s)
Adenilil Ciclasas , Nodo Sinoatrial , Animales , Ratones , Nodo Sinoatrial/metabolismo , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Calcio/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Isoformas de Proteínas/metabolismo
14.
J Mol Cell Cardiol ; 50(1): 194-202, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20951705

RESUMEN

Atrioventricular node (AV node) is the hub where electrical input from the atria is propagated and conveyed to the ventricles. Despite its strategic position and role in governing impulse conduction between atria and ventricles, there is paucity of data regarding the contribution of specific ion channels to the function of the AV node. Here, we examined the roles of Ca(v)1.3 L-type Ca(2+) channel in AV node by taking advantage of a mouse model with null mutation of Ca(v)1.3 (Ca(v)1.3(-/-)). Ca(v)1.3 null mutant mice show evidence of AV node dysfunction with AV block, suggesting the tissue-specific function of the Ca(v)1.3 channel. In keeping with this assertion, we demonstrate that Ca(v)1.3 isoform is highly expressed in the isolated AV node cells. Furthermore, AV node isolated from Ca(v)1.3 null mutant mice show a significant decrease in the firing frequency of spontaneous action potentials suggesting that Ca(v)1.3 L-type Ca(2+) channel plays significant roles in the automaticity of the AV node. Because of the distinct voltage-dependence of Ca(v)1.2 and Ca(v)1.3 Ca(2+) channels, Ca(v)1.2 alone does not suffice to maintain normal AV node function. Ca(v)1.3 currents activate at more hyperpolarizing voltage compared to Ca(v)1.2 currents. Consequently, Ca(v)1.2 Ca(2+) channel cannot functionally substitute for Ca(v)1.3 isoform in the AV node of Ca(v)1.3 null mutant mice. Thus, our study demonstrates that the distinct biophysical properties of Ca(v)1.3 Ca(2+) channel play critical roles in the firing frequency of AV node tissues.


Asunto(s)
Nodo Atrioventricular/metabolismo , Canales de Calcio Tipo L/metabolismo , Potenciales de Acción/fisiología , Animales , Inmunohistoquímica , Ratones , Ratones Mutantes , Microscopía Confocal , Modelos Teóricos , Técnicas de Placa-Clamp
15.
Am J Physiol Heart Circ Physiol ; 300(2): H617-26, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21131481

RESUMEN

Endothelin-1 (ET-1) and activation of protein kinase C (PKC) have been implicated in alterations of myocyte function in cardiac hypertrophy and heart failure. Changes in cellular Ca2+ handling and electrophysiological properties also occur in these states and may contribute to mechanical dysfunction and arrhythmias. While ET-1 or PKC stimulation induces cellular hypertrophy in cultured neonatal rat ventricular myocytes (NRVMs), a system widely used in studies of hypertrophic signaling, there is little data about electrophysiological changes. Here we studied the effects of ET-1 (100 nM) or the PKC activator phorbol 12-myristate 13-acetate (PMA, 1 µM) on ionic currents in NRVMs. The acute effects of PMA or ET-1 (≤30 min) were small or insignificant. However, PMA or ET-1 exposure for 48-72 h increased cell capacitance by 100 or 25%, respectively, indicating cellular hypertrophy. ET-1 also slightly increased Ca2+ current density (T and L type). Na+/Ca2+ exchange current was increased by chronic pretreatment with either PMA or ET-1. In contrast, transient outward and delayed rectifier K+ currents were strongly downregulated by PMA or ET-1 pretreatment. Inward rectifier K+ current tended toward a decrease at larger negative potential, but time-independent outward K+ current was unaltered by either treatment. The enhanced inward and reduced outward currents also result in action potential prolongation after PMA or ET-1 pretreatment. We conclude that chronic PMA or ET-1 exposure in cultured NRVMs causes altered functional expression of cardiac ion currents, which mimic electrophysiological changes seen in whole animal and human hypertrophy and heart failure.


Asunto(s)
Canales de Calcio/biosíntesis , Endotelina-1/farmacología , Miocitos Cardíacos/metabolismo , Canales de Potasio/biosíntesis , Intercambiador de Sodio-Calcio/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Western Blotting , Canales de Calcio/efectos de los fármacos , Tamaño de la Célula , Células Cultivadas , Fenómenos Electrofisiológicos , Técnicas In Vitro , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Fosforilación , Canales de Potasio/efectos de los fármacos , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley
16.
STAR Protoc ; 2(4): 100891, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34704077

RESUMEN

Prestin (Slc26a5) is a motor protein previously considered to be expressed exclusively in outer hair cells (OHCs) of the inner ear. However, we recently identified the functional expression of prestin in the heart. Nonlinear capacitance (NLC) measurement in OHCs is used to evaluate the signature function of prestin, which exhibits membrane potential-dependent conformational changes. Here, we describe detailed recording techniques and quantification methods for NLC to evaluate the prestin function in mouse ventricular myocytes. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2021).


Asunto(s)
Potenciales de la Membrana/fisiología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp/métodos , Animales , Células Cultivadas , Capacidad Eléctrica , Ratones
17.
Cell Rep ; 35(5): 109097, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33951436

RESUMEN

Cardiac cells generate and amplify force in the context of cardiac load, yet the membranous sheath enclosing the muscle fibers-the sarcolemma-does not experience displacement. That the sarcolemma sustains beat-to-beat pressure changes without experiencing significant distortion is a muscle-contraction paradox. Here, we report that an elastic element-the motor protein prestin (Slc26a5)-serves to amplify actin-myosin force generation in mouse and human cardiac myocytes, accounting partly for the nonlinear capacitance of cardiomyocytes. The functional significance of prestin is underpinned by significant alterations of cardiac contractility in Prestin-knockout mice. Prestin was previously considered exclusive to the inner ear's outer hair cells; however, our results show that prestin serves a broader cellular motor function.


Asunto(s)
Corazón/fisiología , Proteínas Motoras Moleculares/metabolismo , Transportadores de Sulfato/metabolismo , Animales , Humanos , Ratones
18.
Am J Physiol Heart Circ Physiol ; 299(5): H1459-67, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20709863

RESUMEN

Adenylyl cyclase (AC) is the principal effector molecule in the ß-adrenergic receptor pathway. AC(V) and AC(VI) are the two predominant isoforms in mammalian cardiac myocytes. The disparate roles among AC isoforms in cardiac hypertrophy and progression to heart failure have been under intense investigation. Specifically, the salutary effects resulting from the disruption of AC(V) have been established in multiple models of cardiomyopathy. It has been proposed that a continual activation of AC(V) through elevated levels of protein kinase C could play an integral role in mediating a hypertrophic response leading to progressive heart failure. Elevated protein kinase C is a common finding in heart failure and was demonstrated in murine cardiomyopathy from cardiac-specific overexpression of G(αq) protein. Here we assessed whether the disruption of AC(V) expression can improve cardiac function, limit electrophysiological remodeling, or improve survival in the G(αq) mouse model of heart failure. We directly tested the effects of gene-targeted disruption of AC(V) in transgenic mice with cardiac-specific overexpression of G(αq) protein using multiple techniques to assess the survival, cardiac function, as well as structural and electrical remodeling. Surprisingly, in contrast to other models of cardiomyopathy, AC(V) disruption did not improve survival or cardiac function, limit cardiac chamber dilation, halt hypertrophy, or prevent electrical remodeling in G(αq) transgenic mice. In conclusion, unlike other established models of cardiomyopathy, disrupting AC(V) expression in the G(αq) mouse model is insufficient to overcome several parallel pathophysiological processes leading to progressive heart failure.


Asunto(s)
Adenilil Ciclasas/metabolismo , Cardiomiopatías/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Adenilil Ciclasas/genética , Animales , Bradicardia/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnicas Electrofisiológicas Cardíacas , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Proteína Quinasa C/metabolismo
19.
Circ Res ; 102(4): 465-71, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18096820

RESUMEN

Since the first description of the anatomical atrioventricular nodes (AVNs), a large number of studies have provided insights into the heterogeneity of the structure as well as a repertoire of ion channel proteins that govern this complex conduction pathway between the atria and ventricles. These studies have revealed the intricate organization of multiple nodal and nodal-like myocytes contributing to the unique electrophysiology of the AVN in health and diseases. On the other hand, information regarding the contribution of specific ion channels to the function of the AVN remains incomplete. We reason that the identification of AVN-specific ion channels may provide a more direct and rational design of therapeutic target in the control of AVN conduction in atrial flutter/fibrillation, one of the most common arrhythmias seen clinically. In this study, we took advantage of 2 genetically altered mouse models with overexpression or null mutation of 1 of a small conductance Ca2+-activated K+ channel isoform, SK2 channel, and demonstrated robust phenotypes of AVN dysfunction in these experimental models. Overexpression of SK2 channels results in the shortening of the spontaneous action potentials of the AVN cells and an increase in the firing frequency. On the other hand, ablation of the SK2 channel results in the opposite effects on the spontaneous action potentials of the AVN. Furthermore, we directly documented the expression of SK2 channel in mouse AVN using multiple techniques. The new insights may have important implications in providing novel drug targets for the modification of AVN conduction in the treatment of atrial arrhythmias.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Nodo Atrioventricular/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Potenciales de Acción/fisiología , Animales , Arritmias Cardíacas/genética , Nodo Atrioventricular/citología , Electrocardiografía , Técnica del Anticuerpo Fluorescente , Expresión Génica/fisiología , Ratones , Ratones Transgénicos , Microscopía Confocal , Técnicas de Placa-Clamp , Nodo Sinoatrial/citología , Nodo Sinoatrial/fisiología
20.
Sci Adv ; 6(15): eaba1104, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32285007

RESUMEN

The mammalian cochlea relies on active electromotility of outer hair cells (OHCs) to resolve sound frequencies. OHCs use ionic channels and somatic electromotility to achieve the process. It is unclear, though, how the kinetics of voltage-gated ionic channels operate to overcome extrinsic viscous drag on OHCs at high frequency. Here, we report ultrafast electromechanical gating of clustered Kv7.4 in OHCs. Increases in kinetics and sensitivity resulting from cooperativity among clustered-Kv7.4 were revealed, using optogenetics strategies. Upon clustering, the half-activation voltage shifted negative, and the speed of activation increased relative to solitary channels. Clustering also rendered Kv7.4 channels mechanically sensitive, confirmed in consolidated Kv7.4 channels at the base of OHCs. Kv7.4 clusters provide OHCs with ultrafast electromechanical channel gating, varying in magnitude and speed along the cochlea axis. Ultrafast Kv7.4 gating provides OHCs with a feedback mechanism that enables the cochlea to overcome viscous drag and resolve sounds at auditory frequencies.


Asunto(s)
Fenómenos Electrofisiológicos , Células Ciliadas Auditivas Externas/citología , Células Ciliadas Auditivas Externas/fisiología , Canales de Potasio KCNQ/metabolismo , Fenómenos Mecánicos , Animales , Línea Celular , Cóclea/fisiología , Humanos , Activación del Canal Iónico , Ratones , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA