Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
S D Med ; 73(4): 171-177, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32445305

RESUMO

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.


Assuntos
Reanimação Cardiopulmonar , Serviços Médicos de Emergência , Parada Cardíaca Extra-Hospitalar , Humanos , Parada Cardíaca Extra-Hospitalar/terapia , Estudos Retrospectivos , South Dakota
2.
J Mol Cell Cardiol ; 132: 24-35, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31077706

RESUMO

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.


Assuntos
Potenciais de Ação , Fibrilação Atrial/etiologia , Fator Natriurético Atrial/genética , Mutação da Fase de Leitura , Átrios do Coração/fisiopatologia , Miócitos Cardíacos/patologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Animais , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Fator Natriurético Atrial/metabolismo , Fenômenos Eletrofisiológicos , Humanos , Potenciais da Membrana , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo
3.
Holist Nurs Pract ; 33(4): 197-203, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31192831

RESUMO

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.


Assuntos
Exercícios Respiratórios/normas , Respiração , Testes de Função Respiratória/estatística & dados numéricos , Yoga , Adulto , Exercícios Respiratórios/métodos , Feminino , Voluntários Saudáveis/estatística & dados numéricos , Humanos , Pulmão/fisiologia , Masculino , Projetos Piloto , Testes de Função Respiratória/métodos , South Dakota
4.
S D Med ; 71(2): 72-79, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29990416

RESUMO

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.


Assuntos
Ciência de Dados , Serviços Médicos de Emergência , Parada Cardíaca Extra-Hospitalar/epidemiologia , População Rural/estatística & dados numéricos , Reanimação Cardiopulmonar , Humanos , South Dakota/epidemiologia
5.
Stem Cells ; 32(7): 1774-88, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24648383

RESUMO

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.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Sistema de Sinalização das MAP Quinases , Proteínas/fisiologia , Animais , Células Cultivadas , Citocinas , Átrios do Coração/citologia , Camundongos , Miócitos Cardíacos/fisiologia
6.
J Allied Health ; 53(1): 3-9, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38430490

RESUMO

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.


Assuntos
Atenção à Saúde , Participação dos Interessados , Adulto , Humanos , Aprendizagem Baseada em Problemas
7.
J Biol Chem ; 287(47): 39613-25, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23033485

RESUMO

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.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/tratamento farmacológico , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/metabolismo , Naftiridinas/química , Naftiridinas/farmacologia , Fenetilaminas/efeitos adversos , Bloqueadores dos Canais de Potássio/efeitos adversos , Piridinas/química , Piridinas/farmacologia , Sulfonamidas/efeitos adversos , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Relação Dose-Resposta a Droga , Descoberta de Drogas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Proteínas Musculares/genética , Miocárdio/metabolismo , Miocárdio/patologia , Fenetilaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Coelhos , Relação Estrutura-Atividade , Sulfonamidas/farmacologia
8.
Circulation ; 124(9): 1001-11, 2011 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-21824921

RESUMO

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.


Assuntos
Flutter Atrial/genética , Cardiomiopatia Dilatada/genética , Mutação de Sentido Incorreto , Síndrome do Nó Sinusal/genética , Canais de Sódio/genética , Animais , Flutter Atrial/terapia , Células CHO , Cricetinae , Cricetulus , Desfibriladores Implantáveis , Eletrocardiografia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Contração Miocárdica/genética , Canal de Sódio Disparado por Voltagem NAV1.5 , Síndrome do Nó Sinusal/terapia , Resultado do Tratamento , Adulto Jovem
9.
J Mol Cell Cardiol ; 50(1): 50-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20950623

RESUMO

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.


Assuntos
Glucosiltransferases/genética , Síndrome do QT Longo/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Células CHO , Cricetinae , Cricetulus , Análise Mutacional de DNA , Eletrofisiologia , Feminino , Teste de Complementação Genética , Glucosiltransferases/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
10.
J Hosp Palliat Nurs ; 22(4): 305-311, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32568939

RESUMO

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.


Assuntos
Planejamento Antecipado de Cuidados/normas , Redes Comunitárias/tendências , Docentes de Enfermagem/tendências , Planejamento Antecipado de Cuidados/tendências , Comportamento Cooperativo , Humanos , Estudos Longitudinais , South Dakota
11.
J Mol Cell Cardiol ; 46(2): 257-67, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19038263

RESUMO

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.


Assuntos
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Canal de Potássio ERG1 , Eletrofisiologia , Canais de Potássio Éter-A-Go-Go/genética , Proteínas Ativadoras de GTPase/genética , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Potássio/metabolismo , Canais de Potássio/genética , Canais de Potássio/metabolismo , Interferência de RNA
12.
J Physiol ; 587(Pt 11): 2555-66, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19406877

RESUMO

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.


Assuntos
Sequência Conservada , Canais de Potássio Éter-A-Go-Go/metabolismo , Evolução Molecular , Ativação do Canal Iônico , Alanina , Sequência de Aminoácidos , Animais , Células CHO , Simulação por Computador , Cricetinae , Cricetulus , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Cinética , Potenciais da Membrana , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Oócitos , Fenetilaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Conformação Proteica , Relação Estrutura-Atividade , Sulfonamidas/farmacologia , Transfecção , Xenopus laevis
13.
J Clin Invest ; 115(11): 3045-56, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16276415

RESUMO

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.


Assuntos
Bradicardia/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hipertrofia Ventricular Esquerda/genética , Mutação , Miócitos Cardíacos/metabolismo , Receptor Tipo 1 de Angiotensina/deficiência , Receptor Tipo 1 de Angiotensina/genética , Animais , Apoptose/genética , Bradicardia/metabolismo , Bradicardia/patologia , Células Cultivadas , Eletrocardiografia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose/genética , Fibrose/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Fenótipo , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina/metabolismo
14.
J Clin Invest ; 113(8): 1130-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15085192

RESUMO

Heterozygous mutations of the cardiac transcription factor Nkx2-5 cause atrioventricular conduction defects in humans by unknown mechanisms. We show in KO mice that the number of cells in the cardiac conduction system is directly related to Nkx2-5 gene dosage. Null mutant embryos appear to lack the primordium of the atrioventricular node. In Nkx2-5 haploinsufficiency, the conduction system has half the normal number of cells. In addition, an entire population of connexin40(-)/connexin45(+) cells is missing in the atrioventricular node of Nkx2-5 heterozygous KO mice. Specific functional defects associated with Nkx2-5 loss of function can be attributed to hypoplastic development of the relevant structures in the conduction system. Surprisingly, the cellular expression of connexin40, the major gap junction isoform of Purkinje fibers and a putative Nkx2-5 target, is unaffected, consistent with normal conduction times through the His-Purkinje system measured in vivo. Postnatal conduction defects in Nkx2-5 mutation may result at least in part from a defect in the genetic program that governs the recruitment or retention of embryonic cardiac myocytes in the conduction system.


Assuntos
Sistema de Condução Cardíaco/patologia , Proteínas de Homeodomínio/genética , Mutação , Animais , Conexinas/análise , Eletrocardiografia , Proteínas de Homeodomínio/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína alfa-5 de Junções Comunicantes
15.
Circ Res ; 97(12): 1262-9, 2005 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-16284182

RESUMO

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.


Assuntos
Arritmias Cardíacas/etiologia , Peroxidação de Lipídeos , Miocárdio/metabolismo , Canais de Sódio/fisiologia , Aldeídos/farmacologia , Linhagem Celular , Flecainida/farmacologia , Humanos , Isoprostanos/metabolismo , Infarto do Miocárdio/metabolismo , Estresse Oxidativo , terc-Butil Hidroperóxido/farmacologia
16.
Circ Res ; 97(1): 62-9, 2005 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-15947250

RESUMO

Although atrial fibrillation is the most common serious cardiac arrhythmia, the fundamental molecular pathways remain undefined. Mutations in KCNQ1, one component of a sympathetically activated cardiac potassium channel complex, cause familial atrial fibrillation, although the mechanisms in vivo are unknown. We show here that mice with deletion of the KCNQ1 protein partner KCNE1 have spontaneous episodes of atrial fibrillation despite normal atrial size and structure. Isoproterenol abolishes these abnormalities, but vagomimetic interventions have no effect. Whereas loss of KCNE1 function prolongs ventricular action potentials in humans, KCNE1-/- mice displayed unexpectedly shortened atrial action potentials, and multiple potential mechanisms were identified: (1) K+ currents (total and those sensitive to the KCNQ1 blocker chromanol 293B) were significantly increased in atrial cells from KCNE1-/- mice compared with controls, and (2) when CHO cells expressing KCNQ1 and KCNE1 were pulsed very rapidly (at rates comparable to the normal mouse heart and to human atrial fibrillation), the sigmoidicity of IKs activation prevented current accumulation, whereas cells expressing KCNQ1 alone displayed marked current accumulation at these very rapid rates. Thus, KCNE1 deletion in mice unexpectedly leads to increased outward current in atrial myocytes, shortens atrial action potentials, and enhances susceptibility to atrial fibrillation.


Assuntos
Fibrilação Atrial/etiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Potenciais de Ação , Animais , Fibrilação Atrial/fisiopatologia , Suscetibilidade a Doenças , Isoproterenol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética
17.
J Gen Physiol ; 126(4): 353-63, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186562

RESUMO

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.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Potássio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Alcanos/farmacologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Glucose/farmacologia , Humanos , Hidantoínas , Imidazolidinas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Potenciais da Membrana , Camundongos , Camundongos Knockout , Piperazinas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Compostos de Quinolínio/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Fatores de Tempo , Transfecção
18.
Circulation ; 107(22): 2850-6, 2003 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-12782567

RESUMO

BACKGROUND: Mutations in the gamma2 subunit (PRKAG2) of AMP-activated protein kinase produce an unusual human cardiomyopathy characterized by ventricular hypertrophy and electrophysiological abnormalities: Wolff-Parkinson-White syndrome (WPW) and progressive degenerative conduction system disease. Pathological examinations of affected human hearts reveal vacuoles containing amylopectin, a glycogen-related substance. METHODS AND RESULTS: To elucidate the mechanism by which PRKAG2 mutations produce hypertrophy with electrophysiological abnormalities, we constructed transgenic mice overexpressing the PRKAG2 cDNA with or without a missense N488I human mutation. Transgenic mutant mice showed elevated AMP-activated protein kinase activity, accumulated large amounts of cardiac glycogen (30-fold above normal), developed dramatic left ventricular hypertrophy, and exhibited ventricular preexcitation and sinus node dysfunction. Electrophysiological testing demonstrated alternative atrioventricular conduction pathways consistent with WPW. Cardiac histopathology revealed that the annulus fibrosis, which normally insulates the ventricles from inappropriate excitation by the atria, was disrupted by glycogen-filled myocytes. These anomalous microscopic atrioventricular connections, rather than morphologically distinct bypass tracts, appeared to provide the anatomic substrate for ventricular preexcitation. CONCLUSIONS: Our data establish PRKAG2 mutations as a glycogen storage cardiomyopathy, provide an anatomic explanation for electrophysiological findings, and implicate disruption of the annulus fibrosis by glycogen-engorged myocytes as the cause of preexcitation in Pompe, Danon, and other glycogen storage diseases.


Assuntos
Cardiomiopatias/fisiopatologia , Doença de Depósito de Glicogênio/fisiopatologia , Complexos Multienzimáticos/biossíntese , Proteínas Serina-Treonina Quinases/biossíntese , Síndrome de Wolff-Parkinson-White/etiologia , Síndrome de Wolff-Parkinson-White/fisiopatologia , Proteínas Quinases Ativadas por AMP , Animais , Cardiomiopatias/complicações , Cardiomiopatias/patologia , Modelos Animais de Doenças , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Doença de Depósito de Glicogênio/complicações , Doença de Depósito de Glicogênio/patologia , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Camundongos , Camundongos Transgênicos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutação , Miocárdio/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Taxa de Sobrevida , Síndrome de Wolff-Parkinson-White/patologia
19.
Heart Rhythm ; 2(6): 634-40, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15922273

RESUMO

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.


Assuntos
Nó Atrioventricular/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Análise de Variância , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Regulação Enzimológica da Expressão Gênica , Camundongos , Camundongos Transgênicos , Transdução de Sinais
20.
FASEB J ; 17(15): 2263-5, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14525949

RESUMO

The cardiac potassium channel encoded by the human ether-à-go-go related gene (HERG) is blocked by a diverse array of common therapeutic compounds. Even transient exposure to such agents may provoke the life-threatening cardiac arrhythmia torsades de pointes in some, but not all, individuals. Although the molecular and genetic factors predicting such wide variability in drug response remain unclear, known sequence variations within the coding region of HERG do not explain the adverse drug response in many cases. Although other proteins can modulate HERG function, no studies have identified protein partners capable of limiting the pharmacological sensitivity of HERG. Here we show that KCR1, a protein identified previously in rat cerebellum, is a plasma membrane-associated protein expressed at the RNA level in the human heart and can be immunoprecipitated with HERG. Functionally, KCR1 reduces the sensitivity of HERG to classic proarrhythmic HERG blockers (sotalol, quinidine, dofetilide) in both cardiac and noncardiac cell lines. We propose that KCR1, when coupled to HERG, may limit the sensitivity of HERG to proarrhythmic drug blockade and may be a rational target for modifying the proarrhythmic effects of otherwise clinically useful compounds.


Assuntos
Antiarrítmicos/farmacologia , Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Miócitos Cardíacos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , Transativadores , Animais , Antiarrítmicos/antagonistas & inibidores , Células CHO , Linhagem Celular , Cricetinae , Canal de Potássio ERG1 , Condutividade Elétrica , Canais de Potássio Éter-A-Go-Go , Glucosiltransferases/metabolismo , Humanos , Ativação do Canal Iônico , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Bloqueadores dos Canais de Potássio/antagonistas & inibidores , Canais de Potássio/genética , Regulador Transcricional ERG , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA