Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Trans Am Clin Climatol Assoc ; 128: 353-362, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28790518

RESUMEN

Academic health centers (AHCs) are the nation's primary resource for healthcare discovery, innovation, and training. US healthcare revenue growth has declined sharply since 2009, and is forecast to remain well below historic levels for the foreseeable future. As the cost of education and research at nearly all AHCs is heavily subsidized through large transfers from clinical care margins, our institutions face a mounting crisis. Choices centering on how to increase the cost-effectiveness of the AHC enterprise require unprecedented levels of alignment to preserve an environment that nurtures creativity. Management processes require governance models that clarify decision rights while harnessing the talents and the intellectual capital of a large, diverse enterprise to nimbly address unfamiliar organizational challenges. This paper describes key leadership tactics aimed at propelling AHCs along this journey - one that requires from all leaders a commitment to resilience, optimism, and willingness to embrace change.


Asunto(s)
Centros Médicos Académicos/organización & administración , Atención a la Salud , Administración Hospitalaria , Humanos , Liderazgo , Modelos Organizacionales , Afiliación Organizacional , Estados Unidos
3.
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
4.
Am J Hum Genet ; 86(4): 560-72, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20362271

RESUMEN

Large-scale DNA databanks linked to electronic medical record (EMR) systems have been proposed as an approach for rapidly generating large, diverse cohorts for discovery and replication of genotype-phenotype associations. However, the extent to which such resources are capable of delivering on this promise is unknown. We studied whether an EMR-linked DNA biorepository can be used to detect known genotype-phenotype associations for five diseases. Twenty-one SNPs previously implicated as common variants predisposing to atrial fibrillation, Crohn disease, multiple sclerosis, rheumatoid arthritis, or type 2 diabetes were successfully genotyped in 9483 samples accrued over 4 mo into BioVU, the Vanderbilt University Medical Center DNA biobank. Previously reported odds ratios (OR(PR)) ranged from 1.14 to 2.36. For each phenotype, natural language processing techniques and billing-code queries were used to identify cases (n = 70-698) and controls (n = 808-3818) from deidentified health records. Each of the 21 tests of association yielded point estimates in the expected direction. Previous genotype-phenotype associations were replicated (p < 0.05) in 8/14 cases when the OR(PR) was > 1.25, and in 0/7 with lower OR(PR). Statistically significant associations were detected in all analyses that were adequately powered. In each of the five diseases studied, at least one previously reported association was replicated. These data demonstrate that phenotypes representing clinical diagnoses can be extracted from EMR systems, and they support the use of DNA resources coupled to EMR systems as tools for rapid generation of large data sets required for replication of associations found in research cohorts and for discovery in genome science.


Asunto(s)
Artritis Reumatoide/genética , Fibrilación Atrial/genética , Enfermedad de Crohn/genética , Diabetes Mellitus Tipo 2/genética , Registros Electrónicos de Salud , Estudios de Asociación Genética/tendencias , Esclerosis Múltiple/genética , Estudios de Casos y Controles , ADN/sangre , ADN/genética , Genoma Humano , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple/genética
5.
Acad Med ; 85(2): 266-72, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20107354

RESUMEN

One hundred years after Flexner wrote his report for the Carnegie Foundation, calls are heard for another "Flexnerian revolution," a reform movement that would overhaul an approach to medical education that is criticized for its expense and inefficiency, its failure to respond to the health needs of our communities, and the high cost and inefficiency of the health care system it supports. To address these concerns, a group of Vanderbilt educators, national experts, administrators, residents, and students attended a retreat in November 2008. The goal of this meeting was to craft a new vision of physician learning based on the continuous development and assessment of competencies needed for effective and compassionate care under challenging circumstances. The vision that emerged from this gathering was that of a health care workforce comprised of physicians and other professionals, all capable of assessing practice outcomes, identifying learning needs, and engaging in continuous learning to achieve the best care for their patients. Several principles form the foundation for this vision. Learning should be competency based and embedded in the workplace. It should be linked to patient needs and undertaken by individual providers, by teams, and by institutions. Health professionals should be trained in this new model from the start of the educational experience, leading to true interprofessional education, with shared facilities and the same basic coursework. Multiple entry and exit points would provide flexibility and would allow health professionals to redirect their careers as their goals evolved. This article provides a detailed account of the model developed at the retreat and the obstacles that might be encountered in attempting to implement it.


Asunto(s)
Educación Médica/métodos , Personal de Salud/educación , Modelos Educacionales , Grupo de Atención al Paciente , Centros Médicos Académicos/organización & administración , Centros Médicos Académicos/tendencias , Educación Basada en Competencias , Educación en Enfermería/métodos , Educación en Farmacia/métodos , Humanos
6.
Mol Pharmacol ; 76(4): 861-71, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19620257

RESUMEN

The pore of the Na+ channel is lined by asymmetric loops formed by the linkers between the fifth and sixth transmembrane segments (S5-S6). We investigated the role of the N-terminal portion (SS1) of the S5-S6 linkers in channel gating and local anesthetic (LA) block using site-directed cysteine mutagenesis of the rat skeletal muscle (Na(V)1.4) channel. The mutants examined have variable effects on voltage dependence and kinetics of fast inactivation. Of the cysteine mutants immediately N-terminal to the putative DEKA selectivity filter in four domains, only Q399C in domain I and F1236C in domain III exhibit reduced use-dependent block. These two mutations also markedly accelerated the recovery from use-dependent block. Moreover, F1236C and Q399C significantly decreased the affinity of QX-314 for binding to its channel receptor by 8.5- and 3.3-fold, respectively. Oddly enough, F1236C enhanced stabilization of slow inactivation by both hastening entry into and delaying recovery from slow inactivation states. It is noteworthy that symmetric applications of QX-314 on both external and internal sides of F1236C mutant channels reduced recovery from use-dependent block, indicating an allosteric effect of external QX-314 binding on the recovery of availability of F1236C. These observations suggest that cysteine mutation in the SS1 region, particularly immediate adjacent to the DEKA ring, may lead to a structural rearrangement that alters binding of permanently charged QX-314 to its receptor. The results lend further support for a role for the selectivity filter region as a structural determinant for local anesthetic block.


Asunto(s)
Anestésicos Locales/farmacología , Proteínas Musculares/metabolismo , Músculo Esquelético/efectos de los fármacos , Canales de Sodio/metabolismo , Anestésicos Locales/metabolismo , Animales , Activación del Canal Iónico , Cinética , Proteínas Musculares/química , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Mutagénesis Sitio-Dirigida , Ratas , Canales de Sodio/química , Canales de Sodio/genética
7.
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
8.
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
9.
J Biol Chem ; 284(10): 6436-45, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19074138

RESUMEN

The voltage-gated sodium channel NaV1.5 is responsible for the initial upstroke of the action potential in cardiac tissue. Levels of intracellular calcium modulate inactivation gating of NaV1.5, in part through a C-terminal EF-hand calcium binding domain. The significance of this structure is underscored by the fact that mutations within this domain are associated with specific cardiac arrhythmia syndromes. In an effort to elucidate the molecular basis for calcium regulation of channel function, we have determined the solution structure of the C-terminal EF-hand domain using multidimensional heteronuclear NMR. The structure confirms the existence of the four-helix bundle common to EF-hand domain proteins. However, the location of this domain is shifted with respect to that predicted on the basis of a consensus 12-residue EF-hand calcium binding loop in the sequence. This finding is consistent with the weak calcium affinity reported for the isolated EF-hand domain; high affinity binding is observed only in a construct with an additional 60 residues C-terminal to the EF-hand domain, including the IQ motif that is central to the calcium regulatory apparatus. The binding of an IQ motif peptide to the EF-hand domain was characterized by isothermal titration calorimetry and nuclear magnetic resonance spectroscopy. The peptide binds between helices I and IV in the EF-hand domain, similar to the binding of target peptides to other EF-hand calcium-binding proteins. These results suggest a molecular basis for the coupling of the intrinsic (EF-hand domain) and extrinsic (calmodulin) components of the calcium-sensing apparatus of NaV1.5.


Asunto(s)
Proteínas de Unión al Calcio/química , Calcio/química , Proteínas Musculares/química , Miocardio/química , Canales de Sodio/química , Secuencias de Aminoácidos/fisiología , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Calmodulina/química , Calmodulina/genética , Calmodulina/metabolismo , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Resonancia Magnética Nuclear Biomolecular , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Canales de Sodio/genética , Canales de Sodio/metabolismo , Síndrome
10.
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
11.
Acad Med ; 83(10): 969-75, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18820531

RESUMEN

PURPOSE: In 2000, faced with a national concern over the decreasing number of physician-scientists, Vanderbilt School of Medicine established the institutionally funded Vanderbilt Physician-Scientist Development (VPSD) program to provide centralized oversight and financial support for physician-scientist career development. In 2002, Vanderbilt developed the National Institutes of Health (NIH)-funded Vanderbilt Clinical Research Scholars (VCRS) program using a similar model of centralized oversight. The authors evaluate the impact of the VPSD and VCRS programs on early career outcomes of physician-scientists. METHOD: Physician-scientists who entered the VPSD or VCRS programs from 2000 through 2006 were compared with Vanderbilt physician-scientists who received NIH career development funding during the same period without participating in the VPSD or VCRS programs. RESULTS: Seventy-five percent of VPSD and 60% of VCRS participants achieved individual career award funding at a younger age than the comparison cohort. This shift to career development award funding at a younger age among VPSD and VCRS scholars was accompanied by a 2.6-fold increase in the number of new K awards funded and a rate of growth in K-award dollars at Vanderbilt that outpaced the national rate of growth in K-award funding. CONCLUSIONS: Analysis of the early outcomes of the VPSD and VCRS programs suggests that centralized oversight can catalyze growth in the number of funded physician-scientists at an institution. Investment in this model of career development for physician-scientists may have had an additive effect on the recruitment and retention of talented trainees and junior faculty.


Asunto(s)
Investigación Biomédica/organización & administración , Docentes Médicos/organización & administración , Médicos/economía , Investigadores/economía , Apoyo a la Investigación como Asunto/organización & administración , Centros Médicos Académicos/organización & administración , Selección de Profesión , Educación Médica/economía , Docentes Médicos/provisión & distribución , Organización de la Financiación/economía , Humanos , National Institutes of Health (U.S.) , Médicos/provisión & distribución , Desarrollo de Programa , Evaluación de Programas y Proyectos de Salud , Control de Calidad , Investigadores/provisión & distribución , Tennessee , Estados Unidos
12.
Acad Med ; 83(9): 827-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18728436

RESUMEN

Biomedical science is at an evolutionary turning point. Many of the rate-limiting steps to realizing the next generation of personalized, highly targeted diagnostics and therapeutics rest at the interstices between biomedical science and the classic, university-based disciplines, such as physics, mathematics, computational science, engineering, social sciences, business, and law. Institutes, centers, or other entities created to foster interdisciplinary science are rapidly forming to tackle these formidable challenges, but they are plagued with substantive barriers, born of traditions, processes, and culture, which impede scientific progress and endanger success. Without a more seamless interdisciplinary framework, academic health centers will struggle to move transformative advances in technology into the foundation of biomedical science, and the equally challenging advancement of models that effectively integrate new molecular diagnostics and therapies into the business and social fabric of our population will be similarly hampered. At the same time, excess attention on rankings tied to competition for National Institutes of Health and other federal funds adversely encourages academic medical centers (AMCs) and universities to hoard, rather than share, resources effectively and efficiently. To fully realize their discovery potential, AMCs must consider a substantive realignment relative to one another, as well as with their associated universities, as the academy looks toward innovative approaches to provide a more supportive foundation for the emergent biomedical research enterprise. The authors discuss potential models that could serve to lower barriers to interdisciplinary science, promoting a new synergy between AMCs and their parent universities.


Asunto(s)
Centros Médicos Académicos/organización & administración , Conducta Competitiva , Conducta Cooperativa , Transferencia de Tecnología , Humanos , Cultura Organizacional , Innovación Organizacional , Investigación
13.
J Cardiovasc Pharmacol ; 50(5): 480-6, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18030056

RESUMEN

Unabated reactive oxygen species (ROS) are potentiated by an ischemia-induced shift in anaerobic metabolism, which generates superoxide anion upon reperfusion and reintroduction of oxygen. ROS can modify protein structure and function in fundamental ways, one of which is by forming reactive lipid species from the oxidation of lipids. In this review, we discuss these pathways and discuss the literature that shows that these species can produce dramatic effects on cardiac ion channel function (eg, Na+ channel function). Furthermore, we review what is known about the generation of such in the highly remodeled post myocardial infarction substrate. We suggest prevention of adduction of these highly reactive compounds would be antiarrhythmic.


Asunto(s)
Arritmias Cardíacas/etiología , F2-Isoprostanos/metabolismo , Peroxidación de Lípido/fisiología , Isquemia Miocárdica/complicaciones , Animales , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/prevención & control , F2-Isoprostanos/química , Humanos , Estructura Molecular , Isquemia Miocárdica/fisiopatología , Estrés Oxidativo/fisiología , Piridoxamina/análogos & derivados , Piridoxamina/uso terapéutico , Complejo Vitamínico B/uso terapéutico
14.
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
15.
Proc Natl Acad Sci U S A ; 103(10): 3592-7, 2006 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-16505387

RESUMEN

The function of the human cardiac voltage-gated sodium channel Na(V)1.5 (hH1) is regulated in part by binding of calcium to an EF hand in the C-terminal cytoplasmic domain. hH1 is also regulated via an extrinsic calcium-sensing pathway mediated by calmodulin (CaM) via binding to an IQ motif immediately adjacent to the EF-hand domain. The intrinsic EF-hand domain is shown here to interact with the IQ motif, which controls calcium affinity. Remarkably, mutation of the IQ residues has only a minor effect on CaM affinity but drastically reduces calcium affinity of the EF-hand domain, whereas the Brugada mutation A1924T significantly reduces CaM affinity but has no effect on calcium affinity of the EF-hand domain. Moreover, the differences in the biochemical effects of the mutations directly correlate with contrasting effects on channel electrophysiology. A comprehensive model is proposed in which the hH1 IQ motif serves as a molecular switch, coupling the intrinsic and extrinsic calcium sensors.


Asunto(s)
Proteínas Musculares/química , Proteínas Musculares/metabolismo , Canales de Sodio/química , Canales de Sodio/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión/genética , Calcio/metabolismo , Línea Celular , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Musculares/genética , Mutagénesis Sitio-Dirigida , Mutación , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia , Canales de Sodio/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 Mol Cell Cardiol ; 38(6): 969-81, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15910881

RESUMEN

BACKGROUND: Mutations in the gene encoding the human cardiac sodium channel (SCN5A) have been associated with three distinct cardiac arrhythmia disorders: the long QT syndrome, the Brugada syndrome and cardiac conduction disease. Here we report the biophysical features of a novel sodium channel mutation, E161K, which we identified in individuals of two non-related families with symptoms of bradycardia, sinus node dysfunction, generalized conduction disease and Brugada syndrome, or combinations thereof. METHODS AND RESULTS: Wild-type (WT) or E161K sodium channel alpha-subunit and beta-subunit were cotransfected into tsA201 cells to study the functional consequences of mutant sodium channels. Characterization of whole-cell sodium current (I(Na)) using the whole cell patch-clamp technique revealed that the E161K mutation caused an almost threefold reduction in current density (P < 0.001), and an 11.9 mV positive shift of the voltage-dependence of activation (P < 0.0001). The inactivation properties of mutant and WT sodium channels were similar. These results suggest an overall reduction of E161K I(Na). Incorporation of the experimental findings into computational models demonstrate atrial and ventricular conduction slowing as well as a reduction in sinus rate by slowing of the diastolic depolarization rate and upstroke velocity of the sinus node action potential. This reduction in sinus rate was aggravated by application of acetylcholine, simulating the dominant vagal tone during night. CONCLUSION: Our experimental and computational analysis of the E161K mutation suggests that a loss of sodium channel function is not only associated with Brugada syndrome and conduction disease, but may also cause sinus node dysfunction in carriers of this mutation.


Asunto(s)
Síndrome del Seno Enfermo/genética , Canales de Sodio/genética , Canales de Sodio/fisiología , Acetilcolina/metabolismo , Adulto , Arritmias Cardíacas/genética , Simulación por Computador , Análisis Mutacional de ADN , Electrocardiografía , Electrofisiología , Salud de la Familia , Femenino , Genotipo , Haplotipos , Sistema de Conducción Cardíaco , Humanos , Síndrome de QT Prolongado/genética , Masculino , Persona de Mediana Edad , Mutación , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Linaje , Fenotipo , Síndrome , Factores de Tiempo , Transfección
19.
J Physiol ; 564(Pt 1): 21-31, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15677685

RESUMEN

Upon prolonged depolarizations, voltage-dependent Na+ channels open and subsequently inactivate, occupying fast and slow inactivated conformational states. Like C-type inactivation in K+ channels, slow inactivation is thought to be accompanied by rearrangement of the channel pore. Cysteine-labelling studies have shown that lidocaine, a local anaesthetic (LA) that elicits depolarization-dependent ('use-dependent') Na+ channel block, does not slow recovery from fast inactivation, but modulates the kinetics of slow inactivated states. While these observations suggest LA-induced stabilization of slow inactivation could be partly responsible for use dependence, a more stringent test would require that slow inactivation gating track the distinct use-dependent kinetic properties of diverse LA compounds, such as lidocaine and bupivacaine. For this purpose, we assayed the slow inactivation-dependent accessibility of cysteines engineered into domain III, P-segment (mu1: F1236C, K1237C) to sulfhydryl (MTSEA) modification using a high-speed solution exchange system. As expected, we found that bupivacaine, like lidocaine, protected cysteine residues from MTSEA modification in a depolarization-dependent manner. However, under pulse-train conditions where bupivacaine block of Na+ channels was extensive (due to ultra-slow recovery), but lidocaine block of Na+ channels was not, P-segment cysteines were protected from MTSEA modification. Here we show that conformational changes associated with slow inactivation track the vastly different rates of recovery from use-dependent block for bupivacaine and lidocaine. Our findings suggest that LA compounds may produce their kinetically distinct voltage-dependent behaviour by modulating slow inactivation gating to varying degrees.


Asunto(s)
Anestésicos Locales/farmacología , Canales de Sodio/química , Canales de Sodio/fisiología , Animales , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Conformación Proteica , Ratas
20.
Proc Natl Acad Sci U S A ; 101(32): 11773-8, 2004 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-15280551

RESUMEN

Human ether-a-go-go-related gene (HERG) encodes the pore-forming subunit of I(Kr), a cardiac K(+) channel. Although many commonly used drugs block I(Kr), in certain individuals, this action evokes a paradoxical life-threatening cardiac rhythm disturbance, known as the acquired long QT syndrome (aLQTS). Although aLQTS has become the leading cause of drug withdrawal by the U.S. Food and Drug Administration, DNA sequencing in aLQTS patients has revealed HERG mutations only in rare cases, suggesting that unknown HERG modulators are often responsible. By using the worm Caenorhabditis elegans, we have developed in vivo behavioral assays that identify candidate modulators of unc-103, the worm HERG orthologue. By using RNA-interference methods, we have shown that worm homologues of two HERG-interacting proteins, Hyperkinetic and K channel regulator 1 (KCR1), modify unc-103 function. Examination of the human KCR1 sequence in patients with drug-induced cardiac repolarization defects revealed a sequence variation (the substitution of isoleucine 447 by valine, I447V) that occurs at a reduced frequency (1.1%) relative to a matched control population (7.0%), suggesting that I447V may be an allele for reduced aLQTS susceptibility. This clinical result is supported by in vitro studies of HERG dofetilide sensitivity by using coexpression of HERG with wild-type and I447V KCR1 cDNAs. Our studies demonstrate the feasibility of using C. elegans to assay and potentially identify aLQTS candidate genes.


Asunto(s)
Arritmias Cardíacas/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Canales de Potasio/genética , Canales de Potasio/fisiología , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Electrofisiología , Humanos , Locomoción/genética , Movimiento (Física) , Mutación , Enfermedades Neuromusculares/genética , Canales de Potasio/metabolismo , Unión Proteica , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...