Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.484
Filtrar
1.
Acta Biomater ; 181: 391-401, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38704114

RESUMEN

Potassium ion transport across myocardial cell membrane is essential for type 2 long QT syndrome (LQT2). However, the dysfunction of potassium ion transport due to genetic mutations limits the therapeutic effect in treating LQT2. Biomimetic ion channels that selectively and efficiently transport potassium ions across the cellular membranes are promising for the treatment of LQT2. To corroborate this, we synthesized a series of foldamer-based ion channels with different side chains, and found a biomimetic ion channel of K+ (BICK) with the highest transport activity among them. The selected BICK can restore potassium ion transport and increase transmembrane potassium ion current, thus shortening phase 3 of action potential (AP) repolarization and QT interval in LQT2. Moreover, BICK does not affect heart rate and cardiac rhythm in treating LQT2 model induced by E4031 in isolated heart as well as in guinea pigs. By restoring ion transmembrane transport tactic, biomimetic ion channels, such as BICK, will show great potential in treating diseases related to ion transport blockade. STATEMENT OF SIGNIFICANCE: Type 2 long QT syndrome (LQT2) is a disease caused by K+ transport disorder, which can cause malignant arrhythmia and even death. There is currently no radical cure, so it is critical to explore ways to improve K+ transmembrane transport. In this study, we report that a small-molecule biomimetic ion channel BICK can efficiently simulate natural K+ channel proteins on the cardiomyocyte and cure E4031-induced LQT2 in guinea pig by restoring K+ transport function for the first time. This study found that the potassium transmembrane transport by BICK significantly reduced the QT interval, which provides a conceptually new strategy for the treatment of LQT2 disease.


Asunto(s)
Síndrome de QT Prolongado , Potasio , Síndrome de QT Prolongado/metabolismo , Animales , Potasio/metabolismo , Cobayas , Humanos , Potenciales de Acción/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Masculino , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Canales de Potasio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Frecuencia Cardíaca/efectos de los fármacos
2.
Biomed Pharmacother ; 175: 116649, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692059

RESUMEN

BACKGROUND: Second-generation antipsychotics increase the risk of atrial fibrillation. This study explores whether the atypical antipsychotic ziprasidone triggers inflammasome signaling, leading to atrial arrhythmia. METHODS: Electromechanical and pharmacological assessments were conducted on the rabbit left atria (LA). The patch-clamp technique was used to measure ionic channel currents in single cardiomyocytes. Detection of cytosolic reactive oxygen species production was performed in atrial cardiomyocytes. RESULTS: The duration of action potentials at 50 % and 90 % repolarization was dose-dependently shortened in ziprasidone-treated LA. Diastolic tension in LA increased after ziprasidone treatment. Ziprasidone-treated LA showed rapid atrial pacing (RAP) triggered activity. PI3K inhibitor, Akt inhibitor and mTOR inhibitor abolished the triggered activity elicited by ziprasidone in LA. The NLRP3 inhibitor MCC950 suppressed the ziprasidone-induced post-RAP-triggered activity. MCC950 treatment reduced the reverse-mode Na+/Ca2+ exchanger current in ziprasidone-treated myocytes. Cytosolic reactive oxygen species production decreased in ziprasidone-treated atrial myocytes after MCC950 treatment. Protein levels of inflammasomes and proinflammatory cytokines, including NLRP3, caspase-1, IL-1ß, IL-18, and IL-6 were observed to be upregulated in myocytes treated with ziprasidone. CONCLUSIONS: Our findings suggest ziprasidone induces atrial arrhythmia, potentially through upregulation of the NLRP3 inflammasome and enhancement of reactive oxygen species production via the PI3K/Akt/mTOR pathway.


Asunto(s)
Fibrilación Atrial , Inflamasomas , Miocitos Cardíacos , Piperazinas , Proteínas Proto-Oncogénicas c-akt , Especies Reactivas de Oxígeno , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Conejos , Especies Reactivas de Oxígeno/metabolismo , Piperazinas/farmacología , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Tiazoles/farmacología , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Potenciales de Acción/efectos de los fármacos , Antipsicóticos/farmacología
3.
Biointerphases ; 19(3)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38738941

RESUMEN

This paper introduces a physical neuron model that incorporates magnetoelectric nanoparticles (MENPs) as an essential electrical circuit component to wirelessly control local neural activity. Availability of such a model is important as MENPs, due to their magnetoelectric effect, can wirelessly and noninvasively modulate neural activity, which, in turn, has implications for both finding cures for neurological diseases and creating a wireless noninvasive high-resolution brain-machine interface. When placed on a neuronal membrane, MENPs act as magnetic-field-controlled finite-size electric dipoles that generate local electric fields across the membrane in response to magnetic fields, thus allowing to controllably activate local ion channels and locally initiate an action potential. Herein, the neuronal electrical characteristic description is based on ion channel activation and inhibition mechanisms. A MENP-based memristive Hodgkin-Huxley circuit model is extracted by combining the Hodgkin-Huxley model and an equivalent circuit model for a single MENP. In this model, each MENP becomes an integral part of the neuron, thus enabling wireless local control of the neuron's electric circuit itself. Furthermore, the model is expanded to include multiple MENPs to describe collective effects in neural systems.


Asunto(s)
Neuronas , Neuronas/fisiología , Neuronas/efectos de los fármacos , Nanopartículas/química , Humanos , Modelos Neurológicos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Campos Magnéticos
4.
Neuropharmacology ; 252: 109946, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38599494

RESUMEN

The spontaneous firing activity of nigral dopaminergic neurons is associated with some important roles including modulation of dopamine release, expression of tyrosine hydroxylase (TH), as well as neuronal survival. The decreased neuroactivity of nigral dopaminergic neurons has been revealed in Parkinson's disease. Central glucagon-like peptide-1 (GLP-1) functions as a neurotransmitter or neuromodulator to exert multiple brain functions. Although morphological studies revealed the expression of GLP-1 receptors (GLP-1Rs) in the substantia nigra pars compacta, the possible modulation of GLP-1 on spontaneous firing activity of nigral dopaminergic neurons is unknown. The present extracellular in vivo single unit recordings revealed that GLP-1R agonist exendin-4 significantly increased the spontaneous firing rate and decreased the firing regularity of partial nigral dopaminergic neurons of adult male C57BL/6 mice. Blockade of GLP-1Rs by exendin (9-39) decreased the firing rate of nigral dopaminergic neurons suggesting the involvement of endogenous GLP-1 in the modulation of firing activity. Furthermore, the PKA and the transient receptor potential canonical (TRPC) 4/5 channels are involved in activation of GLP-1Rs-induced excitatory effects of nigral dopaminergic neurons. Under parkinsonian state, both the exogenous and endogenous GLP-1 could still induce excitatory effects on the surviving nigral dopaminergic neurons. As the mild excitatory stimuli exert neuroprotective effects on nigral dopaminergic neurons, the present GLP-1-induced excitatory effects may partially contribute to its antiparkinsonian effects.


Asunto(s)
Potenciales de Acción , Neuronas Dopaminérgicas , Exenatida , Péptido 1 Similar al Glucagón , Receptor del Péptido 1 Similar al Glucagón , Ratones Endogámicos C57BL , Sustancia Negra , Animales , Masculino , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Exenatida/farmacología , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Ratones , Ponzoñas/farmacología , Péptidos/farmacología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/fisiopatología , Fragmentos de Péptidos/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo
5.
Toxicon ; 242: 107693, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38519012

RESUMEN

Aconitine is the main active component of Aconitum plants. Although aconitine has effects that include strengthening the heart, analgesia, anti-tumor, and immune-regulating effects, aconitine has both efficacy and toxicity, especially cardiotoxicity. Severe effects can include arrhythmia and cardiac arrest, which limits the clinical application of aconitine-containing traditional Chinese medicine. Ginsenoside Rb1(Rb1) is mainly found in plants, such as ginseng and Panax notoginseng, and has cardiovascular-protective and anti-arrhythmia effects. This study aimed to investigate the detoxifying effects of Rb1 on aconitine cardiotoxicity and the electrophysiological effect of Rb1 on aconitine-induced arrhythmia in rats. Pathological analysis, myocardial enzymatic indexes, and Western blotting were used to investigate the ameliorating effect of Rb1 on aconitine cardiotoxicity. Optical mapping was used to evaluate the effect of Rb1 on action potential and calcium signaling after aconitine-induced arrhythmia. Rb1 inhibited pathological damage caused by aconitine, decreased myocardial enzyme levels, and restored the balance of apoptotic protein expression by reducing the expression of Bax and cleaved caspase 3 and increasing the expression of Bcl-2, thereby reducing myocardial damage caused by aconitine. Rb1 also reduced the increase in heart rate caused by aconitine, accelerated action potential conduction and calcium signaling, and reduced the dispersion of action potential and calcium signal conduction. Rb1 reduced the cardiotoxicity of aconitine by attenuating aconitine-induced myocardial injury and inhibiting the aconitine-induced retardation of ventricular action potential and calcium signaling in rats.


Asunto(s)
Aconitina , Señalización del Calcio , Cardiotoxicidad , Ginsenósidos , Animales , Ginsenósidos/farmacología , Aconitina/análogos & derivados , Cardiotoxicidad/prevención & control , Ratas , Señalización del Calcio/efectos de los fármacos , Masculino , Potenciales de Acción/efectos de los fármacos , Ratas Sprague-Dawley , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/prevención & control , Miocardio/metabolismo , Miocardio/patología
6.
Am J Physiol Heart Circ Physiol ; 326(6): H1337-H1349, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38551482

RESUMEN

Nicotine is the primary addictive component of tobacco products. Through its actions on the heart and autonomic nervous system, nicotine exposure is associated with electrophysiological changes and increased arrhythmia susceptibility. To assess the underlying mechanisms, we treated rabbits with transdermal nicotine (NIC, 21 mg/day) or control (CT) patches for 28 days before performing dual optical mapping of transmembrane potential (RH237) and intracellular Ca2+ (Rhod-2 AM) in isolated hearts with intact sympathetic innervation. Sympathetic nerve stimulation (SNS) was performed at the first to third thoracic vertebrae, and ß-adrenergic responsiveness was additionally evaluated following norepinephrine (NE) perfusion. Baseline ex vivo heart rate (HR) and SNS stimulation threshold were higher in NIC versus CT (P = 0.004 and P = 0.003, respectively). Action potential duration alternans emerged at longer pacing cycle lengths (PCL) in NIC versus CT at baseline (P = 0.002) and during SNS (P = 0.0003), with similar results obtained for Ca2+ transient alternans. SNS shortened the PCL at which alternans emerged in CT but not in NIC hearts. NIC-exposed hearts tended to have slower and reduced HR responses to NE perfusion, but ventricular responses to NE were comparable between groups. Although fibrosis was unaltered, NIC hearts had lower sympathetic nerve density (P = 0.03) but no difference in NE content versus CT. These results suggest both sympathetic hypoinnervation of the myocardium and regional differences in ß-adrenergic responsiveness with NIC. This autonomic remodeling may contribute to the increased risk of arrhythmias associated with nicotine exposure, which may be further exacerbated with long-term use.NEW & NOTEWORTHY Here, we show that chronic nicotine exposure was associated with increased heart rate, increased susceptibility to alternans, and reduced sympathetic electrophysiological responses in the intact rabbit heart. We suggest that this was due to sympathetic hypoinnervation of the myocardium and diminished ß-adrenergic responsiveness of the sinoatrial node following nicotine treatment. Though these differences did not result in increased arrhythmia propensity in our study, we hypothesize that prolonged nicotine exposure may exacerbate this proarrhythmic remodeling.


Asunto(s)
Potenciales de Acción , Frecuencia Cardíaca , Corazón , Nicotina , Sistema Nervioso Simpático , Animales , Nicotina/toxicidad , Nicotina/efectos adversos , Conejos , Frecuencia Cardíaca/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Corazón/inervación , Corazón/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Masculino , Agonistas Nicotínicos/toxicidad , Agonistas Nicotínicos/administración & dosificación , Señalización del Calcio/efectos de los fármacos , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/metabolismo , Parche Transdérmico , Preparación de Corazón Aislado , Administración Cutánea , Norepinefrina/metabolismo
7.
Transl Res ; 268: 1-12, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38244770

RESUMEN

Interleukin (IL)-33, a cytokine involved in immune responses, can activate its receptor, suppression of tumorigenicity 2 (ST2), is elevated during atrial fibrillation (AF). However, the role of IL-33/ST2 signaling in atrial arrhythmia is unclear. This study explored the pathological effects of the IL-33/ST2 axis on atrial remodeling and arrhythmogenesis. Patch clamping, confocal microscopy, and Western blotting were used to analyze the electrical characteristics of and protein activity in atrial myocytes (HL-1) treated with recombinant IL-33 protein and/or ST2-neutralizing antibodies for 48 hrs. Telemetric electrocardiographic recordings, Masson's trichrome staining, and immunohistochemistry staining of the atrium were performed in mice receiving tail vein injections with nonspecific immunoglobulin (control), IL-33, and IL-33 combined with anti-ST2 antibody for 2 weeks. IL-33-treated HL-1 cells had a reduced action potential duration, lower L-type Ca2+ current, greater sarcoplasmic reticulum (SR) Ca2+ content, increased Na+/Ca2+ exchanger (NCX) current, elevation of K+ currents, and increased intracellular calcium transient. IL-33-treated HL-1 myocytes had greater activation of the calcium-calmodulin-dependent protein kinase II (CaMKII)/ryanodine receptor 2 (RyR2) axis and nuclear factor kappa B (NF-κB) / NLR family pyrin domain containing 3 (NLRP3) signaling than did control cells. IL-33 treated cells also had greater expression of Nav1.5, Kv1.5, NCX, and NLRP3 than did control cells. Pretreatment with neutralizing anti-ST2 antibody attenuated IL-33-mediated activation of CaMKII/RyR2 and NF-κB/NLRP3 signaling. IL-33-injected mice had more atrial ectopic beats and increased AF episodes, greater atrial fibrosis, and elevation of NF-κB/NLRP3 signaling than did controls or mice treated with IL-33 combined with anti-ST2 antibody. Thus, IL-33 recombinant protein treatment promotes atrial remodeling through ST2 signaling. Blocking the IL-33/ST2 axis might be an innovative therapeutic approach for patients with atrial arrhythmia and elevated serum IL-33.


Asunto(s)
Remodelación Atrial , Interleucina-33 , Miocitos Cardíacos , Animales , Masculino , Ratones , Potenciales de Acción/efectos de los fármacos , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/metabolismo , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/metabolismo , Remodelación Atrial/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular , Atrios Cardíacos/fisiopatología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal
8.
Stem Cell Reports ; 17(3): 522-537, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35180394

RESUMEN

Patients with coronavirus disease 2019 (COVID-19) commonly have manifestations of heart disease. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome encodes 27 proteins. Currently, SARS-CoV-2 gene-induced abnormalities of human heart muscle cells remain elusive. Here, we comprehensively characterized the detrimental effects of a SARS-CoV-2 gene, Orf9c, on human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) by preforming multi-omic analyses. Transcriptomic analyses of hPSC-CMs infected by SARS-CoV-2 with Orf9c overexpression (Orf9cOE) identified concordantly up-regulated genes enriched into stress-related apoptosis and inflammation signaling pathways, and down-regulated CM functional genes. Proteomic analysis revealed enhanced expressions of apoptotic factors, whereas reduced protein factors for ATP synthesis by Orf9cOE. Orf9cOE significantly reduced cellular ATP level, induced apoptosis, and caused electrical dysfunctions of hPSC-CMs. Finally, drugs approved by the U.S. Food and Drug Administration, namely, ivermectin and meclizine, restored ATP levels and ameliorated CM death and functional abnormalities of Orf9cOE hPSC-CMs. Overall, we defined the molecular mechanisms underlying the detrimental impacts of Orf9c on hPSC-CMs and explored potentially therapeutic approaches to ameliorate Orf9c-induced cardiac injury and abnormalities.


Asunto(s)
COVID-19/patología , Proteínas de la Nucleocápside de Coronavirus/genética , Estudio de Asociación del Genoma Completo/métodos , SARS-CoV-2/genética , Potenciales de Acción/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , COVID-19/virología , Regulación hacia Abajo , Humanos , Ivermectina/farmacología , Meclizina/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosfoproteínas/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Mapas de Interacción de Proteínas/genética , ARN Mensajero/química , ARN Mensajero/metabolismo , SARS-CoV-2/aislamiento & purificación , Transducción de Señal/genética , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba
9.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35074873

RESUMEN

The King Baboon spider, Pelinobius muticus, is a burrowing African tarantula. Its impressive size and appealing coloration are tempered by reports describing severe localized pain, swelling, itchiness, and muscle cramping after accidental envenomation. Hyperalgesia is the most prominent symptom after bites from P. muticus, but the molecular basis by which the venom induces pain is unknown. Proteotranscriptomic analysis of P. muticus venom uncovered a cysteine-rich peptide, δ/κ-theraphotoxin-Pm1a (δ/κ-TRTX-Pm1a), that elicited nocifensive behavior when injected into mice. In small dorsal root ganglion neurons, synthetic δ/κ-TRTX-Pm1a (sPm1a) induced hyperexcitability by enhancing tetrodotoxin-resistant sodium currents, impairing repolarization and lowering the threshold of action potential firing, consistent with the severe pain associated with envenomation. The molecular mechanism of nociceptor sensitization by sPm1a involves multimodal actions over several ion channel targets, including NaV1.8, KV2.1, and tetrodotoxin-sensitive NaV channels. The promiscuous targeting of peptides like δ/κ-TRTX-Pm1a may be an evolutionary adaptation in pain-inducing defensive venoms.


Asunto(s)
Nociceptores/efectos de los fármacos , Papio/metabolismo , Péptidos/farmacología , Venenos de Araña/farmacología , Arañas/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Ganglios Espinales/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Canales Iónicos/metabolismo , Ratones , Dolor/tratamiento farmacológico , Tetrodotoxina/farmacología
10.
Eur J Pharmacol ; 915: 174670, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34863995

RESUMEN

Hydroxychloroquine (HCQ) is a derivative of the antimalaria drug chloroquine primarily prescribed for autoimmune diseases. Recent attempts to repurpose HCQ in the treatment of corona virus disease 2019 has raised concerns because of its propensity to prolong the QT-segment on the electrocardiogram, an effect associated with increased pro-arrhythmic risk. Since chirality can affect drug pharmacological properties, we have evaluated the functional effects of the R(-) and S(+) enantiomers of HCQ on six ion channels contributing to the cardiac action potential and on electrophysiological parameters of isolated Purkinje fibers. We found that R(-)HCQ and S(+)HCQ block human Kir2.1 and hERG potassium channels in the 1 µM-100 µM range with a 2-4 fold enantiomeric separation. NaV1.5 sodium currents and CaV1.2 calcium currents, as well as KV4.3 and KV7.1 potassium currents remained unaffected at up to 90 µM. In rabbit Purkinje fibers, R(-)HCQ prominently depolarized the membrane resting potential, inducing autogenic activity at 10 µM and 30 µM, while S(+)HCQ primarily increased the action potential duration, inducing occasional early afterdepolarization at these concentrations. These data suggest that both enantiomers of HCQ can alter cardiac tissue electrophysiology at concentrations above their plasmatic levels at therapeutic doses, and that chirality does not substantially influence their arrhythmogenic potential in vitro.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Corazón/efectos de los fármacos , Hidroxicloroquina/química , Hidroxicloroquina/farmacología , Canales Iónicos/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/inducido químicamente , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Canales de Potasio Éter-A-Go-Go , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Ramos Subendocárdicos/efectos de los fármacos , Conejos , Estereoisomerismo
11.
Cardiovasc Res ; 118(1): 184-195, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33098411

RESUMEN

AIMS: Systemic inflammation and increased activity of atrial NOX2-containing NADPH oxidases have been associated with the new onset of atrial fibrillation (AF) after cardiac surgery. In addition to lowering LDL-cholesterol, statins exert rapid anti-inflammatory and antioxidant effects, the clinical significance of which remains controversial. METHODS AND RESULTS: We first assessed the impact of cardiac surgery and cardiopulmonary bypass (CPB) on atrial nitroso-redox balance by measuring NO synthase (NOS) and GTP cyclohydrolase-1 (GCH-1) activity, biopterin content, and superoxide production in paired samples of the right atrial appendage obtained before (PRE) and after CPB and reperfusion (POST) in 116 patients. The effect of perioperative treatment with atorvastatin (80 mg once daily) on these parameters, blood biomarkers, and the post-operative atrial effective refractory period (AERP) was then evaluated in a randomized, double-blind, placebo-controlled study in 80 patients undergoing cardiac surgery on CPB. CPB and reperfusion led to a significant increase in atrial superoxide production (74% CI 71-76%, n = 46 paired samples, P < 0.0001) and a reduction in atrial tetrahydrobiopterin (BH4) (34% CI 33-35%, n = 36 paired samples, P < 0.01), and in GCH-1 (56% CI 55-58%, n = 26 paired samples, P < 0.001) and NOS activity (58% CI 52-67%, n = 20 paired samples, P < 0.001). Perioperative atorvastatin treatment prevented the effect of CPB and reperfusion on all parameters but had no significant effect on the postoperative right AERP, troponin release, or NT-proBNP after cardiac surgery. CONCLUSION: Perioperative statin therapy prevents post-reperfusion atrial nitroso-redox imbalance in patients undergoing on-pump cardiac surgery but has no significant impact on postoperative atrial refractoriness, perioperative myocardial injury, or markers of postoperative LV function. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT01780740.


Asunto(s)
Atorvastatina/uso terapéutico , Fibrilación Atrial/prevención & control , Función del Atrio Derecho/efectos de los fármacos , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Puente Cardiopulmonar/efectos adversos , Atrios Cardíacos/efectos de los fármacos , Compuestos Nitrosos/metabolismo , Periodo Refractario Electrofisiológico/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Atorvastatina/efectos adversos , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Método Doble Ciego , Inglaterra , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Oxidación-Reducción , Superóxidos/metabolismo , Factores de Tiempo , Resultado del Tratamiento
12.
Cells ; 10(11)2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34831329

RESUMEN

Spontaneous AP (action potential) firing of sinoatrial nodal cells (SANC) is critically dependent on protein kinase A (PKA) and Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent protein phosphorylation, which are required for the generation of spontaneous, diastolic local Ca2+ releases (LCRs). Although phosphoprotein phosphatases (PP) regulate protein phosphorylation, the expression level of PPs and phosphatase inhibitors in SANC and the impact of phosphatase inhibition on the spontaneous LCRs and other players of the oscillatory coupled-clock system is unknown. Here, we show that rabbit SANC express both PP1, PP2A, and endogenous PP inhibitors I-1 (PPI-1), dopamine and cyclic adenosine 3',5'-monophosphate (cAMP)-regulated phosphoprotein (DARPP-32), kinase C-enhanced PP1 inhibitor (KEPI). Application of Calyculin A, (CyA), a PPs inhibitor, to intact, freshly isolated single SANC: (1) significantly increased phospholamban (PLB) phosphorylation (by 2-3-fold) at both CaMKII-dependent Thr17 and PKA-dependent Ser16 sites, in a time and concentration dependent manner; (2) increased ryanodine receptor (RyR) phosphorylation at the Ser2809 site; (3) substantially increased sarcoplasmic reticulum (SR) Ca2+ load; (4) augmented L-type Ca2+ current amplitude; (5) augmented LCR's characteristics and decreased LCR period in intact and permeabilized SANC, and (6) increased the spontaneous basal AP firing rate. In contrast, the selective PP2A inhibitor okadaic acid (100 nmol/L) had no significant effect on spontaneous AP firing, LCR parameters, or PLB phosphorylation. Application of purified PP1 to permeabilized SANC suppressed LCR, whereas purified PP2A had no effect on LCR characteristics. Our numerical model simulations demonstrated that PP inhibition increases AP firing rate via a coupled-clock mechanism, including respective increases in the SR Ca2+ pumping rate, L-type Ca2+ current, and Na+/Ca2+-exchanger current. Thus, PP1 and its endogenous inhibitors modulate the basal spontaneous firing rate of cardiac pacemaker cells by suppressing SR Ca2+ cycling protein phosphorylation, the SR Ca2+ load and LCRs, and L-type Ca2+ current.


Asunto(s)
Relojes Biológicos , Fosfoproteínas Fosfatasas/metabolismo , Nodo Sinoatrial/citología , Potenciales de Acción/efectos de los fármacos , Animales , Relojes Biológicos/efectos de los fármacos , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Proteínas de Unión al Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Simulación por Computador , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ventrículos Cardíacos/citología , Toxinas Marinas/farmacología , Modelos Biológicos , Oxazoles/farmacología , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos
13.
Int J Mol Sci ; 22(22)2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34830281

RESUMEN

Solifenacin (Vesicare®, SOL), known to be a member of isoquinolines, is a muscarinic antagonist that has anticholinergic effect, and it has been beneficial in treating urinary incontinence and neurogenic detrusor overactivity. However, the information regarding the effects of SOL on membrane ionic currents is largely uncertain, despite its clinically wide use in patients with those disorders. In this study, the whole-cell current recordings revealed that upon membrane depolarization in pituitary GH3 cells, the exposure to SOL concentration-dependently increased the amplitude of M-type K+ current (IK(M)) with effective EC50 value of 0.34 µM. The activation time constant of IK(M) was concurrently shortened in the SOL presence, hence yielding the KD value of 0.55 µM based on minimal reaction scheme. As cells were exposed to SOL, the steady-state activation curve of IK(M) was shifted along the voltage axis to the left with no change in the gating charge of the current. Upon an isosceles-triangular ramp pulse, the hysteretic area of IK(M) was increased by adding SOL. As cells were continually exposed to SOL, further application of acetylcholine (1 µM) failed to modify SOL-stimulated IK(M); however, subsequent addition of thyrotropin releasing hormone (TRH, 1 µM) was able to counteract SOL-induced increase in IK(M) amplitude. In cell-attached single-channel current recordings, bath addition of SOL led to an increase in the activity of M-type K+ (KM) channels with no change in the single channel conductance; the mean open time of the channel became lengthened. In whole-cell current-clamp recordings, the SOL application reduced the firing of action potentials (APs) in GH3 cells; however, either subsequent addition of TRH or linopirdine was able to reverse SOL-mediated decrease in AP firing. In hippocampal mHippoE-14 neurons, the IK(M) was also stimulated by adding SOL. Altogether, findings from this study disclosed for the first time the effectiveness of SOL in interacting with KM channels and hence in stimulating IK(M) in electrically excitable cells, and this noticeable action appears to be independent of its antagonistic activity on the canonical binding to muscarinic receptors expressed in GH3 or mHippoE-14 cells.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Antagonistas Muscarínicos/farmacología , Canales de Potasio con Entrada de Voltaje/metabolismo , Transducción de Señal/efectos de los fármacos , Succinato de Solifenacina/farmacología , Acetilcolina/farmacología , Animales , Línea Celular Tumoral , Hipocampo/citología , Indoles/farmacología , Transporte Iónico/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Técnicas de Placa-Clamp/métodos , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Piridinas/farmacología , Ratas , Hormona Liberadora de Tirotropina/farmacología
14.
Biomed Pharmacother ; 144: 112307, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34653762

RESUMEN

Combination therapy between paclitaxel (PTX) and doxorubicin (DXR) is applied as the first-line treatment of breast cancer. Co-administration of drugs at synergistic ratio for treatment is facilitated with the use of nanocarriers, such as liposomes. However, despite the high response rate of solid tumors to this combination, a synergism of cardiotoxicity may limit the use. Thus, the objective of this work was to investigate the toxicity of long-circulating and fusogenic liposomes co-encapsulating PTX and DXR at the synergistic molar ratio (1:10) (LCFL-PTX/DXR). For this, clinical chemistry, histopathological analysis and electrocardiographic exams were performed on female Balb/c mice that received a single intravenous dose of LCFL-PTX/DXR. The results of the study indicated that the LD50 dose range (lethal dose for 50% of animals) of the LCFL-PTX/DXR treatment (28.9-34.7 mg/kg) is much higher than that found for free PTX/DXR treatment (20.8-23.1 mg/kg). In addition, liposomes promoted cardiac protection by not raising CK-MB levels in animals, keeping cardiomyocytes without injury or electrocardiographic changes. After 14 days of treatment, free PTX/DXR caused prolongation of the QRS interval when compared to LCFL-PTX/DXR treatment at the same dose (37.0 ± 5.01 ms and 30.83 ± 2.62 ms, respectively, with p = 0.017). The survival rate of animals treated with LCFL-PTX/DXR was three times higher than that of those treated with free drugs. Thus, it was established that the toxicity of LCFL-PTX/DXR is reduced compared to the combination of free PTX/DXR and this platform has advantages for the clinical treatment of breast cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Doxorrubicina/toxicidad , Cardiopatías/inducido químicamente , Lípidos/química , Miocitos Cardíacos/efectos de los fármacos , Paclitaxel/toxicidad , Potenciales de Acción/efectos de los fármacos , Administración Intravenosa , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/química , Cardiotoxicidad , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Composición de Medicamentos , Sinergismo Farmacológico , Electrocardiografía , Femenino , Cardiopatías/metabolismo , Cardiopatías/patología , Dosificación Letal Mediana , Liposomas , Ratones Endogámicos BALB C , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Paclitaxel/administración & dosificación , Paclitaxel/química
15.
Bull Exp Biol Med ; 171(6): 732-735, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34705175

RESUMEN

In primary dissociated hippocampal cell cultures from 18-day-old mouse embryos, streptozotocin in concentrations of 2-5 mM produced a dose-dependent cytotoxic effect on day 3 in vitro, whereas on day 11 of culturing, the neurons were resistant to streptozotocin. The neurons in the 3-day cultures were functionally immature, which was seen from their weak spontaneous bioelectric activity in the form of rare single action potentials; by day 11 of culturing, the neurons reached a high level of differentiation and their functional properties acquired a character of network burst activity. Thus, streptozotocin had the most pronounced cytotoxic effect on immature hippocampal neurons in vitro.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Neuronas/efectos de los fármacos , Estreptozocina/toxicidad , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Ratones , Ratones Endogámicos BALB C , Neuronas/citología , Neuronas/fisiología , Cultivo Primario de Células , Factores de Tiempo
16.
Nat Commun ; 12(1): 6208, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34707084

RESUMEN

Inhibitory GABA-ergic neurotransmission is fundamental for the adult vertebrate central nervous system and requires low chloride concentration in neurons, maintained by KCC2, a neuroprotective ion transporter that extrudes intracellular neuronal chloride. To identify Kcc2 gene expression­enhancing compounds, we screened 1057 cell growth-regulating compounds in cultured primary cortical neurons. We identified kenpaullone (KP), which enhanced Kcc2/KCC2 expression and function in cultured rodent and human neurons by inhibiting GSK3ß. KP effectively reduced pathologic pain-like behavior in mouse models of nerve injury and bone cancer. In a nerve-injury pain model, KP restored Kcc2 expression and GABA-evoked chloride reversal potential in the spinal cord dorsal horn. Delta-catenin, a phosphorylation-target of GSK3ß in neurons, activated the Kcc2 promoter via KAISO transcription factor. Transient spinal over-expression of delta-catenin mimicked KP analgesia. Our findings of a newly repurposed compound and a novel, genetically-encoded mechanism that each enhance Kcc2 gene expression enable us to re-normalize disrupted inhibitory neurotransmission through genetic re-programming.


Asunto(s)
Analgésicos/uso terapéutico , Benzazepinas/uso terapéutico , Reposicionamiento de Medicamentos , Indoles/uso terapéutico , Transmisión Sináptica/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Analgésicos/farmacología , Animales , Benzazepinas/farmacología , Dolor en Cáncer/tratamiento farmacológico , Cateninas/genética , Cateninas/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Indoles/farmacología , Ratones , Neuralgia/tratamiento farmacológico , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo , Asta Dorsal de la Médula Espinal/patología , Simportadores/genética , Simportadores/metabolismo , Factores de Transcripción/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Catenina delta
17.
J Neurosci ; 41(43): 8859-8875, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34551939

RESUMEN

Neural response properties that typify primary sensory afferents are critical to fully appreciate because they establish and, ultimately represent, the fundamental coding design used for higher-level processing. Studies illuminating the center-surround receptive fields of retinal ganglion cells, for example, were ground-breaking because they determined the foundation of visual form detection. For the auditory system, a basic organizing principle of the spiral ganglion afferents is their extensive electrophysiological heterogeneity establishing diverse intrinsic firing properties in neurons throughout the spiral ganglion. Moreover, these neurons display an impressively large array of neurotransmitter receptor types that are responsive to efferent feedback. Thus, electrophysiological diversity and its neuromodulation are a fundamental encoding mechanism contributed by the primary afferents in the auditory system. To place these features into context, we evaluated the effects of hyperpolarization and cAMP on threshold level as indicators of overall afferent responsiveness in CBA/CaJ mice of either sex. Hyperpolarization modified threshold gradients such that distinct voltage protocols could shift the relationship between sensitivity and stimulus input to reshape resolution. This resulted in an "accordion effect" that appeared to stretch, compress, or maintain responsivity across the gradient of afferent thresholds. cAMP targeted threshold and kinetic shifts to rapidly adapting neurons, thus revealing multiple cochleotopic properties that could potentially be independently regulated. These examples of dynamic heterogeneity in primary auditory afferents not only have the capacity to shift the range, sensitivity, and resolution, but to do so in a coordinated manner that appears to orchestrate changes with a seemingly unlimited repertoire.SIGNIFICANCE STATEMENT How do we discriminate the more nuanced qualities of the sound around us? Beyond the basics of pitch and loudness, aspects, such as pattern, distance, velocity, and location, are all attributes that must be used to encode acoustic sensations effectively. While higher-level processing is required for perception, it would not be unexpected if the primary auditory afferents optimized receptor input to expedite neural encoding. The findings reported herein are consistent with this design. Neuromodulation compressed, expanded, shifted, or realigned intrinsic electrophysiological heterogeneity to alter neuronal responses selectively and dynamically. This suggests that diverse spiral ganglion phenotypes provide a rich substrate to support an almost limitless array of coding strategies within the first neural element of the auditory pathway.


Asunto(s)
Potenciales de Acción/fisiología , Ganglio Espiral de la Cóclea/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , AMP Cíclico/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos CBA , Técnicas de Cultivo de Órganos , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/efectos de los fármacos
18.
Neurobiol Dis ; 159: 105514, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34555537

RESUMEN

Synchronized and properly balanced electrical activity of neurons is the basis for the brain's ability to process information, to learn, and to remember. In Alzheimer's disease (AD), which causes cognitive decline in patients, this synchronization and balance is disturbed by the accumulation of neuropathological biomarkers such as amyloid-beta peptide (Aß42). Failure of Aß42 clearance mechanisms as well as desynchronization of crucial neuronal classes such as fast-spiking interneurons (FSN) are root causes for the disruption of the cognition-relevant gamma brain rhythm (30-80 Hz) and consequent cognitive impairment observed in AD. Here we show that recombinant BRICHOS molecular chaperone domains from ProSP-C or Bri2, which interfere with Aß42 aggregation, can rescue the gamma rhythm. We demonstrate that Aß42 progressively decreases gamma oscillation power and rhythmicity, disrupts the inhibition/excitation balance in pyramidal cells, and desynchronizes FSN firing during gamma oscillations in the hippocampal CA3 network of mice. Application of the more efficacious Bri2 BRICHOS chaperone rescued the cellular and neuronal network performance from all ongoing Aß42-induced functional impairments. Collectively, our findings offer critical missing data to explain the importance of FSN for normal network function and underscore the therapeutic potential of Bri2 BRICHOS to rescue the disruption of cognition-relevant brain rhythms in AD.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/farmacología , Hipocampo/efectos de los fármacos , Interneuronas/efectos de los fármacos , Chaperonas Moleculares/farmacología , Células Piramidales/efectos de los fármacos , Potenciales de Acción/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos beta-Amiloides , Animales , Modelos Animales de Enfermedad , Ritmo Gamma , Hipocampo/fisiopatología , Técnicas In Vitro , Interneuronas/fisiología , Ratones , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiopatología , Fragmentos de Péptidos , Dominios Proteicos , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Proteína C Asociada a Surfactante Pulmonar/farmacología , Células Piramidales/metabolismo , Células Piramidales/fisiología , Proteínas Recombinantes
19.
Eur J Pharmacol ; 910: 174441, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34474028

RESUMEN

Gefitinib, a tyrosine kinase inhibitor, was the first targeted therapy for non-small cell lung cancer (NSCLC). Gefitinib could block human Ether-à-go-go-Related Gene (hERG) channel, an important target in drug-induced long QT syndrome. However, it is unclear whether gefitinib could induce QT interval prolongation. Here, whole-cell patch-clamp technique was used for evaluating the effect of gefitinib on rapidly-activating delayed rectifier K+ current (IKr), slowly-activating delayed rectifier K+ current (IKs), transient outward potassium current (Ito), inward rectifier K+ current (IK1) and on action potentials in guinea pig ventricular myocytes. The Langendorff heart perfusion technique was used to determine drug effect on the ECG. Gefitinib depressed IKr by binding to open and closed hERG channels in a concentration-dependent way (IC50: 1.91 µM). The inhibitory effect of gefitinib on wildtype hERG channels was reduced at the hERG mutants Y652A, S636A, F656V and S631A (IC50: 8.51, 13.97, 18.86, 32.99 µM), indicating that gefitinib is a pore inhibitor of hERG channels. In addition, gefitinib accelerated hERG channel inactivation and decreased channel steady-state inactivation. Gefitinib also decreased IKs with IC50 of 23.8 µM. Moreover, gefitinib increased action potential duration (APD) in guinea pig ventricular myocytes and the corrected QT interval (QTc) in isolated perfused guinea pig hearts in a concentration-dependent way (1-30 µM). These findings indicate that gefitinib could prolong QTc interval by potently blocking hERG channel, modulating kinetic properties of hERG channel. Partial block of KCNQ1/KCNE1 could also contribute to delayed repolarization and prolonged QT interval. Thus, caution should be taken when gefitinib is used for NSCLC treatment.


Asunto(s)
Gefitinib/farmacología , Síndrome de QT Prolongado/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/metabolismo , Electrocardiografía/efectos de los fármacos , Cobayas , Células HEK293 , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Síndrome de QT Prolongado/inducido químicamente , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp
20.
J Mol Cell Cardiol ; 161: 86-97, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34375616

RESUMEN

Delayed rectifier K+ current (IKs) is a key contributor to repolarization of action potentials. This study investigated the mechanisms underlying the adrenoceptor-induced potentiation of IKs in pulmonary vein cardiomyocytes (PVC). PVC were isolated from guinea pig pulmonary vein. The action potentials and IKs current were recorded using perforated and conventional whole-cell patch-clamp techniques. The expression of IKs was examined using immunocytochemistry and Western blotting. KCNQ1, a IKs pore-forming protein was detected as a signal band approximately 100 kDa in size, and its immunofluorescence signal was found to be mainly localized on the cell membrane. The IKs current in PVC was markedly enhanced by both ß1- and ß2-adrenoceptor stimulation with a negative voltage shift in the current activation, although the potentiation was more effectively induced by ß2-adrenoceptor stimulation than ß1-adrenoceptor stimulation. Both ß-adrenoceptor-mediated increases in IKs were attenuated by treatment with the adenylyl cyclase (AC) inhibitor or protein kinase A (PKA) inhibitor. Furthermore, the IKs current was increased by α1-adrenoceptor agonist but attenuated by the protein kinase C (PKC) inhibitor. PVC exhibited action potentials in normal Tyrode solution which was slightly reduced by HMR-1556 a selective IKs blocker. However, HMR-1556 markedly reduced the ß-adrenoceptor-potentiated firing rate. The stimulatory effects of ß- and α1-adrenoceptor on IKs in PVC are mediated via the PKA and PKC signal pathways. HMR-1556 effectively reduced the firing rate under ß-adrenoceptor activation, suggesting that the functional role of IKs might increase during sympathetic excitation under in vivo conditions.


Asunto(s)
Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Miocitos Cardíacos/metabolismo , Venas Pulmonares/metabolismo , Receptores Adrenérgicos/metabolismo , Potenciales de Acción/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Cobayas , Atrios Cardíacos/metabolismo , Isoproterenol/farmacología , Canal de Potasio KCNQ1/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Proteína Quinasa C/metabolismo , Venas Pulmonares/citología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA