Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 150(5): 1055-67, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22939628

RESUMEN

The type 2 ryanodine receptor/calcium release channel (RyR2), required for excitation-contraction coupling in the heart, is abundant in the brain. Chronic stress induces catecholamine biosynthesis and release, stimulating ß-adrenergic receptors and activating cAMP signaling pathways in neurons. In a murine chronic restraint stress model, neuronal RyR2 were phosphorylated by protein kinase A (PKA), oxidized, and nitrosylated, resulting in depletion of the stabilizing subunit calstabin2 (FKBP12.6) from the channel complex and intracellular calcium leak. Stress-induced cognitive dysfunction, including deficits in learning and memory, and reduced long-term potentiation (LTP) at the hippocampal CA3-CA1 connection were rescued by oral administration of S107, a compound developed in our laboratory that stabilizes RyR2-calstabin2 interaction, or by genetic ablation of the RyR2 PKA phosphorylation site at serine 2808. Thus, neuronal RyR2 remodeling contributes to stress-induced cognitive dysfunction. Leaky RyR2 could be a therapeutic target for treatment of stress-induced cognitive dysfunction.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Calcio/metabolismo , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos de Estrés Traumático/metabolismo
2.
Bioorg Med Chem Lett ; 43: 128058, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33895276

RESUMEN

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of the three endoplasmic reticulum (ER) transmembrane sensors of the unfolded protein response (UPR) that regulates protein synthesis, alleviates cellular ER stress and has been implicated in tumorigenesis and prolonged cancer cell survival. In this study, we report a series of 2-amino-3-amido-5-aryl-pyridines that we have identified as potent, selective, and orally bioavailable PERK inhibitors. Amongst the series studied herein, compound (28) a (R)-2-Amino-5-(4-(2-(3,5-difluorophenyl)-2-hydroxyacetamido)-2-ethylphenyl)-N-isopropylnicotinamide has demonstrated potent biochemical and cellular activity, robust pharmacokinetics and 70% oral bioavailability in mice. Given these data, this compound (28) was studied in the 786-O renal cell carcinoma xenograft model. We observed dose-dependent, statistically significant tumor growth inhibition, supporting the use of this tool compound in additional mechanistic studies.


Asunto(s)
Descubrimiento de Drogas , Piridinas/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , Administración Oral , Disponibilidad Biológica , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Piridinas/administración & dosificación , Piridinas/química , Relación Estructura-Actividad , eIF-2 Quinasa/metabolismo
3.
Circ Res ; 111(6): 708-17, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22828895

RESUMEN

RATIONALE: Atrial fibrillation (AF) is the most common cardiac arrhythmia, however the mechanism(s) causing AF remain poorly understood and therapy is suboptimal. The ryanodine receptor (RyR2) is the major calcium (Ca2+) release channel on the sarcoplasmic reticulum (SR) required for excitation-contraction coupling in cardiac muscle. OBJECTIVE: In the present study, we sought to determine whether intracellular diastolic SR Ca2+ leak via RyR2 plays a role in triggering AF and whether inhibiting this leak can prevent AF. METHODS AND RESULTS: We generated 3 knock-in mice with mutations introduced into RyR2 that result in leaky channels and cause exercise induced polymorphic ventricular tachycardia in humans [catecholaminergic polymorphic ventricular tachycardia (CPVT)]. We examined AF susceptibility in these three CPVT mouse models harboring RyR2 mutations to explore the role of diastolic SR Ca2+ leak in AF. AF was stimulated with an intra-esophageal burst pacing protocol in the 3 CPVT mouse models (RyR2-R2474S+/-, 70%; RyR2-N2386I+/-, 60%; RyR2-L433P+/-, 35.71%) but not in wild-type (WT) mice (P<0.05). Consistent with these in vivo results, there was a significant diastolic SR Ca2+ leak in atrial myocytes isolated from the CPVT mouse models. Calstabin2 (FKBP12.6) is an RyR2 subunit that stabilizes the closed state of RyR2 and prevents a Ca2+ leak through the channel. Atrial RyR2 from RyR2-R2474S+/- mice were oxidized, and the RyR2 macromolecular complex was depleted of calstabin2. The Rycal drug S107 stabilizes the closed state of RyR2 by inhibiting the oxidation/phosphorylation induced dissociation of calstabin2 from the channel. S107 reduced the diastolic SR Ca2+ leak in atrial myocytes and decreased burst pacing-induced AF in vivo. S107 did not reduce the increased prevalence of burst pacing-induced AF in calstabin2-deficient mice, confirming that calstabin2 is required for the mechanism of action of the drug. CONCLUSIONS: The present study demonstrates that RyR2-mediated diastolic SR Ca2+ leak in atrial myocytes is associated with AF in CPVT mice. Moreover, the Rycal S107 inhibited diastolic SR Ca2+ leak through RyR2 and pacing-induced AF associated with CPVT mutations.


Asunto(s)
Fibrilación Atrial/metabolismo , Calcio/metabolismo , Modelos Animales de Enfermedad , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Taquicardia Ventricular/metabolismo , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Cafeína/farmacología , Estimulación Cardíaca Artificial , Células Cultivadas , Electrocardiografía/efectos de los fármacos , Epinefrina/farmacología , Técnicas de Sustitución del Gen , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Immunoblotting , Ratones , Ratones Noqueados , Mutación , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Condicionamiento Físico Animal/fisiología , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Tiazepinas/farmacología
4.
Proc Natl Acad Sci U S A ; 107(22): 10274-9, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20479242

RESUMEN

The force frequency relationship (FFR), first described by Bowditch 139 years ago as the observation that myocardial contractility increases proportionally with increasing heart rate, is an important mediator of enhanced cardiac output during exercise. Individuals with heart failure have defective positive FFR that impairs their cardiac function in response to stress, and the degree of positive FFR deficiency correlates with heart failure progression. We have identified a mechanism for FFR involving heart rate dependent phosphorylation of the major cardiac sarcoplasmic reticulum calcium release channel/ryanodine receptor (RyR2), at Ser2814, by calcium/calmodulin-dependent serine/threonine kinase-delta (CaMKIIdelta). Mice engineered with an RyR2-S2814A mutation have RyR2 channels that cannot be phosphorylated by CaMKIIdelta, and exhibit a blunted positive FFR. Ex vivo hearts from RyR2-S2814A mice also have blunted positive FFR, and cardiomyocytes isolated from the RyR2-S2814A mice exhibit impaired rate-dependent enhancement of cytosolic calcium levels and fractional shortening. The cardiac RyR2 macromolecular complexes isolated from murine and human failing hearts have reduced CaMKIIdelta levels. These data indicate that CaMKIIdelta phosphorylation of RyR2 plays an important role in mediating positive FFR in the heart, and that defective regulation of RyR2 by CaMKIIdelta-mediated phosphorylation is associated with the loss of positive FFR in failing hearts.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca/fisiología , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Gasto Cardíaco/genética , Gasto Cardíaco/fisiología , Cartilla de ADN/genética , Frecuencia Cardíaca/genética , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Contracción Miocárdica/genética , Miocitos Cardíacos/fisiología , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética
5.
Clin Cancer Res ; 29(23): 4870-4882, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37733811

RESUMEN

PURPOSE: Tumors activate protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK, also called EIF2AK3) in response to hypoxia and nutrient deprivation as a stress-mitigation strategy. Here, we tested the hypothesis that inhibiting PERK with HC-5404 enhances the antitumor efficacy of standard-of-care VEGF receptor tyrosine kinase inhibitors (VEGFR-TKI). EXPERIMENTAL DESIGN: HC-5404 was characterized as a potent and selective PERK inhibitor, with favorable in vivo properties. Multiple renal cell carcinoma (RCC) tumor models were then cotreated with both HC-5404 and VEGFR-TKI in vivo, measuring tumor volume across time and evaluating tumor response by protein analysis and IHC. RESULTS: VEGFR-TKI including axitinib, cabozantinib, lenvatinib, and sunitinib induce PERK activation in 786-O RCC xenografts. Cotreatment with HC-5404 inhibited PERK in tumors and significantly increased antitumor effects of VEGFR-TKI across multiple RCC models, resulting in tumor stasis or regression. Analysis of tumor sections revealed that HC-5404 enhanced the antiangiogenic effects of axitinib and lenvatinib by inhibiting both new vasculature and mature tumor blood vessels. Xenografts that progress on axitinib monotherapy remain sensitive to the combination treatment, resulting in ∼20% tumor regression in the combination group. When tested across a panel of 18 RCC patient-derived xenograft (PDX) models, the combination induced greater antitumor effects relative to monotherapies. In this single animal study, nine out of 18 models responded with ≥50% tumor regression from baseline in the combination group. CONCLUSIONS: By disrupting an adaptive stress response evoked by VEGFR-TKI, HC-5404 presents a clinical opportunity to improve the antitumor effects of well-established standard-of-care therapies in RCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Animales , Humanos , Carcinoma de Células Renales/patología , Axitinib/farmacología , Axitinib/uso terapéutico , Neoplasias Renales/patología , Inhibidores de Proteínas Quinasas/uso terapéutico
6.
J Physiol ; 590(24): 6381-7, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23070698

RESUMEN

Enhancement of contractile force (inotropy) occurs in skeletal muscle following neuroendocrine release of catecholamines and activation of muscle ß-adrenergic receptors. Despite extensive study, the molecular mechanism underlying the inotropic response in skeletal muscle is not well understood. Here we show that phosphorylation of a single serine residue (S2844) in the sarcoplasmic reticulum (SR) Ca(2+) release channel/ryanodine receptor type 1 (RyR1) by protein kinase A (PKA) is critical for skeletal muscle inotropy. Treating fast twitch skeletal muscle from wild-type mice with the ß-receptor agonist isoproterenol (isoprenaline) increased RyR1 PKA phosphorylation, twitch Ca(2+) and force generation. In contrast, the enhanced muscle Ca(2+), force and in vivo muscle strength responses following isoproterenol stimulation were abrogated in RyR1-S2844A mice in which the serine in the PKA site in RyR1 was replaced with alanine. These data suggest that the molecular mechanism underlying skeletal muscle inotropy requires enhanced SR Ca(2+) release due to PKA phosphorylation of S2844 in RyR1.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Isoproterenol/farmacología , Contracción Muscular/efectos de los fármacos , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fuerza Muscular/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Musculares de Contracción Rápida/enzimología , Fosforilación , Mutación Puntual , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Serina , Factores de Tiempo
7.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G97-G104, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21960523

RESUMEN

Secretagogue-stimulated intracellular Ca(2+) signals are fundamentally important for initiating the secretion of the fluid and ion component of saliva from parotid acinar cells. The Ca(2+) signals have characteristic spatial and temporal characteristics, which are defined by the specific properties of Ca(2+) release mediated by inositol 1,4,5-trisphosphate receptors (InsP(3)R). In this study we have investigated the role of adenine nucleotides in modulating Ca(2+) release in mouse parotid acinar cells. In permeabilized cells, the Ca(2+) release rate induced by submaximal [InsP(3)] was increased by 5 mM ATP. Enhanced Ca(2+) release was not observed at saturating [InsP(3)]. The EC(50) for the augmented Ca(2+) release was ∼8 µM ATP. The effect was mimicked by nonhydrolysable ATP analogs. ADP and AMP also potentiated Ca(2+) release but were less potent than ATP. In acini isolated from InsP(3)R-2-null transgenic animals, the rate of Ca(2+) release was decreased under all conditions but now enhanced by ATP at all [InsP(3)]. In addition the EC(50) for ATP potentiation increased to ∼500 µM. These characteristics are consistent with the properties of the InsP(3)R-2 dominating the overall features of InsP(3)R-induced Ca(2+) release despite the expression of all isoforms. Finally, Ca(2+) signals were measured in intact parotid lobules by multiphoton microscopy. Consistent with the release data, carbachol-stimulated Ca(2+) signals were reduced in lobules exposed to experimental hypoxia compared with control lobules only at submaximal concentrations. Adenine nucleotide modulation of InsP(3)R in parotid acinar cells likely contributes to the properties of Ca(2+) signals in physiological and pathological conditions.


Asunto(s)
Células Acinares/efectos de los fármacos , Nucleótidos de Adenina/farmacología , Calcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Glándula Parótida/efectos de los fármacos , Células Acinares/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Hipoxia/metabolismo , Ratones , Glándula Parótida/metabolismo
8.
Appl Environ Microbiol ; 78(4): 1215-27, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22179247

RESUMEN

NADH oxidase (Nox) is a flavin-containing enzyme used by Streptococcus mutans to reduce dissolved oxygen encountered during growth in the oral cavity. In this study, we characterized the role of the NADH oxidase in the oxidative and acid stress responses of S. mutans. A nox-defective mutant strain of S. mutans and its parental strain, the genomic type strain UA159, were exposed to various oxygen concentrations at pH values of 5 and 7 to better understand the adaptive mechanisms used by the organism to withstand environmental pressures. With the loss of nox, the activities of oxygen stress response enzymes such as superoxide dismutase and glutathione oxidoreductase were elevated compared to those in controls, resulting in a greater adaptation to oxygen stress. In contrast, the loss of nox led to a decreased ability to grow in a low-pH environment despite an increased resistance to severe acid challenge. Analysis of the membrane fatty acid composition revealed that for both the nox mutant and UA159 parent strain, growth in an oxygen-rich environment resulted in high proportions of unsaturated membrane fatty acids, independent of external pH. The data indicate that S. mutans membrane fatty acid composition is responsive to oxidative stress, as well as changes in environmental pH, as previously reported (E. M. Fozo and R. G. Quivey, Jr., Appl. Environ. Microbiol. 70:929-936, 2004). The heightened ability of the nox strain to survive acidic and oxidative environmental stress suggests a multifaceted response system that is partially dependent on oxygen metabolites.


Asunto(s)
Ácidos/toxicidad , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Oxígeno/toxicidad , Streptococcus mutans/efectos de los fármacos , Streptococcus mutans/fisiología , Estrés Fisiológico , Membrana Celular/química , Ácidos Grasos/análisis , Concentración de Iones de Hidrógeno , Viabilidad Microbiana/efectos de los fármacos , Complejos Multienzimáticos/deficiencia , Mutación , NADH NADPH Oxidorreductasas/deficiencia , Estrés Oxidativo , Streptococcus mutans/enzimología
9.
Pharmaceutics ; 14(10)2022 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-36297668

RESUMEN

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of three endoplasmic reticulum (ER) transmembrane sensors of the unfolded protein response (UPR) responsible for regulating protein synthesis and alleviating ER stress. PERK has been implicated in tumorigenesis, cancer cell survival as well metabolic diseases such as diabetes. The structure-based design and optimization of a novel mandelamide-derived pyrrolopyrimidine series of PERK inhibitors as described herein, resulted in the identification of compound 26, a potent, selective, and orally bioavailable compound suitable for interrogating PERK pathway biology in vitro and in vivo, with pharmacokinetics suitable for once-a-day oral dosing in mice.

10.
J Biol Chem ; 285(48): 37927-38, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20876535

RESUMEN

Ca(2+) release through inositol 1,4,5-trisphosphate receptors (InsP(3)R) can be modulated by numerous factors, including input from other signal transduction cascades. These events shape the spatio-temporal characteristics of the Ca(2+) signal and provide fidelity essential for the appropriate activation of effectors. In this study, we investigate the regulation of Ca(2+) release via InsP(3)R following activation of cyclic nucleotide-dependent kinases in the presence and absence of expression of a binding partner InsP(3)R-associated cGMP kinase substrate (IRAG). cGMP-dependent kinase (PKG) phosphorylation of only the S2+ InsP(3)R-1 subtype resulted in enhanced Ca(2+) release in the absence of IRAG expression. In contrast, IRAG bound to each InsP(3)R subtype, and phosphorylation of IRAG by PKG attenuated Ca(2+) release through all InsP(3)R subtypes. Surprisingly, simply the expression of IRAG attenuated phosphorylation and inhibited the enhanced Ca(2+) release through InsP(3)R-1 following cAMP-dependent protein kinase (PKA) activation. In contrast, IRAG expression did not influence the PKA-enhanced activity of the InsP(3)R-2. Phosphorylation of IRAG resulted in reduced Ca(2+) release through all InsP(3)R subtypes during concurrent activation of PKA and PKG, indicating that IRAG modulation is dominant under these conditions. These studies yield mechanistic insight into how cells with various complements of proteins integrate and prioritize signals from ubiquitous signaling pathways.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Fosfoproteínas/metabolismo , Animales , Células COS , Calcio/metabolismo , Línea Celular , Pollos , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Proteínas de la Membrana , Ratones , Fosfoproteínas/genética , Fosforilación , Ratas
11.
J Biol Chem ; 284(37): 25116-25, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19608738

RESUMEN

Protein kinase A (PKA) phosphorylation of inositol 1,4,5-trisphosphate receptors (InsP(3)Rs) represents a mechanism for shaping intracellular Ca(2+) signals following a concomitant elevation in cAMP. Activation of PKA results in enhanced Ca(2+) release in cells that express predominantly InsP(3)R2. PKA is known to phosphorylate InsP(3)R2, but the molecular determinants of this effect are not known. We have expressed mouse InsP(3)R2 in DT40-3KO cells that are devoid of endogenous InsP(3)R and examined the effects of PKA phosphorylation on this isoform in unambiguous isolation. Activation of PKA increased Ca(2+) signals and augmented the single channel open probability of InsP(3)R2. A PKA phosphorylation site unique to the InsP(3)R2 was identified at Ser(937). The enhancing effects of PKA activation on this isoform required the phosphorylation of Ser(937), since replacing this residue with alanine eliminated the positive effects of PKA activation. These results provide a mechanism responsible for the enhanced Ca(2+) signaling following PKA activation in cells that express predominantly InsP(3)R2.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Serina/química , Secuencia de Aminoácidos , Animales , Células COS , Calcio/química , Pollos , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/química , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Fosforilación , Isoformas de Proteínas , Transducción de Señal
12.
Pflugers Arch ; 460(2): 467-80, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20179962

RESUMEN

Ryanodine receptors (RyR) are intracellular Ca2+-permeable channels that provide the sarcoplasmic reticulum Ca2+ release required for skeletal and cardiac muscle contractions. RyR1 underlies skeletal muscle contraction, and RyR2 fulfills this role in cardiac muscle. Over the past 20 years, numerous mutations in both RyR isoforms have been identified and linked to skeletal and cardiac diseases. Malignant hyperthermia, central core disease, and catecholaminergic polymorphic ventricular tachycardia have been genetically linked to mutations in either RyR1 or RyR2. Thus, RyR channelopathies are both of interest because they cause significant human diseases and provide model systems that can be studied to elucidate important structure-function relationships of these ion channels.


Asunto(s)
Canalopatías/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Calcio/metabolismo , Canalopatías/tratamiento farmacológico , Humanos , Hipertermia Maligna/genética , Hipertermia Maligna/fisiopatología , Ratones , Ratones Transgénicos , Mutación , Contracción Miocárdica , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos , Retículo Sarcoplasmático/fisiología , Taquicardia Ventricular/genética
13.
Mol Pharmacol ; 76(5): 992-7, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19666700

RESUMEN

The highly specialized metabotropic glutamate receptor type 6 (mGluR6) is postsynaptically localized and expressed only in the dendrites of ON bipolar cells. Upon activation of mGluR6 by glutamate released from photoreceptors, a nonselective cation channel is inhibited, causing these cells to hyperpolarize. Mutations in this gene have been implicated in the development of congenital stationary night blindness type 1 (CSNB1). We investigated five known mGluR6 point mutants that lead to CSNB1 to determine the molecular mechanism of each phenotype. In agreement with other studies, four mutants demonstrated trafficking impairment. However, mGluR6 E775K (E781K in humans) suggested no trafficking or signaling deficiencies measured by our initial assays. Most importantly, our results indicate a switch in G-protein coupling, in which E775K loses G(o) coupling but retains coupling to G(i), which may explain the phenotype.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Ceguera Nocturna/genética , Mutación Puntual/fisiología , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Línea Celular , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Humanos , Mutagénesis Sitio-Dirigida , Ceguera Nocturna/congénito , Ceguera Nocturna/metabolismo , Fenotipo , Transporte de Proteínas/genética , Ratas , Ganglio Cervical Superior/fisiología
14.
Methods ; 46(3): 177-82, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18929664

RESUMEN

Inositol 1,4,5-trisphosphate receptors (InsP3R) are a family of ubiquitously expressed intracellular Ca2+ channels. Isoform-specific properties of the three family members may play a prominent role in defining the rich diversity of the spatial and temporal characteristics of intracellular Ca2+ signals. Studying the properties of the particular family members is complicated because individual receptor isoforms are typically never expressed in isolation. In this article, we discuss strategies for studying Ca2+ release through individual InsP3R family members with particular reference to methods applicable following expression of recombinant InsP3R and mutant constructs in the DT40-3KO cell line, an unambiguously null InsP3R expression system.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Animales , Línea Celular Tumoral , Pollos , Fura-2/análogos & derivados , Fura-2/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Permeabilidad , Fotólisis , Isoformas de Proteínas/metabolismo , Ratas
17.
Curr Mol Pharmacol ; 8(2): 133-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25981977

RESUMEN

Voltage gated calcium channels are essential for cardiac physiology by serving as sarcolemma- restricted gatekeepers for calcium in cardiac myocytes. Activation of the L-type voltagegated calcium channel provides the calcium entry required for excitation-contraction coupling and contributes to the plateau phase of the cardiac action potential. Given these critical physiological roles, subtle disturbances in L-type channel function can lead to fatal cardiac arrhythmias. Indeed, numerous human arrhythmia syndromes have been linked to mutations in the L-type channel leading to gain-of-function or loss-of-function mutations. In this review, we discuss the current state of knowledge regarding these mutations present in Timothy Syndrome, Long and Short QT Syndromes, Brugada Syndrome and Early Repolarization Syndrome. We discuss the pathological consequences of the mutations, the biophysical effects of the mutations on the channel as well as possible therapeutic considerations and challenges for future studies.


Asunto(s)
Arritmias Cardíacas/genética , Canales de Calcio Tipo L/genética , Predisposición Genética a la Enfermedad/genética , Mutación , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Trastorno Autístico/fisiopatología , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de Brugada/fisiopatología , Calcio/metabolismo , Canales de Calcio Tipo L/fisiología , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología , Modelos Genéticos , Sindactilia/genética , Sindactilia/metabolismo , Sindactilia/fisiopatología
18.
FEMS Microbiol Lett ; 209(1): 75-9, 2002 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-12007657

RESUMEN

Galactokinase and beta-galactosidase-deficient strains of Streptococcus salivarius were constructed to define the pathways for lactose and galactose catabolism. It was found that S. salivarius does not possess a lactose-specific phosphoenolpyruvate phosphotransferase system (PTS), that intracellular lactose was hydrolyzed by beta-galactosidase, and that galactose is catabolized exclusively through the Leloir pathway. The lack of a high-affinity PTS for lactose may reflect the higher availability of the substrates to soft tissue organisms, such as S. salivarius, compared to dental plaque bacteria.


Asunto(s)
Proteínas Bacterianas/metabolismo , Galactosa/metabolismo , Lactosa/metabolismo , Streptococcus/metabolismo , Galactoquinasa/deficiencia , Galactoquinasa/metabolismo , Humanos , Operón Lac , Boca/microbiología , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato , Streptococcus/genética , Streptococcus/crecimiento & desarrollo , beta-Galactosidasa/deficiencia , beta-Galactosidasa/metabolismo
19.
J Am Coll Cardiol ; 64(1): 66-79, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24998131

RESUMEN

BACKGROUND: BrS is an inherited sudden cardiac death syndrome. Less than 35% of BrS probands have genetically identified pathogenic variants. Recent evidence has implicated SCN10A, a neuronal sodium channel gene encoding Nav1.8, in the electrical function of the heart. OBJECTIVES: The purpose of this study was to test the hypothesis that SCN10A variants contribute to the development of Brugada syndrome (BrS). METHODS: Clinical analysis and direct sequencing of BrS susceptibility genes were performed for 150 probands and family members as well as >200 healthy controls. Expression and coimmunoprecipitation studies were performed to functionally characterize the putative pathogenic mutations. RESULTS: We identified 17 SCN10A mutations in 25 probands (20 male and 5 female); 23 of the 25 probands (92.0%) displayed overlapping phenotypes. SCN10A mutations were found in 16.7% of BrS probands, approaching our yield for SCN5A mutations (20.1%). Patients with BrS who had SCN10A mutations were more symptomatic and displayed significantly longer PR and QRS intervals compared with SCN10A-negative BrS probands. The majority of mutations localized to the transmembrane-spanning regions. Heterologous coexpression of wild-type (WT) SCN10A with WT-SCN5A in HEK cells caused a near doubling of sodium channel current compared with WT-SCN5A alone. In contrast, coexpression of SCN10A mutants (R14L and R1268Q) with WT-SCN5A caused a 79.4% and 84.4% reduction in sodium channel current, respectively. The coimmunoprecipitation studies provided evidence for the coassociation of Nav1.8 and Nav1.5 in the plasma membrane. CONCLUSIONS: Our study identified SCN10A as a major susceptibility gene for BrS, thus greatly enhancing our ability to genotype and risk stratify probands and family members.


Asunto(s)
Síndrome de Brugada/diagnóstico , Síndrome de Brugada/genética , Variación Genética/genética , Mutación Missense/genética , Canal de Sodio Activado por Voltaje NAV1.8/genética , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
20.
Heart Rhythm ; 10(7): 1054-62, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23499631

RESUMEN

BACKGROUND: Wenxin Keli (WK), a Chinese herb extract, is reported to be effective in the treatment of atrial and ventricular cardiac arrhythmias. Recent studies suggest that WK inhibits the transient potassium outward current (I(to)). OBJECTIVE: To examine the effectiveness of WK, alone and in combination with quinidine, to suppress arrhythmogenesis in an experimental model of Brugada syndrome (BrS). METHODS: Action potential and electrocardiographic recordings were obtained from epicardial and endocardial sites of coronary-perfused canine right ventricular wedge preparations. The Ito agonist NS5806 (10-15 µM) was used to pharmacologically mimic a genetic predisposition to BrS. RESULTS: The Ito agonist induced Phase 2 reentry (P2R) in 13/19 preparations and polymorphic ventricular tachycardia (pVT) in 11/19 wedge preparations. WK (10 g/L) suppressed P2R and pVT in 100% (3/3) of preparations. A lower concentration of WK (5 g/L) suppressed P2R in 60% (3/5) and pVT in 50% (2/4), but in combination with a low concentration of quinidine (5 µM), was 100% effective in suppressing P2R and pVT. Quinidine alone suppressed P2R and pVT in 60% (3/5) and 50% (2/4), respectively, and in combination with WK (5 g/L) suppressed P2R and pVT by 80% (4/5) and 75% (3/4), respectively. WK reduced Ito, the L-type calcium current, and contractility in single cardiomyocytes, but dose-dependently increased contractility in intact wedge preparations, an effect mimicked by tyramine. CONCLUSIONS: Our data provide support for the hypothesis that WK, particularly in combination with quinidine, effectively suppresses arrhythmogenesis in an experimental model of BrS via inhibition of Ito and indirect adrenergic sympathomimetic effects.


Asunto(s)
Síndrome de Brugada/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Quinidina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Antiarrítmicos/farmacología , Síndrome de Brugada/patología , Síndrome de Brugada/fisiopatología , Modelos Animales de Enfermedad , Perros , Miocitos Cardíacos/patología , Técnicas de Placa-Clamp
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA