Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(15)2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37569465

RESUMEN

Long QT syndrome (LQTS) can lead to ventricular arrhythmia and sudden cardiac death. The most common congenital cause of LQTS is mutations in the channel subunits generating the cardiac potassium current IKs. Zebrafish (Danio rerio) have been proposed as a powerful system to model human cardiac diseases due to the similar electrical properties of the zebrafish heart and the human heart. We used high-resolution all-optical electrophysiology on ex vivo zebrafish hearts to assess the effects of IKs analogues on the cardiac action potential. We found that chromanol 293B (an IKs inhibitor) prolonged the action potential duration (APD) in the presence of E4031 (an IKr inhibitor applied to drug-induced LQT2), and to a lesser extent, in the absence of E4031. Moreover, we showed that PUFA analogues slightly shortened the APD of the zebrafish heart. However, PUFA analogues failed to reverse the APD prolongation in drug-induced LQT2. However, a more potent IKs activator, ML-277, partially reversed the APD prolongation in drug-induced LQT2 zebrafish hearts. Our results suggest that IKs plays a limited role in ventricular repolarizations in the zebrafish heart under resting conditions, although it plays a more important role when the IKr is compromised, as if the IKs in zebrafish serves as a repolarization reserve as in human hearts. This study shows that potent IKs activators can restore the action potential duration in drug-induced LQT2 in the zebrafish heart.


Asunto(s)
Síndrome de QT Prolongado , Canales de Potasio con Entrada de Voltaje , Animales , Humanos , Antiarrítmicos/farmacología , Pez Cebra , Corazón , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/genética , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/genética , Potenciales de Acción , Canales de Potasio con Entrada de Voltaje/farmacología
2.
Int J Mol Sci ; 22(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572906

RESUMEN

KV1.5 channel function is modified by different regulatory subunits. KVß1.3 subunits assemble with KV1.5 channels and induce a fast and incomplete inactivation. Inhibition of PKC abolishes the KVß1.3-induced fast inactivation, decreases the amplitude of the current KV1.5-KVß1.3 and modifies their pharmacology likely due to changes in the traffic of KV1.5-KVß1.3 channels in a PKC-dependent manner. In order to analyze this hypothesis, HEK293 cells were transfected with KV1.5-KVß1.3 channels, and currents were recorded by whole-cell configuration of the patch-clamp technique. The presence of KV1.5 in the membrane was analyzed by biotinylation techniques, live cell imaging and confocal microscopy approaches. PKC inhibition resulted in a decrease of 33 ± 7% of channels in the cell surface due to reduced recycling to the plasma membrane, as was confirmed by confocal microscopy. Live cell imaging indicated that PKC inhibition almost abolished the recycling of the KV1.5-KVß1.3 channels, generating an accumulation of channels into the cytoplasm. All these results suggest that the trafficking regulation of KV1.5-KVß1.3 channels is dependent on phosphorylation by PKC and, therefore, they could represent a clinically relevant issue, mainly in those diseases that exhibit modifications in PKC activity.


Asunto(s)
Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.5/metabolismo , Proteína Quinasa C/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Humanos , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores
3.
J Mol Cell Cardiol ; 110: 61-69, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28739325

RESUMEN

KV7.1 and KCNE1 co-assemble to give rise to the IKs current, one of the most important repolarizing currents of the cardiac action potential. Its relevance is underscored by the identification of >500 mutations in KV7.1 and, at least, 36 in KCNE1, that cause Long QT Syndrome (LQTS). The aim of this study was to characterize the biophysical and cellular consequences of the D242N KV7.1 mutation associated with the LQTS. The mutation is located in the S4 transmembrane segment, within the voltage sensor of the KV7.1 channel, disrupting the conserved charge balance of this region. Perforated patch-clamp experiments show that, unexpectedly, the mutation did not disrupt the voltage-dependent activation but it removed the inactivation and slowed the activation kinetics of D242N KV7.1 channels. Biotinylation of cell-surface protein and co-immunoprecipitation experiments revealed that neither plasma membrane targeting nor co-assembly between KV7.1 and KCNE1 was altered by the mutation. However, the association of D242N KV7.1 with KCNE1 strongly shifted the voltage dependence of activation to more depolarized potentials (+50mV), hindering IKs current at physiologically relevant membrane potentials. Both functional and computational analysis suggest that the clinical phenotype of the LQTS patients carrying the D242N mutation is due to impaired action potential adaptation to exercise and, in particular, to increase in heart rate. Moreover, our data identify D242 aminoacidic position as a potential residue involved in the KCNE1-mediated regulation of the voltage dependence of activation of the KV7.1 channel.


Asunto(s)
Aminoácidos/genética , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Mutación/genética , Potenciales de Acción , Adaptación Fisiológica , Secuencia de Aminoácidos , Electrocardiografía , Femenino , Células HEK293 , Células HeLa , Corazón/fisiopatología , Heterocigoto , Humanos , Canal de Potasio KCNQ1/química , Síndrome de QT Prolongado/diagnóstico por imagen , Síndrome de QT Prolongado/fisiopatología , Mutación con Pérdida de Función , Masculino , Transporte de Proteínas , Adulto Joven
4.
Arterioscler Thromb Vasc Biol ; 34(7): 1522-30, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24855057

RESUMEN

OBJECTIVE: Voltage-dependent K(+) (Kv) channels from the Kv7 family are expressed in blood vessels and contribute to cardiovascular physiology. Although Kv7 channel blockers trigger muscle contractions, Kv7 activators act as vasorelaxants. Kv7.1 and Kv7.5 are expressed in many vessels. Kv7.1 is under intense investigation because Kv7.1 blockers fail to modulate smooth muscle reactivity. In this study, we analyzed whether Kv7.1 and Kv7.5 may form functional heterotetrameric channels increasing the channel diversity in vascular smooth muscles. APPROACH AND RESULTS: Kv7.1 and Kv7.5 currents elicited in arterial myocytes, oocyte, and mammalian expression systems suggest the formation of heterotetrameric complexes. Kv7.1/Kv7.5 heteromers, exhibiting different pharmacological characteristics, participate in the arterial tone. Kv7.1/Kv7.5 associations were confirmed by coimmunoprecipitation, fluorescence resonance energy transfer, and fluorescence recovery after photobleaching experiments. Kv7.1/Kv7.5 heterotetramers were highly retained at the endoplasmic reticulum. Studies in HEK-293 cells, heart, brain, and smooth and skeletal muscles demonstrated that the predominant presence of Kv7.5 stimulates release of Kv7.1/Kv7.5 oligomers out of lipid raft microdomains. Electrophysiological studies supported that KCNE1 and KCNE3 regulatory subunits further increased the channel diversity. Finally, the analysis of rat isolated myocytes and human blood vessels demonstrated that Kv7.1 and Kv7.5 exhibited a differential expression, which may lead to channel diversity. CONCLUSIONS: Kv7.1 and Kv7.5 form heterotetrameric channels increasing the diversity of structures which fine-tune blood vessel reactivity. Because the lipid raft localization of ion channels is crucial for cardiovascular physiology, Kv7.1/Kv7.5 heteromers provide efficient spatial and temporal regulation of smooth muscle function. Our results shed light on the debate about the contribution of Kv7 channels to vasoconstriction and hypertension.


Asunto(s)
Canales de Potasio KCNQ/metabolismo , Canal de Potasio KCNQ1/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Potasio/metabolismo , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Canales de Potasio KCNQ/química , Canales de Potasio KCNQ/efectos de los fármacos , Canales de Potasio KCNQ/genética , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/efectos de los fármacos , Canal de Potasio KCNQ1/genética , Microdominios de Membrana/metabolismo , Potenciales de la Membrana , Músculo Liso Vascular/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Estructura Cuaternaria de Proteína , Ratas , Transfección , Xenopus
5.
J Immunol ; 191(12): 6136-46, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24249731

RESUMEN

Potassium channels modulate macrophage physiology. Blockade of voltage-dependent potassium channels (Kv) by specific antagonists decreases macrophage cytokine production and inhibits proliferation. In the presence of aspirin, acetylated cyclooxygenase-2 loses the activity required to synthesize PGs but maintains the oxygenase activity to produce 15R-HETE from arachidonate. This intermediate product is transformed via 5-LOX into epimeric lipoxins, termed 15-epi-lipoxins (15-epi-lipoxin A4 [e-LXA4]). Kv have been proposed as anti-inflammatory targets. Therefore, we studied the effects of e-LXA4 on signaling and on Kv and inward rectifier potassium channels (Kir) in mice bone marrow-derived macrophages (BMDM). Electrophysiological recordings were performed in these cells by the whole-cell patch-clamp technique. Treatment of BMDM with e-LXA4 inhibited LPS-dependent activation of NF-κB and IκB kinase ß activity, protected against LPS activation-dependent apoptosis, and enhanced the accumulation of the Nrf-2 transcription factor. Moreover, treatment of LPS-stimulated BMDM with e-LXA4 resulted in a rapid decrease of Kv currents, compatible with attenuation of the inflammatory response. Long-term treatment of LPS-stimulated BMDM with e-LXA4 significantly reverted LPS effects on Kv and Kir currents. Under these conditions, e-LXA4 decreased the calcium influx versus that observed in LPS-stimulated BMDM. These effects were partially mediated via the lipoxin receptor (ALX), because they were significantly reverted by a selective ALX receptor antagonist. We provide evidence for a new mechanism by which e-LXA4 contributes to inflammation resolution, consisting of the reversion of LPS effects on Kv and Kir currents in macrophages.


Asunto(s)
Inmunidad Innata/fisiología , Canal de Potasio Kv1.3/biosíntesis , Canal de Potasio Kv1.5/biosíntesis , Lipoxinas/farmacología , Activación de Macrófagos/fisiología , Canales de Potasio de Rectificación Interna/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Calcio/fisiología , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Interleucina-13/farmacología , Interleucina-4/farmacología , Transporte Iónico , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.5/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Potasio/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/fisiología , Venenos de Escorpión/farmacología , Organismos Libres de Patógenos Específicos , Regulación hacia Arriba
7.
Chirality ; 24(11): 944-50, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22674834

RESUMEN

Local anesthetics are useful probes of ion channel function and structure. Stereoselective interactions are especially interesting because they can reveal three-dimensional relationships between drugs and channels with otherwise identical biophysical and physicochemical properties. Furthermore, stereoselectivity suggests direct and specific receptor-mediated action, and identification of such stereospecific interactions may have important clinical consequences. The fact that drug targets are able to discriminate between the enantiomers present in a racemic drug is the consequence of the ordered asymmetric macromolecular units that form living cells. However, almost 25% of the drugs used in the clinical practice are racemic mixtures, and their individual enantiomers frequently differ in both their pharmacodynamic and pharmacokinetic profiles. Moreover, their effects can be similar to or different from the pharmacological effect of the drug and may contribute to the undesired effects of the drug. In other cases, the pharmacological effects induced by the two enantiomers on the molecular target are opposite. In the present manuscript, we will review the stereoselective effects of bupivacaine-like local anesthetics on cardiac sodium and potassium channels.


Asunto(s)
Anestésicos Locales/química , Anestésicos Locales/metabolismo , Canales Iónicos/metabolismo , Secuencia de Aminoácidos , Anestésicos Locales/farmacología , Humanos , Canales Iónicos/química , Unión Proteica , Estereoisomerismo , Especificidad por Sustrato
8.
Br J Pharmacol ; 178(15): 3034-3048, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33817777

RESUMEN

BACKGROUND AND PURPOSE: Local anaesthetics block sodium and a variety of potassium channels. Although previous studies identified a residue in the pore signature sequence together with three residues in the S6 segment as a putative binding site, the precise molecular basis of inhibition of Kv channels by local anaesthetics remained unknown. Crystal structures of Kv channels predict that some of these residues point away from the central cavity and face into a drug binding site called side pockets. Thus, the question arises whether the binding site of local anaesthetics is exclusively located in the central cavity or also involves the side pockets. EXPERIMENTAL APPROACH: A systematic functional alanine mutagenesis approach, scanning 58 mutants, together with in silico docking experiments and molecular dynamics simulations was utilized to elucidate the binding site of bupivacaine and ropivacaine. KEY RESULTS: Inhibition of Kv 1.5 channels by local anaesthetics requires binding to the central cavity and the side pockets, and the latter requires interactions with residues of the S5 and the back of the S6 segments. Mutations in the side pockets remove stereoselectivity of inhibition of Kv 1.5 channels by bupivacaine. Although binding to the side pockets is conserved for different local anaesthetics, the binding mode in the central cavity and the side pockets shows considerable variations. CONCLUSION AND IMPLICATIONS: Local anaesthetics bind to the central cavity and the side pockets, which provide a crucial key to the molecular understanding of their Kv channel affinity and stereoselectivity, as well as their spectrum of side effects.


Asunto(s)
Anestésicos Locales , Canales de Potasio/química , Anestésicos Locales/farmacología , Sitios de Unión , Bupivacaína/farmacología , Humanos , Simulación del Acoplamiento Molecular , Ropivacaína/farmacología
9.
Sci Adv ; 6(14): eaay4472, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32270035

RESUMEN

The potassium channel Kv7.1 associates with the KCNE1 regulatory subunit to trigger cardiac I Ks currents. Although the Kv7.1/KCNE1 complex has received much attention, the subcellular compartment hosting the assembly is the subject of ongoing debate. Evidence suggests that the complex forms either earlier in the endoplasmic reticulum or directly at the plasma membrane. Kv7.1 and KCNE1 mutations, responsible for long QT syndromes, impair association and traffic, thereby altering I Ks currents. We found that Kv7.1 and KCNE1 do not assemble in the first stages of their biogenesis. Data support an unconventional secretory pathway for Kv7.1-KCNE1 that bypasses Golgi. This route targets channels to endoplasmic reticulum-plasma membrane junctions, where Kv7.1-KCNE1 assemble. This mechanism helps to resolve the ongoing controversy about the subcellular compartment hosting the association. Our results also provide new insights into I Ks channel localization at endoplasmic reticulum-plasma membrane junctions, highlighting an alternative anterograde trafficking mechanism for oligomeric ion channels.


Asunto(s)
Canal de Potasio KCNQ1/metabolismo , Complejos Multiproteicos/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Transporte Biológico , Biomarcadores , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Humanos , Activación del Canal Iónico , Miocitos Cardíacos/metabolismo , Unión Proteica
10.
Elife ; 92020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32207683

RESUMEN

The cardiac ventricular action potential depends on several voltage-gated ion channels, including NaV, CaV, and KV channels. Mutations in these channels can cause Long QT Syndrome (LQTS) which increases the risk for ventricular fibrillation and sudden cardiac death. Polyunsaturated fatty acids (PUFAs) have emerged as potential therapeutics for LQTS because they are modulators of voltage-gated ion channels. Here we demonstrate that PUFA analogues vary in their selectivity for human voltage-gated ion channels involved in the ventricular action potential. The effects of specific PUFA analogues range from selective for a specific ion channel to broadly modulating cardiac ion channels from all three families (NaV, CaV, and KV). In addition, a PUFA analogue selective for the cardiac IKs channel (Kv7.1/KCNE1) is effective in shortening the cardiac action potential in human-induced pluripotent stem cell-derived cardiomyocytes. Our data suggest that PUFA analogues could potentially be developed as therapeutics for LQTS and cardiac arrhythmia.


Asunto(s)
Canales de Calcio Tipo L/efectos de los fármacos , Ácidos Grasos Insaturados/farmacología , Canal de Potasio KCNQ1/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Proteínas de Xenopus/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Antiarrítmicos/farmacología , Canales de Calcio Tipo L/fisiología , Células Madre Pluripotentes Inducidas/citología , Canal de Potasio KCNQ1/fisiología , Síndrome de QT Prolongado/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Proteínas de Xenopus/fisiología , Xenopus laevis
11.
Br J Pharmacol ; 176(13): 2131-2145, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30883701

RESUMEN

BACKGROUND AND PURPOSE: The NO/cGMP pathway represents a major physiological signalling controlling tone in pulmonary arteries (PA), and drugs activating this pathway are used to treat pulmonary arterial hypertension. Kv channels expressed in PA smooth muscle cells (PASMCs) are key determinants of vascular tone. We aimed to analyse the contribution of Kv 1.5 and Kv 7 channels in the electrophysiological and vasodilating effects evoked by NO donors and the GC stimulator riociguat in PA. EXPERIMENTAL APPROACH: Kv currents were recorded in isolated rat PASMCs using the patch-clamp technique. Vascular reactivity was assessed in a wire myograph. KEY RESULTS: The NO donors diethylamine NONOate diethylammonium (DEA-NO) and sodium nitroprusside hyperpolarized the membrane potential and induced a bimodal effect on Kv currents (augmenting the current between -40 and -10 mV and decreasing it at more depolarized potentials). The hyperpolarization and the enhancement of the current were suppressed by Kv 7 channel inhibitors and by the GC inhibitor ODQ but preserved when Kv 1.5 channels were inhibited. Additionally, DEA-NO enhanced Kv 7.5 currents in COS7 cells expressing the KCNQ5 gene. Riociguat increased Kv currents at all potentials ≥-40 mV and induced membrane hyperpolarization. Both effects were prevented by Kv 7 inhibition. Likewise, PA relaxation induced by NO donors and riociguat was attenuated by Kv 7 inhibitors. CONCLUSIONS AND IMPLICATIONS: NO donors and riociguat enhance Kv 7 currents, leading to PASMC hyperpolarization. This mechanism contributes to NO/cGMP-induced PA vasodilation. Our study identifies Kv 7 channels as a novel mechanism of action of vasodilator drugs used in the treatment of pulmonary arterial hypertension.


Asunto(s)
GMP Cíclico/fisiología , Canales de Potasio KCNQ/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Óxido Nítrico/fisiología , Arteria Pulmonar/fisiología , Animales , Células COS , Chlorocebus aethiops , Hidrazinas/farmacología , Canal de Potasio Kv1.5/fisiología , Masculino , Miocitos del Músculo Liso/fisiología , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Arteria Pulmonar/citología , Ratas Wistar , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
12.
Front Pharmacol ; 8: 177, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28408885

RESUMEN

Fludarabine (F-ara-A) is a purine analog commonly used in the treatment of indolent B cell malignancies that interferes with different aspects of DNA and RNA synthesis. KV1.3 K+ channels are membrane proteins involved in the maintenance of K+ homeostasis and the resting potential of the cell, thus controlling signaling events, proliferation and apoptosis in lymphocytes. Here we show that F-ara-A inhibits KV currents in human B lymphocytes. Our data indicate that KV1.3 is expressed in both BL2 and Dana B cell lines, although total KV1.3 levels were higher in BL2 than in Dana cells. However, KV currents in the plasma membrane were similar in both cell lines and were abrogated by the specific KV1.3 channel inhibitor PAP-1, indicating that KV1.3 accounts for most of the KV currents in these cell lines. F-ara-A, at a concentration (3.5 µM) similar to that achieved in the plasma of fludarabine phosphate-treated patients (3 µM), inhibited KV1.3 currents by 61 ± 6.3% and 52.3 ± 6.3% in BL2 and Dana B cells, respectively. The inhibitory effect of F-ara-A was concentration-dependent and showed an IC50 value of 0.36 ± 0.04 µM and a nH value of 1.07 ± 0.15 in BL2 cells and 0.34 ± 0.13 µM (IC50 ) and 0.77 ± 0.11 (nH ) in Dana cells. F-ara-A inhibition of plasma membrane KV1.3 was observed irrespective of its cytotoxic effect on the cells, BL2 cells being sensitive and Dana cells resistant to F-ara-A cytotoxicity. Interestingly, PAP-1, at concentrations as high as 10 µM, did not affect the viability of BL2 and Dana cells, indicating that blockage of KV1.3 in these cells is not toxic. Finally, F-ara-A had no effect on ectopically expressed KV1.3 channels, suggesting an indirect mechanism of current inhibition. In summary, our results describe the inhibitory effect of F-ara-A on the activity of KV1.3 channel. Although KV1.3 inhibition is not sufficient to induce cell death, further research is needed to determine whether it might still contribute to F-ara-A cytotoxicity in sensitive cells or be accountable for some of the clinical side effects of the drug.

13.
Front Physiol ; 7: 578, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27933000

RESUMEN

Voltage gated potassium channels (KV) are membrane proteins that allow selective flow of K+ ions in a voltage-dependent manner. These channels play an important role in several excitable cells as neurons, cardiomyocytes, and vascular smooth muscle. Over the last 20 years, it has been shown that omega-3 polyunsaturated fatty acids (PUFAs) enhance or decrease the activity of several cardiac KV channels. PUFAs-dependent modulation of potassium ion channels has been reported to be cardioprotective. However, the precise cellular mechanism underlying the cardiovascular benefits remained unclear in part because new PUFAs targets and signaling pathways continue being discovered. In this review, we will focus on recent data available concerning the following aspects of the KV channel modulation by PUFAs: (i) the exact residues involved in PUFAs-KV channels interaction; (ii) the structural PUFAs determinants important for their effects on KV channels; (iii) the mechanism of the gating modulation of KV channels and, finally, (iv) the PUFAs modulation of a few new targets present in smooth muscle cells (SMC), KCa1.1, K2P, and KATP channels, involved in vascular relaxation.

14.
J Clin Invest ; 126(2): 627-38, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26752648

RESUMEN

Deregulated protein and Ca2+ homeostasis underlie synaptic dysfunction and neurodegeneration in Huntington disease (HD); however, the factors that disrupt homeostasis are not fully understood. Here, we determined that expression of downstream regulatory element antagonist modulator (DREAM), a multifunctional Ca2+-binding protein, is reduced in murine in vivo and in vitro HD models and in HD patients. DREAM downregulation was observed early after birth and was associated with endogenous neuroprotection. In the R6/2 mouse HD model, induced DREAM haplodeficiency or blockade of DREAM activity by chronic administration of the drug repaglinide delayed onset of motor dysfunction, reduced striatal atrophy, and prolonged life span. DREAM-related neuroprotection was linked to an interaction between DREAM and the unfolded protein response (UPR) sensor activating transcription factor 6 (ATF6). Repaglinide blocked this interaction and enhanced ATF6 processing and nuclear accumulation of transcriptionally active ATF6, improving prosurvival UPR function in striatal neurons. Together, our results identify a role for DREAM silencing in the activation of ATF6 signaling, which promotes early neuroprotection in HD.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Cuerpo Estriado/metabolismo , Enfermedad de Huntington/metabolismo , Neuronas/metabolismo , Transducción de Señal , Factor de Transcripción Activador 6/genética , Animales , Células CHO , Carbamatos/farmacología , Cuerpo Estriado/patología , Cricetulus , Modelos Animales de Enfermedad , Células HEK293 , Células HeLa , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas de Interacción con los Canales Kv/genética , Proteínas de Interacción con los Canales Kv/metabolismo , Ratones , Neuronas/patología , Piperidinas/farmacología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
16.
Cardiovasc Res ; 105(2): 223-32, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25497550

RESUMEN

AIMS: Polyunsaturated fatty n-3 acids (PUFAs) have been reported to exhibit antiarrhythmic properties. However, the mechanisms of action remain unclear. We studied the electrophysiological effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on IKs, and on the expression and location of Kv7.1 and KCNE1. METHODS AND RESULTS: Experiments were performed using patch-clamp, western blot, and sucrose gradient techniques in COS7 cells transfected with Kv7.1/KCNE1 channels. Acute perfusion with both PUFAs increased Kv7.1/KCNE1 current, this effect being greater for DHA than for EPA. Similar results were found in guinea pig cardiomyocytes. Acute perfusion of either PUFA slowed the activation kinetics and EPA shifted the activation curve to the left. Conversely, chronic EPA did not modify Kv7.1/KCNE1 current magnitude and shifted the activation curve to the right. Chronic PUFAs decreased the expression of Kv7.1, but not of KCNE1, and induced spatial redistribution of Kv7.1 over the cell membrane. Cholesterol depletion with methyl-ß-cyclodextrin increased Kv7.1/KCNE1 current magnitude. Under these conditions, acute EPA produced similar effects than those induced in non-cholesterol-depleted cells. A ventricular action potential computational model suggested antiarrhythmic efficacy of acute PUFA application under IKr block. CONCLUSIONS: We provide evidence that acute application of PUFAs increases Kv7.1/KCNE1 through a probably direct effect, and shows antiarrhythmic efficacy under IKr block. Conversely, chronic EPA application modifies the channel activity through a change in the Kv7.1/KCNE1 voltage-dependence, correlated with a redistribution of Kv7.1 over the cell membrane. This loss of function may be pro-arrhythmic. This shed light on the controversial effects of PUFAs regarding arrhythmias.


Asunto(s)
Ácido Eicosapentaenoico/farmacología , Ácidos Grasos Insaturados/metabolismo , Activación del Canal Iónico , Microdominios de Membrana/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Antiarrítmicos/farmacología , Células COS , Chlorocebus aethiops , Ácidos Docosahexaenoicos/farmacología , Humanos , Activación del Canal Iónico/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo
17.
Cardiovasc Res ; 107(4): 613-23, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26168993

RESUMEN

AIMS: KCNQ1 and KCNE1 encode Kv7.1 and KCNE1, respectively, the pore-forming and the accessory subunits of the slow delayed rectifier potassium current, IKs. KCNQ1 mutations are associated with long and short QT syndrome. The aim of this study was to characterize the biophysical and cellular phenotype of a KCNQ1 missense mutation, F279I, found in a 23-year-old man with a corrected QT interval (QTc) of 356 ms and a family history of sudden cardiac death. METHODS AND RESULTS: Experiments were performed using perforated patch-clamp, western blot, co-immunoprecipitation, biotinylation, and immunocytochemistry techniques in HEK293, COS7 cells and in cardiomyocytes transfected with WT Kv7.1/KCNE1 or F279I Kv7.1/KCNE1 channels. In the absence of KCNE1, F279I Kv7.1 current exhibited a lesser degree of inactivation than WT Kv7.1. Also, functional analysis of F279I Kv7.1 in the presence of KCNE1 revealed a negative shift in the activation curve and an acceleration of the activation kinetics leading to a gain of function in IKs. The co-assembly between F279I Kv7.1 channels and KCNE1 was markedly decreased compared with WT Kv7.1 channels, as revealed by co-immunoprecipitation and Föster Resonance Energy Transfer experiments. All these effects contribute to the increase of IKs when channels incorporate F279I Kv7.1 subunits, as shown by a computer model simulation of these data that predicts a shortening of the action potential (AP) consistent with the patient phenotype. CONCLUSION: The F279I mutation induces a gain of function of IKs due to an impaired gating modulation of Kv7.1 induced by KCNE1, leading to a shortening of the cardiac AP.


Asunto(s)
Canal de Potasio KCNQ1/genética , Mutación , Miocitos Cardíacos/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Potenciales de Acción , Células HEK293 , Cardiopatías/genética , Humanos , Inmunoprecipitación/métodos , Mutación/genética
18.
Rev Med Inst Mex Seguro Soc ; 52(1): 76-83, 2014.
Artículo en Español | MEDLINE | ID: mdl-24625488

RESUMEN

OBJECTIVE: To calculate associated costs with dental studies (ACDS) in a public university. METHODS: We performed a cross-sectional study using a costing system on a random sample of 376 dental students enrolled at any semester in a public university. To calculate ACDS (Mexican pesos of 2009-1), we used a questionnaire divided into eight sections. Sociodemographic and socioeconomic variables, housing costs, food, transportation, instruments and equipment, as well as remunerations associated with patient care along 16 weeks of classes in each semester were included. We used linear regression. RESULTS: The average of ACDS was of 18,357.54 ± 12,746.81 Mexican pesos. The largest percentage of ACDS (30.2 %) was for clinical instruments (5,537.66 ± 6,260.50). Students also spent funds in paying to patients for their time during care delivered (2,402.11 ± 4,796.50). Associated variables (p 〈 0.001) with the ACDS were having completed at least one clinical course or one theoretical-practical course, living within the state or out of state (compared to students who live in the city where dental studies take place), and being enrolled in the more advanced dental studies. CONCLUSIONS: The results indicate that a significant percentage of the cost to students (13.1 %) is related with clinical care delivery.


OBJETIVO: calcular los costos relacionados con el estudio de la carrera de cirujano dentista (CRELCD) en una universidad pública del estado de Hidalgo, México. MÉTODOS: se empleó un sistema de costeo en una muestra aleatoria de alumnos de la carrera de cirujano dentista. Se incluyeron 376 estudiantes de segundo a décimo semestre. Para realizar el cálculo de los costos (pesos mexicanos para 2009) se utilizó un cuestionario. Se incluyeron variables sociodemográficas, socioeconómicas, sobre costos en vivienda, alimentación, transporte, instrumental y material, así como en atención a pacientes; para ello se consideraron 16 semanas de clases. Para el modelo final se empleó regresión lineal. RESULTADOS: el promedio del CRELCD fue 18 357.54 ± 12 746.81 pesos; 30.2 % consistió en el costo del instrumental (5537.66 ± 6260.50). Los alumnos gastaron en la remuneración a sus pacientes 2402.11 ± 4796.50 pesos. Las variables relacionadas fueron haber cursado al menos una asignatura clínica y una teórico-práctica, vivir en el interior o fuera del estado (en comparación quienes vivían en la ciudad donde cursaban la carrera) y ser alumno de semestres más avanzados. CONCLUSIONES: un porcentaje importante (13.1 %) de alumnos gasta en el tratamiento de sus pacientes. Es necesario identificar si estos costos representan barreras inequitativas en la decisión de emprender, continuar o finalizar estudios de odontología.


Asunto(s)
Educación en Odontología/economía , Facultades de Odontología/economía , Adolescente , Adulto , Estudios Transversales , Femenino , Humanos , Masculino , México , Facultades de Odontología/organización & administración , Adulto Joven
19.
Front Physiol ; 3: 245, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22934003

RESUMEN

Dietary n-3 polyunsaturated fatty acids (PUFAs) have been reported to exhibit antiarrhythmic properties, and these effects have been attributed to their capability to modulate ion channels. In the present review, we will focus on the effects of PUFAs on a cardiac sodium channel (Na(v)1.5) and two potassium channels involved in cardiac atrial and ventricular repolarization (K(v)) (K(v)1.5 and K(v)11.1). n-3 PUFAs of marine (docosahexaenoic, DHA and eicosapentaenoic acid, EPA) and plant origin (alpha-linolenic acid, ALA) block K(v)1.5 and K(v)11.1 channels at physiological concentrations. Moreover, DHA and EPA decrease the expression levels of K(v)1.5, whereas ALA does not. DHA and EPA also decrease the magnitude of the currents elicited by the activation of Na(v)1.5 and calcium channels. These effects on sodium and calcium channels should theoretically shorten the cardiac action potential duration (APD), whereas the blocking actions of n-3 PUFAs on K(v) channels would be expected to produce a lengthening of cardiac action potential. Indeed, the effects of n-3 PUFAs on the cardiac APD and, therefore, on cardiac arrhythmias vary depending on the method of application, the animal model, and the underlying cardiac pathology.

20.
Front Pharmacol ; 3: 163, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22973228

RESUMEN

Polyunsaturated fatty acids (PUFAs) have been reported to exhibit antiarrhythmic properties, which are attributed to their capability to modulate ion channels. This PUFAs ability has been reported to be due to their effects on the gating properties of ion channels. In the present review, we will focus on the role of PUFAs on the gating of two Kv channels, Kv1.5 and Kv11.1. Kv1.5 channels are blocked by n-3 PUFAs of marine [docosahexaenoic acid (DHA) and eicosapentaenoic acid] and plant origin (alpha-linolenic acid, ALA) at physiological concentrations. The blockade of Kv1.5 channels by PUFAs steeply increased in the range of membrane potentials coinciding with those of Kv1.5 channel activation, suggesting that PUFAs-channel binding may derive a significant fraction of its voltage sensitivity through the coupling to channel gating. A similar shift in the activation voltage was noted for the effects of n-6 arachidonic acid (AA) and DHA on Kv1.1, Kv1.2, and Kv11.1 channels. PUFAs-Kv1.5 channel interaction is time-dependent, producing a fast decay of the current upon depolarization. Thus, Kv1.5 channel opening is a prerequisite for the PUFA-channel interaction. Similar to the Kv1.5 channels, the blockade of Kv11.1 channels by AA and DHA steeply increased in the range of membrane potentials that coincided with the range of Kv11.1 channel activation, suggesting that the PUFAs-Kv channel interactions are also coupled to channel gating. Furthermore, AA regulates the inactivation process in other Kv channels, introducing a fast voltage-dependent inactivation in non-inactivating Kv channels. These results have been explained within the framework that AA closes voltage-dependent potassium channels by inducing conformational changes in the selectivity filter, suggesting that Kv channel gating is lipid dependent.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA