Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Ann Noninvasive Electrocardiol ; 28(5): e13080, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37571804

RESUMEN

BACKGROUND: Congenital Long QT Syndrome (LQTS) is a hereditary arrhythmic disorder. We aimed to assess the performance of current genetic variant annotation scores among LQTS patients and their predictive impact. METHODS: We evaluated 2025 patients with unique mutations for LQT1-LQT3. A patient-specific score was calculated for each of four established genetic variant annotation algorithms: CADD, SIFT, REVEL, and PolyPhen-2. The scores were tested for the identification of LQTS and their predictive performance for cardiac events (CE) and life-threatening events (LTE) and then compared with the predictive performance of LQTS categorization based on mutation location/function. Score performance was tested using Harrell's C-index. RESULTS: A total of 917 subjects were classified as LQT1, 838 as LQT2, and 270 as LQT3. The identification of a pathogenic variant occurred in 99% with CADD, 92% with SIFT, 100% with REVEL, and 86% with PolyPhen-2. However, none of the genetic scores correlated with the risk of CE (Harrell's C-index: CADD = 0.50, SIFT = 0.51, REVEL = 0.50, and PolyPhen-2 = 0.52) or LTE (Harrell's C-index: CADD = 0.50, SIFT = 0.53, REVEL = 0.54, and PolyPhen-2 = 0.52). In contrast, high-risk mutation categorization based on location/function was a powerful independent predictor of CE (HR = 1.88; p < .001) and LTE (HR = 1.89, p < .001). CONCLUSION: In congenital LQTS patients, well-established algorithms (CADD, SIFT, REVEL, and PolyPhen-2) were able to identify the majority of the causal variants as pathogenic. However, the scores did not predict clinical outcomes. These results indicate that mutation location/functional assays are essential for accurate interpretation of the risk associated with LQTS mutations.


Asunto(s)
Electrocardiografía , Síndrome de QT Prolongado , Humanos , Genotipo , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/complicaciones
2.
J Mol Cell Cardiol ; 138: 283-290, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785237

RESUMEN

The slow voltage-gated potassium channel (IKs) is composed of the KCNQ1 and KCNE1 subunits and is one of the major repolarizing currents in the heart. Activation of protein kinase C (PKC) has been linked to cardiac arrhythmias. Although PKC has been shown to be a regulator of a number of cardiac channels, including IKs, little is known about regulation of the channel by specific isoforms of PKC. Here we studied the role of different PKC isoforms on IKs channel membrane localization and function. Our studies focused on PKC isoforms that translocate to the plasma membrane in response to Gq-coupled receptor (GqPCR) stimulation: PKCα, PKCßI, PKCßII and PKCε. Prolonged stimulation of GqPCRs has been shown to decrease IKs membrane expression, but the specific role of each PKC isoform is unclear. Here we show that stimulation of calcium-dependent isoforms of PKC (cPKC) but not PKCε mimic receptor activation. In addition, we show that general PKCß (LY-333531) and PKCßII inhibitors but not PKCα or PKCßI inhibitors blocked the effect of cPKC on the KCNQ1/KCNE1 channel. PKCß inhibitors also blocked GqPCR-mediated decrease in channel membrane expression in cardiomyocytes. Direct activation of PKCßII using constitutively active PKCßII construct mimicked agonist-induced decrease in membrane expression and channel function, while dominant negative PKCßII showed no effect. This suggests that the KCNQ1/KCNE1 channel was not regulated by basal levels of PKCßII activity. Our results indicate that PKCßII is a specific regulator of IKs membrane localization. PKCßII expression and activation are strongly increased in many disease states, including heart disease and diabetes. Thus, our results suggest that PKCßII inhibition may protect against acquired QT prolongation associated with heart disease.


Asunto(s)
Membrana Celular/metabolismo , Canal de Potasio KCNQ1/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteína Quinasa C beta/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fenilefrina/farmacología , Proteína Quinasa C beta/antagonistas & inhibidores , Ratas
3.
J Mol Cell Cardiol ; 129: 314-325, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30898664

RESUMEN

Statins, in addition to their cholesterol lowering effects, can prevent isoprenylation of Rab GTPase proteins, a key protein family for the regulation of protein trafficking. Rab-GTPases have been shown to be involved in the control of membrane expression level of ion channels, including one of the major cardiac repolarizing channels, IKs. Decreased IKs function has been observed in a number of disease states and associated with increased propensity for arrhythmias, but the mechanism underlying IKs decrease remains elusive. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in variety of human diseases and have been suggested to acutely regulate IKs function. We hypothesize that chronic cPKC stimulation leads to Rab-mediated decrease in IKs membrane expression, and that can be prevented by statins. In this study we show that chronic cPKC stimulation caused a dramatic Rab5 GTPase-dependent decrease in plasma membrane localization of the IKs pore forming subunit KCNQ1, reducing IKs function. Our data indicates fluvastatin inhibition of Rab5 restores channel localization and function after cPKC-mediated channel internalization. Our results indicate a novel statin anti-arrhythmic effect that would be expected to inhibit pathological electrical remodeling in a number of disease states associated with high cPKC activation. Because Rab-GTPases are important regulators of membrane trafficking they may underlie other statin pleiotropic effects.


Asunto(s)
Calcio/metabolismo , Endocitosis , Fluvastatina/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Proteína Quinasa C/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas
4.
J Mol Cell Cardiol ; 79: 203-11, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25479336

RESUMEN

BACKGROUND: The most common inherited cardiac arrhythmia, LQT1, is due to IKs potassium channel mutations and is linked to high risk of adrenergic-triggered cardiac events. We recently showed that although exercise-triggered events are very well treated by ß-blockers for these patients, acute arousal-triggered event rate were not significantly reduced after beta-blocker treatment, suggesting that the mechanisms underlying arousal-triggered arrhythmias may be different from those during exercise. IKs is strongly regulated by ß-adrenergic receptor (ß-AR) signaling, but little is known about the role of α1-AR-mediated regulation. METHODS AND RESULTS: Here we show, using a combination of cellular electrophysiology and computational modeling, that IKs phosphorylation and α1-AR regulation via activation of calcium-dependent PKC isoforms (cPKC) may be a key mechanism to control channel voltage-dependent activation and consequently action potential duration (APD) in response to adrenergic-stimulus. We show that simulated mutation-specific combined adrenergic effects (ß+α) on APD were strongly correlated to acute stress-triggered cardiac event rate for patients while ß-AR effects alone were not. CONCLUSION: We were able to show that calcium-dependent PKC signaling is key to normal QT shortening during acute arousal and when impaired, correlates with increased rate of sudden arousal-triggered cardiac events. Our study suggests that the acute α1-AR-cPKC regulation of IKs is important for QT shortening in "fight-or-flight" response and is linked to decreased risk of sudden emotion/arousal-triggered cardiac events in LQT1 patients.


Asunto(s)
Nivel de Alerta , Calcio/metabolismo , Emociones , Activación del Canal Iónico , Canal de Potasio KCNQ1/metabolismo , Síndrome de QT Prolongado/fisiopatología , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteína Quinasa C/metabolismo , Potenciales de Acción , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Isoenzimas/metabolismo , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Fosforilación , Canales de Potasio con Entrada de Voltaje/genética , Modelos de Riesgos Proporcionales , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Factores de Riesgo , Transducción de Señal
5.
Biochem Biophys Res Commun ; 465(3): 464-70, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26277396

RESUMEN

Protein kinase C (PKC) plays key roles in the regulation of signal transduction and cellular function in various cell types. At least ten PKC isoforms have been identified and intracellular localization and trafficking of these individual isoforms are important for regulation of enzyme activity and substrate specificity. PKC can be activated downstream of Gq-protein coupled receptor (GqPCR) signaling and translocate to various cellular compartments including plasma membrane (PM). Recent reports suggested that different types of GqPCRs would activate different PKC isoforms (classic, novel and atypical PKCs) with different trafficking patterns. However, the knowledge of isoform-specific activation of PKC by each GqPCR is limited. α1-Adrenoceptor (α1-AR) is one of the GqPCRs highly expressed in the cardiovascular system. In this study, we examined the isoform-specific dynamic translocation of PKC in living HEK293T cells by α1-AR stimulation (α1-ARS). Rat PKCα, ßI, ßII, δ, ε and ζ fused with GFP at C-term were co-transfected with human α1A-AR into HEK293T cells. The isoform-specific dynamic translocation of PKC in living HEK293T cells by α1-ARS using phenylephrine was measured by confocal microscopy. Before stimulation, GFP-PKCs were localized at cytosolic region. α1-ARS strongly and rapidly translocated a classical PKC (cPKC), PKCα, (<30 s) to PM, with PKCα returning diffusively into the cytosol within 5 min. α1-ARS rapidly translocated other cPKCs, PKCßI and PKCßII, to the PM (<30 s), with sustained membrane localization. One novel PKC (nPKC), PKCε, but not another nPKC, PKCδ, was translocated by α1-AR stimulation to the PM (<30 s) and its membrane localization was also sustained. Finally, α1-AR stimulation did not cause a diacylglycerol-insensitive atypical PKC, PKCζ translocation. Our data suggest that PKCα, ß and ε activation may underlie physiological and pathophysiological responses of α1-AR signaling for the phosphorylation of membrane-associated substrates including ion-channel and transporter proteins in the cardiovascular system.


Asunto(s)
Membrana Celular/metabolismo , Proteína Quinasa C/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal/fisiología , Células HEK293 , Humanos , Isoformas de Proteínas/metabolismo , Transporte de Proteínas/fisiología
6.
Circ Res ; 110(1): 59-70, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22076634

RESUMEN

RATIONALE: The Rad-Gem/Kir-related family (RGKs) consists of small GTP-binding proteins that strongly inhibit the activity of voltage-gated calcium channels. Among RGKs, Rem1 is strongly and specifically expressed in cardiac tissue. However, the physiological role and regulation of RGKs, and Rem1 in particular, are largely unknown. OBJECTIVE: To determine if Rem1 function is physiologically regulated by adrenergic signaling and thus impacts voltage-gated L-type calcium channel (VLCC) activity in the heart. METHODS AND RESULTS: We found that activation of protein kinase D1, a protein kinase downstream of α(1)-adrenergic signaling, leads to direct phosphorylation of Rem1 at Ser18. This results in an increase of the channel activity and plasma membrane expression observed by using a combination of electrophysiology, live cell confocal microscopy, and immunohistochemistry in heterologous expression system and neonatal cardiomyocytes. In addition, we show that stimulation of α(1)-adrenergic receptor-protein kinase D1-Rem1 signaling increases transverse-tubule VLCC expression that results in increased L-type Ca(2+) current density in adult ventricular myocytes. CONCLUSION: The α(1)-adrenergic stimulation releases Rem1 inhibition of VLCCs through direct phosphorylation of Rem1 at Ser18 by protein kinase D1, resulting in an increase of the channel activity and transverse-tubule expression. Our results uncover a novel molecular regulatory mechanism of VLCC trafficking and function in the heart and provide the first demonstration of physiological regulation of RGK function.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Miocitos Cardíacos/fisiología , Proteínas Quinasas/fisiología , Transporte de Proteínas/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Transducción de Señal/fisiología , Animales , Membrana Celular/fisiología , Células Cultivadas , Masculino , Microtúbulos/fisiología , Modelos Animales , Proteínas de Unión al GTP Monoméricas/fisiología , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Fosforilación , Proteína Quinasa C , Ratas , Ratas Sprague-Dawley
7.
Circulation ; 125(16): 1988-96, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22456477

RESUMEN

BACKGROUND: ß-Adrenergic stimulation is the main trigger for cardiac events in type 1 long-QT syndrome (LQT1). We evaluated a possible association between ion channel response to ß-adrenergic stimulation and clinical response to ß-blocker therapy according to mutation location. METHODS AND RESULTS: The study sample comprised 860 patients with genetically confirmed mutations in the KCNQ1 channel. Patients were categorized into carriers of missense mutations located in the cytoplasmic loops (C loops), membrane-spanning domain, C/N terminus, and nonmissense mutations. There were 27 aborted cardiac arrest and 78 sudden cardiac death events from birth through 40 years of age. After multivariable adjustment for clinical factors, the presence of C-loop mutations was associated with the highest risk for aborted cardiac arrest or sudden cardiac death (hazard ratio versus nonmissense mutations=2.75; 95% confidence interval, 1.29-5.86; P=0.009). ß-Blocker therapy was associated with a significantly greater reduction in the risk of aborted cardiac arrest or sudden cardiac death among patients with C-loop mutations than among all other patients (hazard ratio=0.12; 95% confidence interval, 0.02-0.73; P=0.02; and hazard ratio=0.82; 95% confidence interval, 0.31-2.13; P=0.68, respectively; P for interaction=0.04). Cellular expression studies showed that membrane spanning and C-loop mutations produced a similar decrease in current, but only C-loop mutations showed a pronounced reduction in channel activation in response to ß-adrenergic stimulation. CONCLUSIONS: Patients with C-loop missense mutations in the KCNQ1 channel exhibit a high risk for life-threatening events and derive a pronounced benefit from treatment with ß-blockers. Reduced channel activation after sympathetic activation can explain the increased clinical risk and response to therapy in patients with C-loop mutations.


Asunto(s)
Canal de Potasio KCNQ1/genética , Mutación , Síndrome de Romano-Ward/genética , Adolescente , Antagonistas Adrenérgicos beta/uso terapéutico , Adulto , Niño , Femenino , Predisposición Genética a la Enfermedad , Paro Cardíaco/tratamiento farmacológico , Paro Cardíaco/genética , Humanos , Masculino , Riesgo , Síndrome de Romano-Ward/tratamiento farmacológico , Resultado del Tratamiento , Adulto Joven
8.
Carbon Balance Manag ; 18(1): 7, 2023 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-37062006

RESUMEN

BACKGROUND: Sequestration of carbon on forest land is a common and practical component within many climate action plans developed by state or municipal governments. Initial planning often identifies the general magnitude of sequestration expected given the scope of the project. Because age plays a key role in forest carbon dynamics, we summarize both the carbon stock and accumulation rates in live trees by age class and region, allowing managers and policymakers to assess the influence of forest age class structure on forest carbon storage as represented in current inventories. State-level information is provided in supplementary tables. RESULTS: Average regional aboveground live tree carbon stocks (represented on a per area basis) range from 11.6 tC/ha in the Great Plains to 130 tC/ha in the Pacific Northwest West (west-side of Cascades) and increase with age in all regions, although in three regions carbon stock declined in the oldest age class. Regional average annual net change in live aboveground tree carbon varies from a low of - 0.18 tC /ha/yr in the Rocky Mountain South region to a high value of 1.74 tC/ha/yr in Pacific Northwest West. In all regions except Rocky Mountain South, accumulation rates are highest in the younger age classes and decline with age, with older age classes in several western regions showing negative rates. In the Southeast and Pacific Northwest West, intermediate age classes exhibit lower rates, likely due to harvesting activity. CONCLUSIONS: Aboveground live tree carbon stocks increase and rates of average change decrease with age with few exceptions; this pattern holds when examining hardwood and softwood types individually. Because multiple forest management objectives are often considered and tradeoffs need to be assessed, we recommend considering both measures-standing stock and average annual change-of carbon storage. The relative importance of each component depends on management and policy objectives and the time frame related to those objectives. Harvesting and natural disturbance also affect forest carbon stock and change and may need to be considered if developing projections of potential carbon storage. We present forest carbon summaries at a scale and scope to meet information needs of managers and policymakers.

9.
Circ Res ; 106(7): 1190-6, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20185796

RESUMEN

RATIONALE: The mitochondrial ATP sensitive potassium channel (mK(ATP)) is implicated in cardioprotection by ischemic preconditioning (IPC), but the molecular identity of the channel remains controversial. The validity of current methods to assay mK(ATP) activity is disputed. OBJECTIVE: We sought to develop novel methods to assay mK(ATP) activity and its regulation. METHODS AND RESULTS: Using a thallium (Tl(+))-sensitive fluorophore, we developed a novel Tl(+) flux based assay for mK(ATP) activity, and used this assay probe several aspects of mK(ATP) function. The following key observations were made. (1) Time-dependent run down of mK(ATP) activity was reversed by phosphatidylinositol-4,5-bisphosphate (PIP(2)). (2) Dose responses of mK(ATP) to nucleotides revealed a UDP EC(50) of approximately 20 micromol/L and an ATP IC(50) of approximately 5 micromol/L. (3) The antidepressant fluoxetine (Prozac) inhibited mK(ATP) (IC(50)=2.4 micromol/L). Fluoxetine also blocked cardioprotection triggered by IPC, but did not block protection triggered by a mK(ATP)-independent stimulus. The related antidepressant zimelidine was without effect on either mK(ATP) or IPC. CONCLUSIONS: The Tl(+) flux mK(ATP) assay was validated by correlation with a classical mK(ATP) channel osmotic swelling assay (R(2)=0.855). The pharmacological profile of mK(ATP) (response to ATP, UDP, PIP(2), and fluoxetine) is consistent with that of an inward rectifying K(+) channel (K(IR)) and is somewhat closer to that of the K(IR)6.2 than the K(IR)6.1 isoform. The effect of fluoxetine on mK(ATP)-dependent cardioprotection has implications for the growing use of antidepressants in patients who may benefit from preconditioning.


Asunto(s)
Bioensayo/métodos , Mitocondrias Cardíacas/metabolismo , Canales de Potasio/metabolismo , Potasio/metabolismo , Espectrometría de Fluorescencia , Talio/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antidepresivos de Segunda Generación/farmacología , Benzotiazoles , Cumarinas , Colorantes Fluorescentes , Fluoxetina/farmacología , Glicina/análogos & derivados , Técnicas In Vitro , Precondicionamiento Isquémico Miocárdico , Cinética , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Dilatación Mitocondrial , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Ósmosis , Perfusión , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Uridina Difosfato/metabolismo
10.
Circ Res ; 107(4): 532-9, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20576935

RESUMEN

RATIONALE: Excess signaling through cardiac Gbetagamma subunits is an important component of heart failure (HF) pathophysiology. They recruit elevated levels of cytosolic G protein-coupled receptor kinase (GRK)2 to agonist-stimulated beta-adrenergic receptors (beta-ARs) in HF, leading to chronic beta-AR desensitization and downregulation; these events are all hallmarks of HF. Previous data suggested that inhibiting Gbetagamma signaling and its interaction with GRK2 could be of therapeutic value in HF. OBJECTIVE: We sought to investigate small molecule Gbetagamma inhibition in HF. METHODS AND RESULTS: We recently described novel small molecule Gbetagamma inhibitors that selectively block Gbetagamma-binding interactions, including M119 and its highly related analog, gallein. These compounds blocked interaction of Gbetagamma and GRK2 in vitro and in HL60 cells. Here, we show they reduced beta-AR-mediated membrane recruitment of GRK2 in isolated adult mouse cardiomyocytes. Furthermore, M119 enhanced both adenylyl cyclase activity and cardiomyocyte contractility in response to beta-AR agonist. To evaluate their cardiac-specific effects in vivo, we initially used an acute pharmacological HF model (30 mg/kg per day isoproterenol, 7 days). Concurrent daily injections prevented HF and partially normalized cardiac morphology and GRK2 expression in this acute HF model. To investigate possible efficacy in halting progression of preexisting HF, calsequestrin cardiac transgenic mice (CSQ) with extant HF received daily injections for 28 days. The compound alone halted HF progression and partially normalized heart size, morphology, and cardiac expression of HF marker genes (GRK2, atrial natriuretic factor, and beta-myosin heavy chain). CONCLUSIONS: These data suggest a promising therapeutic role for small molecule inhibition of pathological Gbetagamma signaling in the treatment of HF.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Insuficiencia Cardíaca/prevención & control , Transducción de Señal/fisiología , Animales , Ciclohexanos/farmacología , Ciclohexanos/uso terapéutico , Progresión de la Enfermedad , Femenino , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Células HL-60 , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos , Transducción de Señal/efectos de los fármacos , Xantenos/farmacología , Xantenos/uso terapéutico , Xenopus
11.
J Cardiovasc Electrophysiol ; 22(2): 193-200, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20662986

RESUMEN

UNLABELLED: BACKGROUND: Data regarding possible ion channel mechanisms that predispose to ventricular tachyarrhythmias in patients with phenotype-negative long-QT syndrome (LQTS) are limited. METHODS AND RESULTS: We carried out cellular expression studies for the S349W mutation in the KCNQ1 channel, which was identified in 15 patients from the International LQTS Registry who experienced a high rate of cardiac events despite lack of significant QTc prolongation. The clinical outcome of S349W mutation carriers was compared with that of QTc-matched carriers of haploinsufficient missense (n = 30) and nonsense (n = 45) KCNQ1 mutations. The channels containing the mutant S349W subunit showed a mild reduction in current (<50%), in the haploinsuficient range, with an increase in maximal conductance compared with wild-type channels. In contrast, expression of the S349W mutant subunit produced a pronounced effect on both the voltage dependence of activation and the time constant of activation, while haploinsuficient channels showed no effect on either parameter. The cumulative probability of cardiac events from birth through age 20 years was significantly higher among S349W mutation carriers (58%) as compared with carriers of QTc-matched haploinsufficent missense (21%, P = 0.004) and nonsense (25%, P = 0.01) mutations. CONCLUSIONS: The S349W mutation in the KCNQ1 potassium channel exerts a relatively mild effect on the ion channel current, whereas an increase in conductance compensates for impaired voltage activation of the channel. The changes observed in voltage activation of the channel may underlie the mechanisms predisposing to arrhythmic risk among LQTS patients with a normal-range QTc.


Asunto(s)
Muerte Súbita Cardíaca , Predisposición Genética a la Enfermedad/genética , Activación del Canal Iónico/genética , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Niño , Femenino , Genotipo , Humanos , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple
12.
Ecol Appl ; 21(4): 1154-61, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21774420

RESUMEN

One method of increasing forest carbon stocks that is often discussed is increasing stocking levels on existing forested lands. However, estimates of the potential increases in forest carbon sequestration as a result of increased stocking levels are not readily available. Using the USDA Forest Service's Forest Inventory and Analysis data coupled with the Forest Vegetation Simulator, we estimate that, for a seven-state region in the northeastern United States, timberland contains about 1768 Tg of carbon in aboveground live biomass across all stocking classes. If all medium and understocked stands had the carbon density of fully stocked stands, an additional 453 Tg of carbon would be stored. While the carbon gains per unit area are greatest for understocked stands, generally fewer than 10% of stands are in this condition. The increase in carbon storage per unit area is smaller for stands in the medium stocked class, but the large proportion of stands in this condition offers considerable opportunities. Our analysis indicates that, when seeking to increase forest carbon storage, managing stocking levels is an option with considerable potential, especially since no changes in land use are required.


Asunto(s)
Carbono/metabolismo , Ecosistema , Árboles/metabolismo , New England , New York , Pennsylvania
13.
J Am Soc Nephrol ; 21(12): 2117-29, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21088294

RESUMEN

SeSAME/EAST syndrome is a channelopathy consisting of a hypokalemic, hypomagnesemic, metabolic alkalosis associated with seizures, sensorineural deafness, ataxia, and developmental abnormalities. This disease links to autosomal recessive mutations in KCNJ10, which encodes the Kir4.1 potassium channel, but the functional consequences of these mutations are not well understood. In Xenopus oocytes, all of the disease-associated mutant channels (R65P, R65P/R199X, G77R, C140R, T164I, and A167V/R297C) had decreased K(+) current (0 to 23% of wild-type levels). Immunofluorescence demonstrated decreased surface expression of G77R, C140R, and A167V expressed in HEK293 cells. When we coexpressed mutant and wild-type subunits to mimic the heterozygous state, R199X, C140R, and G77R currents decreased to 55, 40, and 20% of wild-type levels, respectively, suggesting that carriers of these mutations may present with an abnormal phenotype. Because Kir4.1 subunits can form heteromeric channels with Kir5.1, we coexpressed the aforementioned mutants with Kir5.1 and found that currents were reduced at least as much as observed when we expressed mutants alone. Reduction of pH(i) from approximately 7.4 to 6.8 significantly decreased currents of all mutants except R199X but did not affect wild-type channels. In conclusion, perturbed pH gating may underlie the loss of channel function for the disease-associated mutant Kir4.1 channels and may have important physiologic consequences.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Pérdida Auditiva Sensorineural/genética , Canal de Potasio Kv.1.1/genética , Mutación , Convulsiones/genética , Alcalosis/genética , Alcalosis/fisiopatología , Análisis de Varianza , Animales , Ataxia/genética , Ataxia/fisiopatología , Técnica del Anticuerpo Fluorescente , Predisposición Genética a la Enfermedad , Células HEK293/metabolismo , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Hipopotasemia/genética , Hipopotasemia/fisiopatología , Immunoblotting , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Modelos Animales , Biología Molecular , Oocitos , Convulsiones/fisiopatología , Síndrome , Xenopus laevis
14.
Carbon Balance Manag ; 16(1): 17, 2021 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-34018077

RESUMEN

BACKGROUND: With the introduction of the Trillion Trees Initiative and similar programs, forests' ability to absorb carbon dioxide is increasingly in the spotlight. Many states have mandates to develop climate action plans, of which forest carbon is an important component, and planners need current information on forest carbon stocks and rates of change at relevant spatial scales. To this end, we examine rates of average annual change in live aboveground tree carbon in different forest type groups and provide state-wide and regional summaries of current live tree carbon stock and rates of change for the forests of the conterminous United States. Forest carbon summaries are presented in a format designed to meet the needs of managers, policymakers, and others requiring current estimates of aboveground live tree carbon at state and regional scales. RESULTS: Regional average aboveground live tree carbon stocks (represented on a per area basis) are generally between 40 and 75 tC/ha but range from 12.8 tC/ha in the Great Plains to 130 tC/ha in the Pacific Northwest West (west-side of Cascades). Regional average annual change in live aboveground tree carbon varies from a low of - 0.18 mtC/ha/y in the Rocky Mountain South to a high value of 1.74 mtC/ha/y in Pacific Northwest West. For individual states, carbon per unit area varies widely, from a low of 11.9 tC/ha in Nevada to a high of 96.4 tC/ha in Washington, with half the states falling between 50 and 75 tC/ha. Rates of average annual change in live aboveground tree carbon vary from a high of 1.82 tC/ha/y in Mississippi to a low of - 0.47 tC/ha/y in Colorado. CONCLUSIONS: Aboveground live tree carbon stocks and rates of average annual change vary by forest type within regions. While softwood forest types currently exhibit a higher rate of increase in the amount of carbon in aboveground live tree biomass, the current standing stock of carbon per unit area does not consistently follow this pattern. For this reason, we recommend computing and considering both measures -standing stock and average annual change-of carbon storage. The relative importance of each component will depend on management and policy objectives and the time frame related to those objectives. Harvesting and natural disturbance also affect forest carbon stock and change and may need to be considered if developing projections of potential carbon storage.

15.
Commun Biol ; 4(1): 1392, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907346

RESUMEN

Plasma membrane phosphatidylinositol 4-phosphate (PI4P) is a precursor of PI(4,5)P2, an important regulator of a large number of ion channels. Although the role of the phospholipid PI(4,5)P2 in stabilizing ion channel function is well established, little is known about the role of phospholipids in channel membrane localization and specifically the role of PI4P in channel function and localization. The phosphatidylinositol 4-kinases (PI4Ks) synthesize PI4P. Our data show that inhibition of PI4K and prolonged decrease of levels of plasma membrane PI4P lead to a decrease in the KCNQ1/KCNE1 channel membrane localization and function. In addition, we show that mutations linked to Long QT syndrome that affect channel interactions with phospholipids lead to a decrease in membrane expression. We show that expression of a LQT1-associated C-terminal deletion mutant abolishes PI4Kinase-mediated decrease in membrane expression and rescues membrane expression for phospholipid-targeting mutations. Our results indicate a novel role for PI4P on ion channel regulation. Our data suggest that decreased membrane PI4P availability to the channel, either due to inhibition of PI4K or as consequence of mutations, dramatically inhibits KCNQ1/KCNE1 channel membrane localization and current. Our results may have implications to regulation of other PI4P binding channels.


Asunto(s)
Membrana Celular/metabolismo , Canal de Potasio KCNQ1/genética , Fosfatos de Fosfatidilinositol/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Animales , Femenino , Canal de Potasio KCNQ1/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Ratas , Ratas Sprague-Dawley
16.
J Electrocardiol ; 43(5): 396-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20728018

RESUMEN

The short and long QT syndromes are inherited diseases associated with an increased risk for life-threatening arrhythmias. The first case of long QT syndrome (LQTS) was reported more than 150 years ago, and the study of this disease led to crucial advancement of our understanding of channelopathies and associated ventricular arrhythmias. Ten years ago, Gussak et al. reported four cases of idiopathic ventricular fibrillation in individuals from a family with a history of sudden cardiac death exhibited very short QT interval and labeled the disease: short QT syndrome (SQTS). Over this decade, the SQTS was found to be a rare inherited syndrome with the potential to provide novel insights into the main mechanisms of cardiac arrhythmogenicity. In this review, we discuss these mechanisms and provocatively question the role of the QT interval duration as a surrogate marker of increased risk for arrhythmia in both the LQTS and the SQTS.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Canalopatías/fisiopatología , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/genética , Canalopatías/complicaciones , Canalopatías/genética , Muerte Súbita Cardíaca/etiología , Electrocardiografía , Humanos , Síndrome de QT Prolongado/complicaciones , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Mutación , Canales de Potasio/genética , Factores de Riesgo
17.
PLoS One ; 15(8): e0237591, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32833978

RESUMEN

The slow cardiac delayed rectifier current (IKs) is formed by KCNQ1 and KCNE1 subunits and is one of the major repolarizing currents in the heart. Decrease of IKs currents either due to inherited mutations or pathological remodeling is associated with increased risk for cardiac arrhythmias and sudden death. Ca2+-dependent PKC isoforms (cPKC) are chronically activated in heart disease and diabetes. Recently, we found that sustained stimulation of the calcium-dependent PKCßII isoform leads to decrease in KCNQ1 subunit membrane localization and KCNQ1/KCNE1 channel activity, although the role of KCNE1 in this regulation was not explored. Here, we show that the auxiliary KCNE1 subunit expression is necessary for channel internalization. A mutation in a KCNE1 phosphorylation site (KCNE1(S102A)) abolished channel internalization in both heterologous expression systems and cardiomyocytes. Altogether, our results suggest that KCNE1(S102) phosphorylation by PKCßII leads to KCNQ1/KCNE1 channel internalization in response to sustained PKC stimulus, while leaving KCNQ1 homomeric channels in the membrane. This preferential internalization is expected to have strong impact on cardiac repolarization. Our results suggest that KCNE1(S102) is an important anti-arrhythmic drug target to prevent IKs pathological remodeling leading to cardiac arrhythmias.


Asunto(s)
Calcio/metabolismo , Canal de Potasio KCNQ1/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteína Quinasa C/metabolismo , Animales , Femenino , Células HEK293 , Humanos , Canal de Potasio KCNQ1/genética , Mutación , Miocitos Cardíacos/citología , Canales de Potasio con Entrada de Voltaje/genética , Proteína Quinasa C/genética , Ratas
18.
J Mol Cell Cardiol ; 46(5): 704-12, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19233191

RESUMEN

KCNQ1 is co-assembled with KCNE1 subunits in the heart to form the cardiac delayed rectifier K(+) current (IKs), which is one of the main currents responsible for myocyte repolarization. The most commonly inherited form of cardiac arrhythmias, long-QT syndrome type 1 (LQT1), is due to mutations on KCNQ1. Gq-coupled receptors (GqPCRs) are known to mediate positive inotropism in human ventricular myocardium. The mechanism of IKs current modulation by GqPCRs remains incompletely understood. Here we studied the molecular mechanisms underlying Gq regulation of the IKs channel. Heterologously expressed IKs (human KCNQ1/KCNE1 subunits) was measured in Xenopus oocytes, expressed together with GqPCRs. Our data from several GqPCRs shows that IKs is regulated in a biphasic manner, showing both an activation and an inhibition phase. Receptor-mediated inhibition phase was irreversible when recycling of agonist-sensitive pools of phosphatidylinositol-4,5-bisphosphate (PIP2) was blocked by the lipid kinase inhibitor wortmannin. In addition, stimulation of PIP(2) production, by overexpression of phosphatidylinositol-4-phosphate-5-kinase (PIP5-kinase), decreased receptor-mediated inhibition. The receptor-mediated activation phase was inhibited by the PKC inhibitor calphostin C and by a mutation in a putative PKC phosphorylation site in the KCNE1 subunit. Our results indicate that the depletion of membrane PIP(2) underlies receptor-mediated inhibition of IKs and that phosphorylation by PKC of the KCNE1 subunit underlies the GqPCR-mediated channel activation.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Activación del Canal Iónico , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio/metabolismo , Proteína Quinasa C/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Acetilcolina/farmacología , Animales , Calcio/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Hidrólisis/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Cinética , Modelos Biológicos , Fosforilación/efectos de los fármacos , Xenopus
19.
J Cardiovasc Electrophysiol ; 20(8): 859-65, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19490272

RESUMEN

BACKGROUND: Type-1 long-QT syndrome (LQT1) is caused by mutations in the KCNQ1 gene. The purpose of this study was to investigate whether KCNQ1 mutations in highly conserved amino acid residues within the voltage-gated potassium channel family are associated with an increased risk of cardiac events. METHODS AND RESULTS: The study population involved 492 LQT1 patients with 54 missense mutations in the transmembrane region of the KCNQ1 channel. The amino acid sequences of the transmembrane region of 38 human voltage-gated potassium channels were aligned. An adjusted Shannon entropy score for each amino acid residue was calculated ranging from 0 (no conservation) to 1.0 (full conservation). Cox analysis was used to identify independent factors associated with the first cardiac event (syncope, aborted cardiac arrest, or death). Patients were subcategorized into tertiles by their adjusted Shannon entropy scores. The lowest tertile (score 0-0.469; n = 146) was used as a reference group; patients with intermediate tertile scores (0.470-0.665; n = 150) had no increased risk of cardiac events (HR = 1.19, P = 0.42) or aborted cardiac arrest/sudden cardiac death (HR = 1.58, P = 0.26), and those with the highest tertile scores (>0.665; n = 196) showed significantly increased risk of cardiac events (HR = 3.32, P <0.001) and aborted cardiac arrest/sudden cardiac death (HR = 2.62, P = 0.04). The increased risk in patients with the highest conservation scores was independent of QTc, gender, age, and beta-blocker therapy. CONCLUSIONS: Mutations in highly conserved amino acid residues in the KCNQ1 gene are associated with a significant risk of cardiac events independent of QTc, gender, and beta-blocker therapy.


Asunto(s)
Secuencia Conservada , Canal de Potasio KCNQ1/genética , Mutación Missense/genética , Síndrome de Romano-Ward/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/genética , Niño , Preescolar , Biología Computacional/métodos , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Factores de Riesgo , Síndrome de Romano-Ward/etiología , Adulto Joven
20.
Nat Neurosci ; 8(5): 626-34, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15852009

RESUMEN

The subjective feeling of cold is mediated by the activation of TRPM8 channels in thermoreceptive neurons by cold or by cooling agents such as menthol. Here, we demonstrate a central role for phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)) in the activation of recombinant TRPM8 channels by both cold and menthol. Moreover, we show that Ca(2+) influx through these channels activates a Ca(2+)-sensitive phospholipase C and that the subsequent depletion of PI(4,5)P(2) limits channel activity, serving as a unique mechanism for desensitization of TRPM8 channels. Finally, we find that mutation of conserved positive residues in the highly conserved proximal C-terminal TRP domain of TRPM8 and two other family members, TRPM5 and TRPV5, reduces the sensitivity of the channels for PI(4,5)P(2) and increases inhibition by PI(4,5)P(2) depletion. These data suggest that the TRP domain of these channels may serve as a PI(4,5)P(2)-interacting site and that regulation by PI(4,5)P(2) is a common feature of members of the TRP channel family.


Asunto(s)
Frío , Canales Iónicos/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Células Receptoras Sensoriales/metabolismo , Sensación Térmica/fisiología , Animales , Células COS , Calcio/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Chlorocebus aethiops , Canales Iónicos/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mentol/farmacología , Mutación/fisiología , Proteínas de Neoplasias/genética , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Células Receptoras Sensoriales/efectos de los fármacos , Canales Catiónicos TRPV , Sensación Térmica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA