Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Allied Health ; 53(1): 3-9, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38430490

RESUMEN

BACKGROUND: The ASAHP established the Clinical Education Task Force (CETF) in 2017 to identify strategies for clinical education. Implementing the CETF recommendations requires continuous collaboration between healthcare industry and academic partners. AIM: ASAHP Regional Summits were planned and implemented to offer an active learning environment for stakeholders, strengthen translational skills, identify gaps in interprofessional collaborative practice (IPC), and create lasting networking opportunities. METHODS: The Regional Summits were organized in a standard format across three hub sites. During a virtual "Harvest" session all sites were video linked to continue the local dialogue on a national level. Outcomes were analyzed using mixed methods, including pre- and post-session surveys quantitative methods. Notes from table discussions were analyzed using a qualitative approach. RESULTS: Qualitative results offered a rich dataset from the industry and academic perspective to provide a better understanding how the CETF recommendations are being understood. Ideas for future action and partnerships were identified. Various regions contributed insights that reflect unique environments. CONCLUSIONS: The ASAHP Collaborative Stakeholder Engagement Model offers a robust and reproducible active adult learning model for IPC that can lead to change and continued engagement. These findings identify opportunities for deepening the connections made through regional hubs.


Asunto(s)
Atención a la Salud , Participación de los Interesados , Adulto , Humanos , Aprendizaje Basado en Problemas
2.
J Hosp Palliat Nurs ; 22(4): 305-311, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32568939

RESUMEN

The purpose of this quality improvement project was to evaluate a statewide initiative promoting Advance Care Planning (ACP) to educate and support multidisciplinary ACP educators and provide tools to start ACP conversations in a predominantly rural state of the Upper Midwest. Individual objectives were to (1) motivate people of different professions and backgrounds to support the vision and (2) implement a system to educate and maintain a pipeline of ACP educators in appropriate methodologies to enable ACP in distant communities. The Advance Care Planning: Quality Conversations coalition was formed in 2015 to improve health care across the life span. The Reach-Effectiveness-Adoption-Implementation-Maintenance framework was applied to evaluate the project. Outcome variables were measured before, during, and after program implementation through service statistics and a questionnaire. Participation in the coalition's membership team between September 2015 and September 2019 ranged from 18 to 36 with a median of 27 and mode of 27. At least 20 different professions were represented. The coalition provided funds for educating 9 ACP instructors and 180 facilitators according to the Respecting Choices-First Steps ACP program. The coalition's mission has generated sustained interest for 4 years. Key elements and obstacles to implementing a statewide coalition were identified.


Asunto(s)
Planificación Anticipada de Atención/normas , Redes Comunitarias/tendencias , Docentes de Enfermería/tendencias , Planificación Anticipada de Atención/tendencias , Conducta Cooperativa , Humanos , Estudios Longitudinales , South Dakota
3.
S D Med ; 73(4): 171-177, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32445305

RESUMEN

INTRODUCTION: Competency in and understanding of the factors impacting cardiopulmonary resuscitation (CPR) are key to emergency medicine. The purpose of this study was to assess the impact of the automated LUCAS-2 device on survival to emergency department (ED) compared to manual CPR as part of the EMS response using a large data set collected in a mostly rural U.S. state. METHODS: We conducted a retrospective analysis of South Dakota's electronic Patient Care Reports (ePCR) collected from Jan. 1, 2013 through Dec. 31, 2015. The primary outcome measure was survival to ED. RESULTS: A mechanical piston device (LUCAS-2), was utilized in 260 (15 percent) of 1,781 total cases during this period. The odds for survival to ED were calculated and compared between manual and LUCAS-2-assisted CPR. The odds ratio for survival to ED using compressions alone was 3.94 compared to LUCAS-2 and those results persisted after adjusting for significant covariates. DISCUSSION: Despite hemodynamic benefits associated with the LUCAS-2 device in the laboratory and in other settings, this and other studies indicate that compression-only CPR outperforms automation-assisted CPR during OHCA. However, the data also suggest that enhanced training of emergency providers to improve response times and levels of expertise with the equipment may improve the outcomes associated with the LUCAS-2 and it is recommended that further training should be pursued.


Asunto(s)
Reanimación Cardiopulmonar , Servicios Médicos de Urgencia , Paro Cardíaco Extrahospitalario , Humanos , Paro Cardíaco Extrahospitalario/terapia , Estudios Retrospectivos , South Dakota
4.
Holist Nurs Pract ; 33(4): 197-203, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31192831

RESUMEN

This study tests the hypothesis that yoga breathing (pranayama) improves lung function in healthy volunteers during a 6-week protocol. A randomized controlled pilot study demonstrated an improvement in peak expiratory flow rate and forced expiratory volume. The easy-to-learn approach can be translated to the inpatient and outpatient settings.


Asunto(s)
Ejercicios Respiratorios/normas , Respiración , Pruebas de Función Respiratoria/estadística & datos numéricos , Yoga , Adulto , Ejercicios Respiratorios/métodos , Femenino , Voluntarios Sanos/estadística & datos numéricos , Humanos , Pulmón/fisiología , Masculino , Proyectos Piloto , Pruebas de Función Respiratoria/métodos , South Dakota
5.
J Mol Cell Cardiol ; 132: 24-35, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31077706

RESUMEN

A frameshift (fs) mutation in the natriuretic peptide precursor A (NPPA) gene, encoding a mutant atrial natriuretic peptide (Mut-ANP), has been linked with familial atrial fibrillation (AF) but the underlying mechanisms by which the mutation causes AF remain unclear. We engineered 2 transgenic (TG) mouse lines expressing the wild-type (WT)-NPPA gene (H-WT-NPPA) and the human fs-Mut-NPPA gene (H-fsMut-NPPA) to test the hypothesis that mice overexpressing the human NPPA mutation are more susceptible to AF and elucidate the underlying electrophysiologic and molecular mechanisms. Transthoracic echocardiography and surface electrocardiography (ECG) were performed in H-fsMut-NPPA, H-WT-NPPA, and Non-TG mice. Invasive electrophysiology, immunohistochemistry, Western blotting and patch clamping of membrane potentials were performed. To examine the role of the Mut-ANP in ion channel remodeling, we measured plasma cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) levels and protein kinase A (PKA) activity in the 3 groups of mice. In H-fsMut-NPPA mice mean arterial pressure (MAP) was reduced when compared to H-WT-NPPA and Non-TG mice. Furthermore, injection of synthetic fs-Mut-ANP lowered the MAP in H-WT-NPPA and Non-TG mice while synthetic WT-ANP had no effect on MAP in the 3 groups of mice. ECG characterization revealed significantly prolonged QRS duration in H-fsMut-NPPA mice when compared to the other two groups. Trans-Esophageal (TE) atrial pacing of H-fsMut-NPPA mice showed increased AF burden and AF episodes when compared with H-WT-NPPA or Non-TG mice. The cardiac Na+ (NaV1.5) and Ca2+ (CaV1.2/CaV1.3) channel expression and currents (INa, ICaL) and action potential durations (APD90/APD50/APD20) were significantly reduced in H-fsMut-NPPA mice while the rectifier K+ channel current (IKs) was markedly increased when compared to the other 2 groups of mice. In addition, plasma cGMP levels were only increased in H-fsMut-NPPA mice with a corresponding reduction in plasma cAMP levels and PKA activity. In summary, we showed that mice overexpressing an AF-linked NPPA mutation are more prone to develop AF and this risk is mediated in part by remodeling of the cardiac Na+, Ca2+ and K+ channels creating an electrophysiologic substrate for reentrant AF.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial/etiología , Factor Natriurético Atrial/genética , Mutación del Sistema de Lectura , Atrios Cardíacos/fisiopatología , Miocitos Cardíacos/patología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Factor Natriurético Atrial/metabolismo , Fenómenos Electrofisiológicos , Humanos , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo
6.
S D Med ; 71(2): 72-79, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29990416

RESUMEN

BACKGROUND: Out-of-hospital cardiac arrest (OHCA) is the cessation of electric or mechanical activity of the heart, confirmed by absence of circulation. Survival to hospital dismissal rates have remained low nationwide despite considerable effort to improve treatment. Current initiatives seek systems approaches that optimize care at each point along the "chain of survival." Systems approaches rely on the availability of robust data sets to understand and control variables that can be highly interdependent. The current report seeks to provide a source of reliable data of OHCA for South Dakota. METHODS: Using the "Utstein" guidelines for reviewing and reporting OHCA resuscitations issued by the American Heart Association in 2014, we analyzed the EMS data that were captured by ePCR between January 1, 2013 and December 31, 2015. Inclusion criteria were 911 calls in 2013-2015, where first impression of the call was cardiac arrest. Exclusion criteria were inconsistent and missing data. RESULTS: There were 1,781 OHCA in the ePCR, and 1,280 cases had survival information, with 378 victims surviving to ED. Overall, SD OHCA rates were lower than those reported nationally. Survival was the highest in patients with a shockable rhythm and when victim received bystander CPR. The odds for survival were greater if the arrest took place in an urban setting compared to a rural setting and if the victim received care from an EMS unit that did not have a "hardship" designation. DISCUSSION: Recommendations for future efforts include: (1) Develop and employ quality improvement methodologies for data collection and utilization to minimize the impact of poor or missing data, (2) Assess the educational and training needs of the EMS staff to properly collect, analyze, and develop actionable outputs, (3) Provide public training to include hands-only CPR and PulsePoint.


Asunto(s)
Ciencia de los Datos , Servicios Médicos de Urgencia , Paro Cardíaco Extrahospitalario/epidemiología , Población Rural/estadística & datos numéricos , Reanimación Cardiopulmonar , Humanos , South Dakota/epidemiología
7.
Stem Cells ; 32(7): 1774-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24648383

RESUMEN

The bone morphogenetic protein antagonist Gremlin 2 (Grem2) is required for atrial differentiation and establishment of cardiac rhythm during embryonic development. A human Grem2 variant has been associated with familial atrial fibrillation, suggesting that abnormal Grem2 activity causes arrhythmias. However, it is not known how Grem2 integrates into signaling pathways to direct atrial cardiomyocyte differentiation. Here, we demonstrate that Grem2 expression is induced concurrently with the emergence of cardiovascular progenitor cells during differentiation of mouse embryonic stem cells (ESCs). Grem2 exposure enhances the cardiogenic potential of ESCs by 20-120-fold, preferentially inducing genes expressed in atrial myocytes such as Myl7, Nppa, and Sarcolipin. We show that Grem2 acts upstream to upregulate proatrial transcription factors CoupTFII and Hey1 and downregulate atrial fate repressors Irx4 and Hey2. The molecular phenotype of Grem2-induced atrial cardiomyocytes was further supported by induction of ion channels encoded by Kcnj3, Kcnj5, and Cacna1d genes and establishment of atrial-like action potentials shown by electrophysiological recordings. We show that promotion of atrial-like cardiomyocytes is specific to the Gremlin subfamily of BMP antagonists. Grem2 proatrial differentiation activity is conveyed by noncanonical BMP signaling through phosphorylation of JNK and can be reversed by specific JNK inhibitors, but not by dorsomorphin, an inhibitor of canonical BMP signaling. Taken together, our data provide novel mechanistic insights into atrial cardiomyocyte differentiation from pluripotent stem cells and will assist the development of future approaches to study and treat arrhythmias.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Sistema de Señalización de MAP Quinasas , Proteínas/fisiología , Animales , Células Cultivadas , Citocinas , Atrios Cardíacos/citología , Ratones , Miocitos Cardíacos/fisiología
8.
Heart Rhythm ; 10(1): 61-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23010577

RESUMEN

BACKGROUND: Long QT syndrome (LQTS) is characterized by prolonged ventricular repolarization and variable clinical course with arrhythmia-related syncope and sudden death. Mutations in the nonpore region of the LQTS-associated KCNH2 gene (also known as hERG) are mostly associated with coassembly or trafficking abnormalities, resulting in haplotype insufficiency and milder clinical phenotypes compared with mutations in the pore domain. OBJECTIVE: To investigate the effect of a nonpore mutation on the channel current, which was identified from an LQTS family with severe clinical phenotypes. METHODS: Two members of a Japanese family with LQTS were searched for mutations in KCNQ1, KCNH2, SCN5A, KCNE1, KCNE2, and KCNJ2 genes by using automated DNA sequencing. We characterized the electrophysiological properties and glycosylation pattern of the mutant channels by using patch clamp recording and Western blot analysis. RESULTS: In the LQTS patient with torsades de pointes and cardiopulmonary arrest, we identified the novel T473P mutation in the transmembrane nonpore region of KCNH2. The proband's father carried the same mutation and showed prolonged corrected QT interval and frequent torsades de pointes in the presence of hypokalemia following the administration of garenoxacin. Patch clamp analysis in heterologous cells showed that hERG T473P channels generated no current and exhibited a dominant negative effect when coexpressed with wild-type protein. Only incompletely glycosylated hERG T473P channels were observed by using Western blot analysis, suggesting impaired trafficking. CONCLUSIONS: These results demonstrated that a trafficking-deficient mutation in the transmembrane nonpore region of KCNH2 causes a dominant negative effect and a severe clinical course in affected patients.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Síndrome de QT Prolongado/genética , Mutación , Adulto , Western Blotting , Canal de Potasio ERG1 , Electrocardiografía , Humanos , Masculino , Linaje , Fenotipo
9.
J Biol Chem ; 287(47): 39613-25, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23033485

RESUMEN

The human Ether-à-go-go-related gene (hERG)-encoded K(+) current, I(Kr) is essential for cardiac repolarization but is also a source of cardiotoxicity because unintended hERG inhibition by diverse pharmaceuticals can cause arrhythmias and sudden cardiac death. We hypothesized that a small molecule that diminishes I(Kr) block by a known hERG antagonist would constitute a first step toward preventing hERG-related arrhythmias and facilitating drug discovery. Using a high-throughput assay, we screened a library of compounds for agents that increase the IC(70) of dofetilide, a well characterized hERG blocker. One compound, VU0405601, with the desired activity was further characterized. In isolated, Langendorff-perfused rabbit hearts, optical mapping revealed that dofetilide-induced arrhythmias were reduced after pretreatment with VU0405601. Patch clamp analysis in stable hERG-HEK cells showed effects on current amplitude, inactivation, and deactivation. VU0405601 increased the IC(50) of dofetilide from 38.7 to 76.3 nM. VU0405601 mitigates the effects of hERG blockers from the extracellular aspect primarily by reducing inactivation, whereas most clinically relevant hERG inhibitors act at an inner pore site. Structure-activity relationships surrounding VU0405601 identified a 3-pyridiyl and a naphthyridine ring system as key structural components important for preventing hERG inhibition by multiple inhibitors. These findings indicate that small molecules can be designed to reduce the sensitivity of hERG to inhibitors.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/tratamiento farmacológico , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/metabolismo , Naftiridinas/química , Naftiridinas/farmacología , Fenetilaminas/efectos adversos , Bloqueadores de los Canales de Potasio/efectos adversos , Piridinas/química , Piridinas/farmacología , Sulfonamidas/efectos adversos , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Proteínas Musculares/genética , Miocardio/metabolismo , Miocardio/patología , Fenetilaminas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Conejos , Relación Estructura-Actividad , Sulfonamidas/farmacología
10.
Circulation ; 124(9): 1001-11, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21824921

RESUMEN

BACKGROUND: The D1275N SCN5A mutation has been associated with a range of unusual phenotypes, including conduction disease and dilated cardiomyopathy, as well as atrial and ventricular tachyarrhythmias. However, when D1275N is studied in heterologous expression systems, most studies show near-normal sodium channel function. Thus, the relationship of the variant to the clinical phenotypes remains uncertain. METHODS AND RESULTS: We identified D1275N in a patient with atrial flutter, atrial standstill, conduction disease, and sinus node dysfunction. There was no major difference in biophysical properties between wild-type and D1275N channels expressed in Chinese hamster ovary cells or tsA201 cells in the absence or presence of ß1 subunits. To determine D1275N function in vivo, the Scn5a locus was modified to knock out the mouse gene, and the full-length wild-type (H) or D1275N (DN) human SCN5A cDNAs were then inserted at the modified locus by recombinase mediated cassette exchange. Mice carrying the DN allele displayed slow conduction, heart block, atrial fibrillation, ventricular tachycardia, and a dilated cardiomyopathy phenotype, with no significant fibrosis or myocyte disarray on histological examination. The DN allele conferred gene-dose-dependent increases in SCN5A mRNA abundance but reduced sodium channel protein abundance and peak sodium current amplitudes (H/H, 41.0±2.9 pA/pF at -30 mV; DN/H, 19.2±3.1 pA/pF, P<0.001 vs. H/H; DN/DN, 9.3±1.1 pA/pF, P<0.001 versus H/H). CONCLUSIONS: Although D1275N produces near-normal currents in multiple heterologous expression experiments, our data establish this variant as a pathological mutation that generates conduction slowing, arrhythmias, and a dilated cardiomyopathy phenotype by reducing cardiac sodium current.


Asunto(s)
Aleteo Atrial/genética , Cardiomiopatía Dilatada/genética , Mutación Missense , Síndrome del Seno Enfermo/genética , Canales de Sodio/genética , Animales , Aleteo Atrial/terapia , Células CHO , Cricetinae , Cricetulus , Desfibriladores Implantables , Electrocardiografía , Humanos , Masculino , Ratones , Ratones Transgénicos , Contracción Miocárdica/genética , Canal de Sodio Activado por Voltaje NAV1.5 , Síndrome del Seno Enfermo/terapia , Resultado del Tratamiento , Adulto Joven
11.
J Mol Cell Cardiol ; 50(1): 50-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20950623

RESUMEN

The acquired long QT syndrome (aLQTS) is frequently associated with extrinsic and intrinsic risk factors including therapeutic agents that inadvertently inhibit the KCNH2 K(+) channel that underlies the repolarizing I(Kr) current in the heart. Previous reports demonstrated that K(+) channel regulator 1 (KCR1) diminishes KCNH2 drug sensitivity and may protect susceptible patients from developing aLQTS. Here, we describe a novel variant of KCR1 (E33D) isolated from a patient with ventricular fibrillation and significant QT prolongation. We recorded the KCNH2 current (I(KCNH2)) from CHO-K1 cells transfected with KCNH2 plus wild type (WT) or mutant KCR1 cDNA, using whole cell patch-clamp techniques and assessed the development of I(KCNH2) inhibition in response to well-characterized KCNH2 inhibitors. Unlike KCR1 WT, the E33D variant did not protect KCNH2 from the effects of class I antiarrhythmic drugs such as quinidine or class III antiarrhythmic drugs including dofetilide and sotalol. The remaining current of the KCNH2 WT+KCR1 E33D channel after 100 pulses in the presence of each drug was similar to that of KCNH2 alone. Simulated conditions of hypokalemia (1mM [K(+)](o)) produced no significant difference in the fraction of the current that was protected from dofetilide inhibition with KCR1 WT or E33D. The previously described α-glucosyltransferase activity of KCR1 was found to be compromised in KCR1 E33D in a yeast expression system. Our findings suggest that KCR1 genetic variations that diminish the ability of KCR1 to protect KCNH2 from inhibition by commonly used therapeutic agents constitute a risk factor for the aLQTS.


Asunto(s)
Glucosiltransferasas/genética , Síndrome de QT Prolongado/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Células CHO , Cricetinae , Cricetulus , Análisis Mutacional de ADN , Electrofisiología , Femenino , Prueba de Complementación Genética , Glucosiltransferasas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
Heart Rhythm ; 7(7): 973-80, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20348026

RESUMEN

BACKGROUND: Mutations in the KCNQ1 and human ether-a-go-go-related gene (HERG) genes cause the long QT syndromes, LQTS1 and LQTS2, due to reductions in the cardiac repolarizing I(Ks) and I(Kr) currents, respectively. It was previously reported that KCNQ1 coexpression modulates HERG function by enhancing membrane expression of HERG, and that the 2 proteins coimmunoprecipitate, and colocalize in myocytes. In vivo studies in genetically modified rabbits also support a HERG-KCNQ1 interaction. OBJECTIVE: We sought to determine whether KCNQ1 influences the current characteristics of HERG genetic variants. METHODS: This study used expression of HERG and KCNQ1 wild-type (WT) and mutant channels in heterologous systems, combined with whole-cell patch clamp analysis and biochemistry. RESULTS: Supporting the notion that KCNQ1 needs to be trafficking competent to influence HERG function, we found that although the tail current density of HERG expressed in Chinese Hamster Ovary (CHO) cells was approximately doubled by WT KCNQ1 coexpression, it was not altered in the presence of the trafficking-defective KCNQ1(T587M) variant. Activation and deactivation kinetics of HERG variants were not altered. The HERG(M124T) variant, previously shown to be mildly impaired functionally, was restored to WT levels by KCNQ1-WT but not KCNQ1(T587M) coexpression. The tail current densities of the severely trafficking-impaired HERG(G601S) and HERG(F805C) variants were only slightly improved by KCNQ1 coexpression. The trafficking competent but incompletely processed HERG(N598Q), and a mutation in the selectivity filter, HERG(G628S), were not improved by KCNQ1 coexpression. CONCLUSION: These findings suggest a functional codependence of HERG on KCNQ1 during channel biogenesis. Moreover, KCNQ1 variably modulates LQTS2 mutations with distinct underlying pathologies.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Canal de Potasio KCNQ1/metabolismo , Síndrome de QT Prolongado/genética , Animales , Western Blotting , Células CHO , Membrana Celular/genética , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Técnicas Electrofisiológicas Cardíacas , Inmunoprecipitación , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/metabolismo , Técnicas de Placa-Clamp , Transporte de Proteínas
13.
J Physiol ; 587(Pt 11): 2555-66, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19406877

RESUMEN

Human ether-a-go-go-related gene (HERG) encodes the rapid, outwardly rectifying K(+) current I(Kr) that is critical for repolarization of the cardiac action potential. Congenital HERG mutations or unintended pharmaceutical block of I(Kr) can lead to life-threatening arrhythmias. Here, we assess the functional role of the alanine at position 653 (HERG-A653) that is highly conserved among evolutionarily divergent K(+) channels. HERG-A653 is close to the 'glycine hinge' implicated in K(+) channel opening, and is flanked by tyrosine 652 and phenylalanine 656, which contribute to the drug binding site. We substituted an array of seven (I, C, S, G, Y, V and T) amino acids at position 653 and expressed individual variants in heterologous systems to assess changes in gating and drug binding. Substitution of A653 resulted in negative shifts of the V(1/2) of activation ranging from -23.6 (A653S) to -62.5 (A653V) compared to -11.2 mV for wild-type (WT). Deactivation was also drastically altered: channels with A653I/C substitutions exhibited delayed deactivation in response to test potentials above the activation threshold, while A653S/G/Y/V/T failed to deactivate under those conditions and required hyperpolarization and prolonged holding potentials at -130 mV. While A653S/G/T/Y variants showed decreased sensitivity to the I(Kr) inhibitor dofetilide, these changes could not be correlated with defects in channel closure. Homology modelling suggests that in the closed state, A653 forms tight contacts with several residues from the neighbouring subunit in the tetramer, playing a key role in S6 helix packing at the narrowest part of the vestibule. Our study suggests that A653 plays an important functional role in the outwardly rectifying gating behaviour of HERG, supporting channel closure at membrane potentials negative to the channel activation threshold.


Asunto(s)
Secuencia Conservada , Canales de Potasio Éter-A-Go-Go/metabolismo , Evolución Molecular , Activación del Canal Iónico , Alanina , Secuencia de Aminoácidos , Animales , Células CHO , Simulación por Computador , Cricetinae , Cricetulus , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Cinética , Potenciales de la Membrana , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Oocitos , Fenetilaminas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Conformación Proteica , Relación Estructura-Actividad , Sulfonamidas/farmacología , Transfección , Xenopus laevis
14.
J Biol Chem ; 284(13): 8846-54, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19171938

RESUMEN

Sodium channels are fundamental signaling molecules in excitable cells, and are molecular targets for local anesthetic agents and intracellular free Ca(2+) ([Ca(2+)](i)). Two regions of Na(V)1.5 have been identified previously as [Ca(2+)](i)-sensitive modulators of channel inactivation. These include a C-terminal IQ motif that binds calmodulin (CaM) in different modes depending on Ca(2+) levels, and an immediately adjacent C-terminal EF-hand domain that directly binds Ca(2+). Here we show that a mutation of the IQ domain (A1924T; Brugada Syndrome) that reduces CaM binding stabilizes Na(V)1.5 inactivation, similarly and more extensively than even reducing [Ca(2+)](i). Because the DIII-DIV linker is an essential structure in Na(V)1.5 inactivation, we evaluated this domain for a potential CaM binding interaction. We identified a novel CaM binding site within the linker, validated its interaction with CaM by NMR spectroscopy, and revealed its micromolar affinity by isothermal titration calorimetry. Mutation of three consecutive hydrophobic residues (Phe(1520)-Ile(1521)-Phe(1522)) to alanines in this CaM-binding domain recapitulated the electrophysiology phenotype observed with mutation of the C-terminal IQ domain: Na(V)1.5 inactivation was stabilized; moreover, mutations of either CaM-binding domain abolish the well described stabilization of inactivation by lidocaine. The direct physical interaction of CaM with the C-terminal IQ domain and the DIII-DIV linker, combined with the similarity in phenotypes when CaM-binding sites in either domain are mutated, suggests these cytoplasmic structures could be functionally coupled through the action of CaM. These findings have bearing upon Na(+) channel function in genetically altered channels and under pathophysiologic conditions where [Ca(2+)](i) impacts cardiac conduction.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Proteínas Musculares/metabolismo , Canales de Sodio/metabolismo , Secuencias de Aminoácidos/genética , Sustitución de Aminoácidos , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Calcio/química , Calmodulina/química , Calmodulina/genética , Línea Celular , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Proteínas Musculares/química , Proteínas Musculares/genética , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.5 , Resonancia Magnética Nuclear Biomolecular , Estabilidad Proteica , Estructura Cuaternaria de Proteína/genética , Estructura Terciaria de Proteína/genética , Canales de Sodio/química , Canales de Sodio/genética
15.
J Mol Cell Cardiol ; 46(2): 257-67, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19038263

RESUMEN

The human ether-a-go-go related gene (HERG) constitutes the pore forming subunit of I(Kr), a K(+) current involved in repolarization of the cardiac action potential. While mutations in HERG predispose patients to cardiac arrhythmias (Long QT syndrome; LQTS), altered function of HERG regulators are undoubtedly LQTS risk factors. We have combined RNA interference with behavioral screening in Caenorhabditis elegans to detect genes that influence function of the HERG homolog, UNC-103. One such gene encodes the worm ortholog of the rho-GTPase activating protein 6 (ARHGAP6). In addition to its GAP function, ARHGAP6 induces cytoskeletal rearrangements and activates phospholipase C (PLC). Here we show that I(Kr) recorded in cells co-expressing HERG and ARHGAP6 was decreased by 43% compared to HERG alone. Biochemical measurements of cell-surface associated HERG revealed that ARHGAP6 reduced membrane expression of HERG by 35%, which correlates well with the reduction in current. In an atrial myocyte cell line, suppression of endogenous ARHGAP6 by virally transduced shRNA led to a 53% enhancement of I(Kr). ARHGAP6 effects were maintained when we introduced a dominant negative rho-GTPase, or ARHGAP6 devoid of rhoGAP function, indicating ARHGAP6 regulation of HERG is independent of rho activation. However, ARHGAP6 lost effectiveness when PLC was inhibited. We further determined that ARHGAP6 effects are mediated by a consensus SH3 binding domain within the C-terminus of HERG, although stable ARHGAP6-HERG complexes were not observed. These data link a rhoGAP-activated PLC pathway to HERG membrane expression and implicate this family of proteins as candidate genes in disorders involving HERG.


Asunto(s)
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Canal de Potasio ERG1 , Electrofisiología , Canales de Potasio Éter-A-Go-Go/genética , Proteínas Activadoras de GTPasa/genética , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Potasio/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Interferencia de ARN
16.
J Biol Chem ; 282(8): 5506-13, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17189275

RESUMEN

The HERG (human ether-à-go-go-related gene) protein, which underlies the cardiac repolarizing current I(Kr), is the unintended target for many pharmaceutical agents. Inadvertent block of I(Kr), known as the acquired long QT syndrome (aLQTS), is a leading cause for drug withdrawal by the United States Food and Drug Administration. Hence, an improved understanding of the regulatory factors that protect most individuals from aLQTS is essential for advancing clinical therapeutics in broad areas, from cancer chemotherapy to antipsychotics and antidepressants. Here, we show that the K(+) channel regulatory protein KCR1, which markedly reduces I(Kr) drug sensitivity, protects HERG through glucosyltransferase function. KCR1 and the yeast alpha-1,2-glucosyltransferase ALG10 exhibit sequence homology, and like KCR1, ALG10 diminished HERG block by dofetilide. Inhibition of cellular glycosylation pathways with tunicamycin abrogated the effects of KCR1, as did expression in Lec1 cells (deficient in glycosylation). Moreover, KCR1 complemented the growth defect of an alg10-deficient yeast strain and enhanced glycosylation of an Alg10 substrate in yeast. HERG itself is not the target for KCR1-mediated glycosylation because the dofetilide response of glycosylation-deficient HERG(N598Q) was still modulated by KCR1. Nonetheless, our data indicate that the alpha-1,2-glucosyltransferase function is a key component of the molecular pathway whereby KCR1 diminishes I(Kr) drug response. Incorporation of in vitro data into a computational model indicated that KCR1 expression is protective against arrhythmias. These findings reveal a potential new avenue for targeted prevention of aLQTS.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Glucosiltransferasas/metabolismo , Síndrome de QT Prolongado/metabolismo , Animales , Antidepresivos/efectos adversos , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Antipsicóticos/efectos adversos , Células CHO , Cricetinae , Cricetulus , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Expresión Génica , Glucosiltransferasas/deficiencia , Glucosiltransferasas/genética , Glicosilación , Humanos , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/prevención & control , Modelos Cardiovasculares , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Modificación Traduccional de las Proteínas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
17.
Circ Res ; 97(12): 1262-9, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16284182

RESUMEN

Sudden cardiac death attributable to ventricular tachycardia/fibrillation (VF) remains a catastrophic outcome of myocardial ischemia and infarction. At the same time, conventional antagonist drugs targeting ion channels have yielded poor survival benefits. Although pharmacological and genetic models suggest an association between sodium (Na+) channel loss-of-function and sudden cardiac death, molecular mechanisms have not been identified that convincingly link ischemia to Na+ channel dysfunction and ventricular arrhythmias. Because ischemia can evoke the generation of reactive oxygen species, we explored the effect of oxidative stress on Na+ channel function. We show here that oxidative stress reduces Na+ channel availability. Both the general oxidant tert-butyl-hydroperoxide and a specific, highly reactive product of the isoprostane pathway of lipid peroxidation, E2-isoketal, potentiate inactivation of cardiac Na+ channels in human embryonic kidney (HEK)-293 cells and cultured atrial (HL-1) myocytes. Furthermore, E2-isoketals were generated in the epicardial border zone of the canine healing infarct, an arrhythmogenic focus where Na+ channels exhibit similar inactivation defects. In addition, we show synergistic functional effects of flecainide, a proarrhythmic Na+ channel blocker, and oxidative stress. These data suggest Na+ channel dysfunction evoked by lipid peroxidation is a candidate mechanism for ischemia-related conduction abnormalities and arrhythmias.


Asunto(s)
Arritmias Cardíacas/etiología , Peroxidación de Lípido , Miocardio/metabolismo , Canales de Sodio/fisiología , Aldehídos/farmacología , Línea Celular , Flecainida/farmacología , Humanos , Isoprostanos/metabolismo , Infarto del Miocardio/metabolismo , Estrés Oxidativo , terc-Butilhidroperóxido/farmacología
18.
J Clin Invest ; 115(11): 3045-56, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16276415

RESUMEN

Ang II type 1 (AT1) receptors activate both conventional heterotrimeric G protein-dependent and unconventional G protein-independent mechanisms. We investigated how these different mechanisms activated by AT1 receptors affect growth and death of cardiac myocytes in vivo. Transgenic mice with cardiac-specific overexpression of WT AT1 receptor (AT1-WT; Tg-WT mice) or an AT1 receptor second intracellular loop mutant (AT1-i2m; Tg-i2m mice) selectively activating G(alpha)q/G(alpha)i-independent mechanisms were studied. Tg-i2m mice developed more severe cardiac hypertrophy and bradycardia coupled with lower cardiac function than Tg-WT mice. In contrast, Tg-WT mice exhibited more severe fibrosis and apoptosis than Tg-i2m mice. Chronic Ang II infusion induced greater cardiac hypertrophy in Tg-i2m compared with Tg-WT mice whereas acute Ang II administration caused an increase in heart rate in Tg-WT but not in Tg-i2m mice. Membrane translocation of PKCepsilon, cytoplasmic translocation of G(alpha)q, and nuclear localization of phospho-ERKs were observed only in Tg-WT mice while activation of Src and cytoplasmic accumulation of phospho-ERKs were greater in Tg-i2m mice, consistent with the notion that G(alpha)q/G(alpha)i-independent mechanisms are activated in Tg-i2m mice. Cultured myocytes expressing AT1-i2m exhibited a left and upward shift of the Ang II dose-response curve of hypertrophy compared with those expressing AT1-WT. Thus, the AT1 receptor mediates downstream signaling mechanisms through G(alpha)q/G(alpha)i-dependent and -independent mechanisms, which induce hypertrophy with a distinct phenotype.


Asunto(s)
Bradicardia/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hipertrofia Ventricular Izquierda/genética , Mutación , Miocitos Cardíacos/metabolismo , Receptor de Angiotensina Tipo 1/deficiencia , Receptor de Angiotensina Tipo 1/genética , Animales , Apoptosis/genética , Bradicardia/metabolismo , Bradicardia/patología , Células Cultivadas , Electrocardiografía , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Fenotipo , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/metabolismo
19.
J Gen Physiol ; 126(4): 353-63, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16186562

RESUMEN

The pharmacological properties of slow Ca(2+)-activated K(+) current (K(slow)) were investigated in mouse pancreatic beta-cells and islets to understand how K(slow) contributes to the control of islet bursting, [Ca(2+)](i) oscillations, and insulin secretion. K(slow) was insensitive to apamin or the K(ATP) channel inhibitor tolbutamide, but UCL 1684, a potent and selective nonpeptide SK channel blocker reduced the amplitude of K(slow) tail current in voltage-clamped mouse beta-cells. K(slow) was also selectively and reversibly inhibited by the class III antiarrythmic agent azimilide (AZ). In isolated beta-cells or islets, pharmacologic inhibition of K(slow) by UCL 1684 or AZ depolarized beta-cell silent phase potential, increased action potential firing, raised [Ca(2+)](i), and enhanced glucose-dependent insulin secretion. AZ inhibition of K(slow) also supported mediation by SK, rather than cardiac-like slow delayed rectifier channels since bath application of AZ to HEK 293 cells expressing SK3 cDNA reduced SK current. Further, AZ-sensitive K(slow) current was extant in beta-cells from KCNQ1 or KCNE1 null mice lacking cardiac slow delayed rectifier currents. These results strongly support a functional role for SK channel-mediated K(slow) current in beta-cells, and suggest that drugs that target SK channels may represent a new approach for increasing glucose-dependent insulin secretion. The apamin insensitivity of beta-cell SK current suggests that beta-cells express a unique SK splice variant or a novel heteromultimer consisting of different SK subunits.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Potasio/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Alcanos/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Glucosa/farmacología , Humanos , Hidantoínas , Imidazolidinas/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Potenciales de la Membrana , Ratones , Ratones Noqueados , Piperazinas/farmacología , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Compuestos de Quinolinio/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Factores de Tiempo , Transfección
20.
Heart Rhythm ; 2(6): 634-40, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15922273

RESUMEN

BACKGROUND: Multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) is abundant in myocardium. CaMKII activity is augmented by catecholamine stimulation, which enhances AV nodal conduction, suggesting the hypothesis that CaMKII also contributes to AV nodal conduction properties. OBJECTIVES: The purpose of this study was to test the potential role of CaMKII in regulating AV nodal conduction in heart. METHODS: We developed a novel mouse with genetic CaMKII inhibition by cardiac-specific expression of autocamtide 3 inhibitory peptide (AC3-I) mimicking a conserved sequence of the CaMKII regulatory domain. We also engineered a control transgenic mouse with cardiac expression of an inactive, scrambled version of AC3-I (autocamtide 3 control peptide [AC3-C]) and performed electrophysiologic measurements in vivo and in Langendorff-perfused isolated hearts. RESULTS: AC3-I and AC3-C were abundantly expressed in AV nodal cells. AC3-I mice with implanted ECG telemeters showed enhanced Wenckebach-type AV conduction block after isoproterenol (present in 9/9 mice) compared with AC3-C mice (present in 1/5 mice, P = .005). Intracardiac recordings showed significant PR and AH interval prolongation in AC3-I mice at baseline and after isoproterenol compared with AC3-C mice. HV durations were not different. Langendorff-perfused AC3-I hearts had significantly prolonged Wenckebach cycle lengths and AV nodal effective refractory periods compared with AC3-C hearts, whereas sinus node recovery time and left ventricular effective refractory times were similar between these genotypes. CONCLUSIONS: These studies define CaMKII as a critical determinant of normal and catecholamine-stimulated AV nodal conduction responses.


Asunto(s)
Nodo Atrioventricular/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Análisis de Varianza , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Regulación Enzimológica de la Expresión Génica , Ratones , Ratones Transgénicos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...