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

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Intervalo de año de publicación
1.
Clin Transl Med ; 12(7): e954, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35872650

RESUMEN

BACKGROUND: Mice with deletion of complex I subunit Ndufs4 develop mitochondrial encephalomyopathy resembling Leigh syndrome (LS). The metabolic derangement and underlying mechanisms of cardio-encephalomyopathy in LS remains incompletely understood. METHODS: We performed echocardiography, electrophysiology, confocal microscopy, metabolic and molecular/morphometric analysis of the mice lacking Ndufs4. HEK293 cells, human iPS cells-derived cardiomyocytes and neurons were used to determine the mechanistic role of mitochondrial complex I deficiency. RESULTS: LS mice develop severe cardiac bradyarrhythmia and diastolic dysfunction. Human-induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) with Ndufs4 deletion recapitulate LS cardiomyopathy. Mechanistically, we demonstrate a direct link between complex I deficiency, decreased intracellular (nicotinamide adenine dinucleotide) NAD+ /NADH and bradyarrhythmia, mediated by hyperacetylation of the cardiac sodium channel NaV 1.5, particularly at K1479 site. Neuronal apoptosis in the cerebellar and midbrain regions in LS mice was associated with hyperacetylation of p53 and activation of microglia. Targeted metabolomics revealed increases in several amino acids and citric acid cycle intermediates, likely due to impairment of NAD+ -dependent dehydrogenases, and a substantial decrease in reduced Glutathione (GSH). Metabolic rescue by nicotinamide riboside (NR) supplementation increased intracellular NAD+ / NADH, restored metabolic derangement, reversed protein hyperacetylation through NAD+ -dependent Sirtuin deacetylase, and ameliorated cardiomyopathic phenotypes, concomitant with improvement of NaV 1.5 current and SERCA2a function measured by Ca2+ -transients. NR also attenuated neuronal apoptosis and microglial activation in the LS brain and human iPS-derived neurons with Ndufs4 deletion. CONCLUSIONS: Our study reveals direct mechanistic explanations of the observed cardiac bradyarrhythmia, diastolic dysfunction and neuronal apoptosis in mouse and human induced pluripotent stem cells (iPSC) models of LS.


Asunto(s)
Cardiomiopatías , Células Madre Pluripotentes Inducidas , Enfermedad de Leigh , Animales , Bradicardia/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Leigh/genética , Enfermedad de Leigh/metabolismo , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales , NAD/metabolismo
2.
BMC Complement Altern Med ; 19(1): 357, 2019 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822281

RESUMEN

BACKGROUND: Shenxian-Shengmai (SXSM) Oral Liquid is a CFDA-approved patent Chinese Herbal medicine, which has been clinically used for the treatment of bradycardia. However, its active components and action mechanism remain to be established. The present study aimed to evaluate the efficacy of SXSM on bradycardia and to identify the possible active components and their pharmacological targets for this action. METHODS: A literature-based meta-analysis was performed to evaluate the clinical efficacy of SXSM on bradycardia, which was confirmed by a rat ex vivo cardiac model. Network pharmacology analysis was then conducted to reveal the potential targets of SXSM active components and their anti-arrhythmia mechanisms. Finally, the identified drug-target interaction was confirmed by immunofluorescence assay in cardiomyocyte. RESULTS: Meta-analysis of the available clinical study data shows that Shenxian-Shengmai Oral Liquid has a favorable effect for bradycardia. In an ex vivo bradycardia model of rat heart, SXSM restored heart rate by affecting Heart rate variability (HRV) which is associated with autonomic nervous system activity. A drug-target-pathway network analysis connecting SXSM components with arrhythmia suggested that a prominent anti-arrhythmia mechanisms of SXSM was via ß1-adrenergic signaling pathway, which was subsequently validated by immunofluorescence assay showing that SXSM indeed increased the expression of ADRB1 in cultured cardiomyocytes. CONCLUSION: By combining approaches of clinical evidence mining, experimental model confirmation, network pharmacology analyses and molecular mechanistic validation, we show that SXSM is an effective treatment for bradycardia and it involves multiple component interacting via multiple pathways, among which is the critical ß1-adrenergic receptor upregulation. Our integrative approach could be applied to other multi-component traditional Chinese medicine investigation where ample clinical data are accumulated but advanced mechanistic studies are lacking.


Asunto(s)
Bradicardia/metabolismo , Medicamentos Herbarios Chinos/farmacología , Receptores Adrenérgicos beta 1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Línea Celular , Electrocardiografía , Corazón/efectos de los fármacos , Masculino , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Circulation ; 139(18): 2157-2169, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30764634

RESUMEN

BACKGROUND: Bradyarrhythmia is a common clinical manifestation. Although the majority of cases are acquired, genetic analysis of families with bradyarrhythmia has identified a growing number of causative gene mutations. Because the only ultimate treatment for symptomatic bradyarrhythmia has been invasive surgical implantation of a pacemaker, the discovery of novel therapeutic molecular targets is necessary to improve prognosis and quality of life. METHODS: We investigated a family containing 7 individuals with autosomal dominant bradyarrhythmias of sinus node dysfunction, atrial fibrillation with slow ventricular response, and atrioventricular block. To identify the causative mutation, we conducted the family-based whole exome sequencing and genome-wide linkage analysis. We characterized the mutation-related mechanisms based on the pathophysiology in vitro. After generating a transgenic animal model to confirm the human phenotypes of bradyarrhythmia, we also evaluated the efficacy of a newly identified molecular-targeted compound to upregulate heart rate in bradyarrhythmias by using the animal model. RESULTS: We identified one heterozygous mutation, KCNJ3 c.247A>C, p.N83H, as a novel cause of hereditary bradyarrhythmias in this family. KCNJ3 encodes the inwardly rectifying potassium channel Kir3.1, which combines with Kir3.4 (encoded by KCNJ5) to form the acetylcholine-activated potassium channel ( IKACh channel) with specific expression in the atrium. An additional study using a genome cohort of 2185 patients with sporadic atrial fibrillation revealed another 5 rare mutations in KCNJ3 and KCNJ5, suggesting the relevance of both genes to these arrhythmias. Cellular electrophysiological studies revealed that the KCNJ3 p.N83H mutation caused a gain of IKACh channel function by increasing the basal current, even in the absence of m2 muscarinic receptor stimulation. We generated transgenic zebrafish expressing mutant human KCNJ3 in the atrium specifically. It is interesting to note that the selective IKACh channel blocker NIP-151 repressed the increased current and improved bradyarrhythmia phenotypes in the mutant zebrafish. CONCLUSIONS: The IKACh channel is associated with the pathophysiology of bradyarrhythmia and atrial fibrillation, and the mutant IKACh channel ( KCNJ3 p.N83H) can be effectively inhibited by NIP-151, a selective IKACh channel blocker. Thus, the IKACh channel might be considered to be a suitable pharmacological target for patients who have bradyarrhythmia with a gain-of-function mutation in the IKACh channel.


Asunto(s)
Fibrilación Atrial , Bloqueo Atrioventricular , Bradicardia , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Enfermedades Genéticas Congénitas , Mutación Missense , Sustitución de Aminoácidos , Animales , Animales Modificados Genéticamente , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Bloqueo Atrioventricular/genética , Bloqueo Atrioventricular/metabolismo , Bloqueo Atrioventricular/patología , Bloqueo Atrioventricular/fisiopatología , Benzopiranos/farmacología , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/patología , Bradicardia/fisiopatología , Técnicas Electrofisiológicas Cardíacas , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Enfermedades Genéticas Congénitas/fisiopatología , Humanos , Masculino , Xenopus laevis , Pez Cebra
4.
Exp Physiol ; 103(10): 1302-1308, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30070742

RESUMEN

NEW FINDINGS: What is the central question of this study? How do gastric stretch and gastric cooling stimuli affect cardiac autonomic control? What is the main finding and its importance? Gastric stretch causes an increase in cardiac sympathetic activity. Stretch combined with cold stimulation result in an elimination of the sympathetic response to stretch and an increase in cardiac parasympathetic activity, in turn resulting in a reduction in heart rate. Gastric cold stimulation causes a shift in sympathovagal balance towards parasympathetic dominance. The cold-induced bradycardia has the potential to decrease cardiac workload, which might be significant in individuals with cardiovascular pathologies. ABSTRACT: Gastric distension increases blood pressure and heart rate in young, healthy humans, but little is known about the effect of gastric stretch combined with cooling. We used a randomized crossover study to assess the cardiovascular responses to drinking 300 ml of ispaghula husk solution at either 6 or 37°C in nine healthy humans (age 24.08 ± 9.36 years) to establish the effect of gastric stretch with and without cooling. The effect of consuming peppermint oil capsules to activate cold thermoreceptors was also investigated. The ECG, respiratory movements and continuous blood pressure were recorded during a 5 min baseline period, followed by a 115 min post-drink period, during which 5 min epochs of data were recorded. Cardiac autonomic activity was assessed using time and frequency domain analyses of respiratory sinus arrhythmia to quantify parasympathetic autonomic activity, and corrected QT (QTc) interval analysis to quantify sympathetic autonomic activity. Gastric stretch only caused a significant reduction in QTc interval lasting up to 15 min, with a concomitant but non-significant increase in heart rate, indicating an increased sympathetic cardiac tone. The additional effect of gastric cold stimulation was significantly to reduce heart rate for up to 15 min, elevate indicators of cardiac parasympathetic tone and eliminate the reduction in QTc interval seen with gastric stretch only. Stimulation of gastric cold thermoreceptors with menthol also caused a significant reduction in heart rate and concomitant increase in the root mean square of successive differences. These findings indicate that gastric cold stimulation causes a shift in the sympathovagal balance of cardiac control towards a more parasympathetic dominant pattern.


Asunto(s)
Frecuencia Cardíaca/efectos de los fármacos , Corazón/efectos de los fármacos , Mentol/administración & dosificación , Adulto , Sistema Nervioso Autónomo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Bradicardia/metabolismo , Frío , Estudios Cruzados , Electrocardiografía/efectos de los fármacos , Voluntarios Sanos , Humanos , Mentha piperita , Aceites de Plantas/administración & dosificación , Psyllium/administración & dosificación , Termorreceptores/metabolismo , Adulto Joven
5.
Int Heart J ; 59(3): 601-606, 2018 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-29628472

RESUMEN

A biological pacemaker is expected to solve the persisting problems of an artificial cardiac pacemaker including short battery life, lead breaks, infection, and electromagnetic interference. We previously reported HCN4 overexpression enhances pacemaking ability of mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) in vitro. However, the effect of these cells on bradycardia in vivo has remained unclear. Therefore, we transplanted HCN4-overexpressing mESC-CMs into bradycardia model animals and investigated whether they could function as a biological pacemaker. The rabbit Hcn4 gene was transfected into mouse embryonic stem cells and induced HCN4-overexpressing mESC-CMs. Non-cardiomyocytes were removed under serum/glucose-free and lactate-supplemented conditions. Cardiac balls containing 5 × 103 mESC-CMs were made by using the hanging drop method. One hundred cardiac balls were injected into the left ventricular free wall of complete atrioventricular block (CAVB) model rats. Heart beats were evaluated using an implantable telemetry system 7 to 30 days after cell transplantation. The result showed that ectopic ventricular beats that were faster than the intrinsic escape rhythm were often observed in CAVB model rats transplanted with HCN4-overexpressing mESC-CMs. On the other hand, the rats transplanted with non-overexpressing mESC-CMs showed sporadic single premature ventricular contraction but not sustained ectopic ventricular rhythms. These results indicated that HCN4-overexpressing mESC-CMs produce rapid ectopic ventricular rhythms as a biological pacemaker.


Asunto(s)
Relojes Biológicos/fisiología , Bradicardia/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Bradicardia/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Frecuencia Cardíaca/fisiología , Ratones , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/citología , Conejos , Ratas , Telemetría , Transfección
6.
Sci Rep ; 8(1): 1900, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382866

RESUMEN

Stimulation of vagal afferent endings with intravenous phenylbiguanide (PBG) causes both bradycardia and vasodepression, simulating neurally mediated syncope. Activation of µ-opioid receptors in the nucleus tractus solitarius (NTS) increases blood pressure. Electroacupuncture (EA) stimulation of somatosensory nerves underneath acupoints P5-6, ST36-37, LI6-7 or G37-39 selectively but differentially modulates sympathoexcitatory responses. We therefore hypothesized that EA-stimulation at P5-6 or ST36-37, but not LI6-7 or G37-39 acupoints, inhibits the bradycardia and vasodepression through a µ-opioid receptor mechanism in the NTS. We observed that stimulation at acupoints P5-6 and ST36-37 overlying the deep somatosensory nerves and LI6-7 and G37-39 overlying cutaneous nerves differentially evoked NTS neural activity in anesthetized and ventilated animals. Thirty-min of EA-stimulation at P5-6 or ST36-37 reduced the depressor and bradycardia responses to PBG while EA at LI6-7 or G37-39 did not. Congruent with the hemodynamic responses, EA at P5-6 and ST36-37, but not at LI6-7 and G37-39, reduced vagally evoked activity of cardiovascular NTS cells. Finally, opioid receptor blockade in the NTS with naloxone or a specific µ-receptor antagonist reversed P5-6 EA-inhibition of the depressor, bradycardia and vagally evoked NTS activity. These data suggest that point specific EA stimulation inhibits PBG-induced vasodepression and bradycardia responses through a µ-opioid mechanism in the NTS.


Asunto(s)
Analgésicos Opioides/farmacología , Bradicardia/tratamiento farmacológico , Bradicardia/fisiopatología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/fisiopatología , Vasodilatadores/farmacología , Puntos de Acupuntura , Animales , Presión Sanguínea/efectos de los fármacos , Bradicardia/metabolismo , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Gatos , Electroacupuntura/métodos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Masculino , Naloxona/farmacología , Receptores Opioides/metabolismo , Reflejo/efectos de los fármacos , Núcleo Solitario/metabolismo , Nervio Vago/efectos de los fármacos , Nervio Vago/metabolismo , Nervio Vago/fisiopatología
7.
J Neuroimmune Pharmacol ; 12(1): 204-209, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28150133

RESUMEN

Fingolimod is a an oral disease modifying drug for relapsing remitting multiple sclerosis (MS) preventing egress of B and T cells from lymph nodes. Relevant first dose adverse events include bradycardia and atrioventricular conduction slowing. Cardiac side effects of fingolimod and combinational pharmacotherapy including duloxetine and tolterodine were monitored in mice of different age using implantable ECG telemetric systems. Cardiac tissue was assessed for S1P-receptor subtype (1 and 3), and for GIRK1 expression. Fingolimod led to a significant heart rate reduction within 60 min, which returned to baseline values within 24 h. In older mice bradycardia was more pronounced compared to younger mice. Atrioventricular conduction was not affected. Older mice showed a higher S1PR3 expression in a naïve state and receptor expression was reduced after fingolimod administration. Combination with duloxetine or tolterodine alleviated fingolimod induced heart rate decrease. Our data provide preclinical evidence that negative chronotropic effects of fingolimod might be age dependent, possibly due to an altered expression and internalization of cardiac S1PR3 in older animals. This data could be relevant for future clinical monitoring and patient selection in the aging MS population. Combinational therapies of fingolimod and duloxetine or tolterodine are well tolerated and safe without an increased risk for pronounced bradycardia or arrhythmia.


Asunto(s)
Envejecimiento/efectos de los fármacos , Bradicardia/inducido químicamente , Clorhidrato de Fingolimod/administración & dosificación , Clorhidrato de Fingolimod/efectos adversos , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Bradicardia/inmunología , Bradicardia/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Femenino , Ratones , Ratones Endogámicos C57BL , Telemetría/métodos
8.
Chin Med J (Engl) ; 130(2): 179-186, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-28091410

RESUMEN

BACKGROUND: The molecular mechanisms of Shenxianshengmai (SXSM), a traditional Chinese medicine, on bradycardia have been incompletely understood. The study tried to investigate the gene expression profile and proteomics of bradycardia rabbits' hearts after SXSM treatment. METHODS: Twenty-four adult rabbits were randomly assigned in four groups: sham, model, model plus SXSM treatment, and sham plus SXSM treatment groups. Heart rate was recorded in all rabbits. Then, total RNA of atria and proteins of ventricle were isolated and quantified, respectively. Gene expression profiling was conducted by gene expression chip, and quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed to confirm the results of gene expression chip. We used isobaric tags for elative and absolute quantitation and Western blotting to identify altered proteins after SXSM treatment. RESULTS: There was a constant decrease in the mean heart rate (32%, from 238 ± 6 beats/min to 149 ± 12 beats/min) after six weeks in model compared with that in sham group. This effect was partially reversed by 4-week SXSM treatment. Complementary DNA microarray demonstrated that the increased acetylcholinesterase and reduced nicotinic receptor were take responsibility for the increased heart rate. In addition, proteins involved in calcium handling and signaling were affected by SXSM treatment. Real-time RT-PCR verified the results from gene chip. Results from proteomics demonstrated that SXSM enhanced oxidative phosphorylation and tricarboxylic acid (TCA) cycle in ventricular myocardium to improve ATP generation. CONCLUSIONS: Long-term SXSM stimulates sympathetic transmission by increasing the expression of acetylcholinesterase and reduces the expression of nicotinic receptor to increase heart rate. SXSM also restored the calcium handling genes and altered genes involved in signaling. In addition, SXSM improves the ATP supply of ventricular myocardium by increasing proteins involved in TCA cycle and oxidation-respiratory chain.


Asunto(s)
Bradicardia/tratamiento farmacológico , Medicamentos Herbarios Chinos/uso terapéutico , Animales , Bradicardia/metabolismo , Bradicardia/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Proteómica , Conejos , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Brain Res ; 1652: 43-52, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27693394

RESUMEN

Previously, we reported that microinjection of L-proline (L-Pro) into the paraventricular nucleus of the hypothalamus (PVN) caused vasopressin-mediated pressor responses in unanesthetized rats. In the present study, we report on the central mechanisms involved in the mediation of the cardiovascular effects caused by the microinjection of L-Pro into the PVN. Microinjection of increasing doses of L-Pro (3-100nmol/100nL) into the PVN caused dose-related pressor and bradycardic responses. No cardiovascular responses were observed after the microinjection of equimolar doses (33nmol/100nL) of its isomer D-Proline (D-Pro) or Mannitol. The PVN pretreatment with either a selective non-NMDA (NBQX) or selective NMDA (LY235959 or DL-AP7) glutamate receptor antagonists blocked the cardiovascular response to L-Pro (33nmol/100nL). The dose-effect curve for the pretreatment with increasing doses of LY235959 was located at the left in relation to the curves for NBQX and DL-AP7, showing that LY235959 is more potent than NBQX, which is more potent than DL-AP7 in inhibiting the cardiovascular response to L-Pro. The cardiovascular response to the microinjection of L-Pro into the PVN was not affected by local pretreatment with Nω-Propyl-l-arginine (N-Propyl), a selective inhibitor of the neuronal nitric oxide synthase (nNOS), suggesting that NO does not mediate the responses to L-Pro in the PVN. In conclusion, the results suggest that ionotropic receptors in the PVN, blocked by both NMDA and non-NMDA receptor antagonists, mediate the pressor response to L-Pro that results from activation of PVN vasopressinergic magnocellular neurons and vasopressin release into the systemic circulation.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Fármacos del Sistema Nervioso Central/administración & dosificación , Neurotransmisores/administración & dosificación , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Prolina/administración & dosificación , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Bradicardia/inducido químicamente , Bradicardia/metabolismo , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Masculino , Microinyecciones , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo
10.
J Physiol ; 594(20): 5869-5879, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27374078

RESUMEN

Pacemaker activity of the sino-atrial node generates the heart rate. Disease of the sinus node and impairment of atrioventricular conduction induce an excessively low ventricular rate (bradycardia), which cannot meet the needs of the organism. Bradycardia accounts for about half of the total workload of clinical cardiologists. The 'sick sinus' syndrome (SSS) is characterized by sinus bradycardia and periods of intermittent atrial fibrillation. Several genetic or acquired risk factors or pathologies can lead to SSS. Implantation of an electronic pacemaker constitutes the only available therapy for SSS. The incidence of SSS is forecast to double over the next 50 years, with ageing of the general population thus urging the development of complementary or alternative therapeutic strategies. In recent years an increasing number of mutations affecting ion channels involved in sino-atrial automaticity have been reported to underlie inheritable SSS. L-type Cav 1.3 channels play a major role in the generation and regulation of sino-atrial pacemaker activity and atrioventricular conduction. Mutation in the CACNA1D gene encoding Cav 1.3 channels induces loss-of-function in channel activity and underlies the sino-atrial node dysfunction and deafness syndrome (SANDD). Mice lacking Cav 1.3 channels (Cav 1.3-/- ) fairly recapitulate SSS and constitute a precious model to test new therapeutic approaches to handle this disease. Work in our laboratory shows that targeting G protein-gated K+ (IKACh ) channels effectively rescues SSS of Cav 1.3-/- mice. This new concept of 'compensatory' ion channel targeting shines new light on the principles underlying the pacemaker mechanism and may open the way to new therapies for SSS.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Canalopatías/metabolismo , Ventrículos Cardíacos/metabolismo , Animales , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/fisiopatología , Canales de Calcio Tipo L/genética , Canalopatías/genética , Canalopatías/fisiopatología , Frecuencia Cardíaca/genética , Frecuencia Cardíaca/fisiología , Ventrículos Cardíacos/fisiopatología , Humanos , Mutación/genética , Síndrome del Seno Enfermo/genética , Síndrome del Seno Enfermo/metabolismo , Síndrome del Seno Enfermo/fisiopatología , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiopatología
11.
Am J Physiol Cell Physiol ; 310(3): C193-204, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26538090

RESUMEN

The extracellular calcium-sensing receptor CaSR is expressed in blood vessels where its role is not completely understood. In this study, we tested the hypothesis that the CaSR expressed in vascular smooth muscle cells (VSMC) is directly involved in regulation of blood pressure and blood vessel tone. Mice with targeted CaSR gene ablation from vascular smooth muscle cells (VSMC) were generated by breeding exon 7 LoxP-CaSR mice with animals in which Cre recombinase is driven by a SM22α promoter (SM22α-Cre). Wire myography performed on Cre-negative [wild-type (WT)] and Cre-positive (SM22α)CaSR(Δflox/Δflox) [knockout (KO)] mice showed an endothelium-independent reduction in aorta and mesenteric artery contractility of KO compared with WT mice in response to KCl and to phenylephrine. Increasing extracellular calcium ion (Ca(2+)) concentrations (1-5 mM) evoked contraction in WT but only relaxation in KO aortas. Accordingly, diastolic and mean arterial blood pressures of KO animals were significantly reduced compared with WT, as measured by both tail cuff and radiotelemetry. This hypotension was mostly pronounced during the animals' active phase and was not rescued by either nitric oxide-synthase inhibition with nitro-l-arginine methyl ester or by a high-salt-supplemented diet. KO animals also exhibited cardiac remodeling, bradycardia, and reduced spontaneous activity in isolated hearts and cardiomyocyte-like cells. Our findings demonstrate a role for CaSR in the cardiovascular system and suggest that physiologically relevant changes in extracellular Ca(2+) concentrations could contribute to setting blood vessel tone levels and heart rate by directly acting on the cardiovascular CaSR.


Asunto(s)
Presión Sanguínea , Señalización del Calcio , Calcio/metabolismo , Hipotensión/metabolismo , Músculo Liso Vascular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Vasoconstricción , Vasodilatación , Animales , Aorta/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/fisiopatología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Relación Dosis-Respuesta a Droga , Predisposición Genética a la Enfermedad , Frecuencia Cardíaca , Hipotensión/genética , Hipotensión/fisiopatología , Arterias Mesentéricas/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Miocitos Cardíacos/metabolismo , Fenotipo , Receptores Sensibles al Calcio , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Vasoconstricción/efectos de los fármacos , Vasoconstricción/genética , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Vasodilatadores/farmacología , Remodelación Ventricular
12.
Cardiology ; 128(1): 15-24, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24514589

RESUMEN

OBJECTIVES: Bradycardia is caused by loss-of-function mutations in potassium channels that regulate phase 3 repolarization of the cardiac action potential. The purpose of this study is to monitor the effects of potassium channel (KCNQ1) inhibition and to evaluate the effects of isoproterenol (ISO) and MgSO4 in restoring sinus rhythm in atrial cells. METHODS: Microelectrode array was used to analyze conduction velocity, voltage amplitude and cycle length of atrial cells (HL-1). A combination of ISO and MgSO4 was used to restore sinus rhythm in these cells. RESULTS: mRNA expression levels of KCNQ1 (42.2 vs. 100%, p < 0.0001), connexin 43 (29.6 vs. 100%, p = 0.0033), atrial natriuretic peptide (31.0 vs. 100%, p = 0.0030), cardiac actin (38.2 vs. 100%, p < 0.0001) and α-myosin heavy chain (31.2 vs. 100%, p = 0.00254) were significantly lower in the KCNQ1 gene-inhibited group compared to the control group. When treated with MgSO4 (1 mM) and ISO (10 µM), conduction velocity (0.0208 ± 0.0036 vs. 0.0086 ± 0.0014 m/s, p = 0.0004) and voltage amplitude (1,210.78 ± 65.81 vs. 124.1 ± 13.30 µV, p < 0.0001) were higher, and cycle length (431.55 ± 2.05 vs. 1,015.15 ± 4.31 ms, p < 0.0001) was shorter than in the gene-inhibited group. CONCLUSION: Inhibition of sinus rhythm in the bradycardia cell model was recovered by treatment with ISO and MgSO4, demonstrating the potency of combination therapy in the treatment of bradycardia.


Asunto(s)
Bradicardia/tratamiento farmacológico , Cardiotónicos/uso terapéutico , Isoproterenol/uso terapéutico , Canal de Potasio KCNQ1/metabolismo , Sulfato de Magnesio/uso terapéutico , Animales , Bradicardia/metabolismo , Cardiotónicos/farmacología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Isoproterenol/farmacología , Canal de Potasio KCNQ1/genética , Sulfato de Magnesio/farmacología , Ratones , Microelectrodos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño
13.
Circ Arrhythm Electrophysiol ; 6(4): 799-808, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23884198

RESUMEN

BACKGROUND: When complete atrioventricular block (AVB) occurs, infranodal escape rhythms are essential to prevent bradycardic death. The role of T-type Ca(2+) channels in pacemaking outside the sinus node is unknown. We investigated the role of T-type Ca(2+) channels in escape rhythms and bradycardia-related ventricular tachyarrhythmias after AVB in mice. METHODS AND RESULTS: Adult male mice lacking the main T-type Ca(2+) channel subunit Cav3.1 (Cav3.1(-/-)) and wild-type (WT) controls implanted with ECG telemetry devices underwent radiofrequency atrioventricular node ablation to produce AVB. Before ablation, Cav3.1(-/-) mice showed sinus bradycardia (mean±SEM; RR intervals, 148±3 versus 128±2 ms WT; P<0.001). Immediately after AVB, Cav3.1(-/-) mice had slower escape rhythms (RR intervals, 650±75 versus 402±26 ms in WT; P<0.01) but a preserved heart-rate response to isoproterenol. Over the next 24 hours, mortality was markedly greater in Cav3.1(-/-) mice (19/31; 61%) versus WT (8/26; 31%; P<0.05), and Torsades de Pointes occurred more frequently (73% Cav3.1(-/-) versus 35% WT; P<0.05). Escape rhythms improved in both groups during the next 4 weeks but remained significantly slower in Cav3.1(-/-). At 4 weeks after AVB, ventricular tachycardia was more frequent in Cav3.1(-/-) than in WT mice (746±116 versus 214±78 episodes/24 hours; P<0.01). Ventricular function remodeling was similar in Cav3.1(-/-) and WT, except for smaller post-AVB fractional-shortening increase in Cav3.1(-/-). Expression changes were seen post-AVB for a variety of genes; these tended to be greater in Cav3.1(-/-) mice, and overexpression of fetal and profibrotic genes occurred only in Cav3.1(-/-). CONCLUSIONS: This study suggests that T-type Ca(2+) channels play an important role in infranodal escape automaticity. Loss of T-type Ca(2+) channels worsens bradycardia-related mortality, increases bradycardia-associated adverse remodeling, and enhances the risk of malignant ventricular tachyarrhythmias complicating AVB.


Asunto(s)
Bloqueo Atrioventricular/metabolismo , Bradicardia/metabolismo , Canales de Calcio Tipo T/metabolismo , Señalización del Calcio , Sistema de Conducción Cardíaco/metabolismo , Frecuencia Cardíaca , Periodicidad , Torsades de Pointes/metabolismo , Potenciales de Acción , Animales , Bloqueo Atrioventricular/diagnóstico , Bloqueo Atrioventricular/genética , Bloqueo Atrioventricular/fisiopatología , Bradicardia/diagnóstico , Bradicardia/genética , Bradicardia/fisiopatología , Bradicardia/prevención & control , Canales de Calcio Tipo T/deficiencia , Canales de Calcio Tipo T/genética , Modelos Animales de Enfermedad , Electrocardiografía Ambulatoria , Técnicas Electrofisiológicas Cardíacas , Regulación de la Expresión Génica , Sistema de Conducción Cardíaco/fisiopatología , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Telemetría , Factores de Tiempo , Torsades de Pointes/diagnóstico , Torsades de Pointes/genética , Torsades de Pointes/fisiopatología , Torsades de Pointes/prevención & control , Remodelación Ventricular
14.
Biol Pharm Bull ; 34(10): 1523-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21963490

RESUMEN

Doxorubicin (DOX) is a potent antitumor agent, but the cardiotoxicity mediated by the formation of reactive oxygen species limit its clinical use. The present study aims to explore electrocardiographic and biochemical evidence for the cardioprotective effect of two antioxidants, Lycium barbarum polysaccharides (LBP, the main antioxidant in Lycium barbarum) and edaravone (a potent free radical scavenger, EDA) against DOX-induced acute cardiotoxicity in beagle dogs. In this study, male beagle dogs received daily treatment of either LBP (20 mg/kg, per os (p.o.)) or EDA (2 mg/kg, intravenously (i.v.)) for 7 d and then followed by an intravenous injection of DOX (1.5 mg/kg). DOX (15 mg/kg) significantly induced acute cardiotoxicity in dogs characterized by conduction abnormalities (including decreased heart rate, ST segment elevation, QT intervals prolongation, inverted T wave, arrhythmia, and myocardial ischemia) and increased serum creatine kinase (CK) and aspartate aminotransferase (AST). Pretreatment with LBP or EDA effectively alleviated both DOX-associated conduction abnormalities and increased serum CK and AST. Moreover, physiological and serum biochemical evidences demonstrated that EDA is more effective than LBP in alleviating these abnormalities produced by DOX in heart. All these results confirm and extend previous observations in rats concerning the effectiveness of LBP or EDA against DOX-induced cardiomyopathy.


Asunto(s)
Antioxidantes/farmacología , Antipirina/análogos & derivados , Cardiotónicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Electrocardiografía/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Cardiopatías/prevención & control , Enfermedad Aguda , Alanina Transaminasa/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/toxicidad , Antipirina/farmacología , Aspartato Aminotransferasas/efectos de los fármacos , Bradicardia/metabolismo , Creatina Quinasa/efectos de los fármacos , Perros , Doxorrubicina/farmacología , Doxorrubicina/toxicidad , Evaluación Preclínica de Medicamentos , Edaravona , Corazón , Sistema de Conducción Cardíaco/patología , Cardiopatías/inducido químicamente , Cardiopatías/patología , Masculino , Distribución Aleatoria
15.
Am J Physiol Heart Circ Physiol ; 299(5): H1459-67, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20709863

RESUMEN

Adenylyl cyclase (AC) is the principal effector molecule in the ß-adrenergic receptor pathway. AC(V) and AC(VI) are the two predominant isoforms in mammalian cardiac myocytes. The disparate roles among AC isoforms in cardiac hypertrophy and progression to heart failure have been under intense investigation. Specifically, the salutary effects resulting from the disruption of AC(V) have been established in multiple models of cardiomyopathy. It has been proposed that a continual activation of AC(V) through elevated levels of protein kinase C could play an integral role in mediating a hypertrophic response leading to progressive heart failure. Elevated protein kinase C is a common finding in heart failure and was demonstrated in murine cardiomyopathy from cardiac-specific overexpression of G(αq) protein. Here we assessed whether the disruption of AC(V) expression can improve cardiac function, limit electrophysiological remodeling, or improve survival in the G(αq) mouse model of heart failure. We directly tested the effects of gene-targeted disruption of AC(V) in transgenic mice with cardiac-specific overexpression of G(αq) protein using multiple techniques to assess the survival, cardiac function, as well as structural and electrical remodeling. Surprisingly, in contrast to other models of cardiomyopathy, AC(V) disruption did not improve survival or cardiac function, limit cardiac chamber dilation, halt hypertrophy, or prevent electrical remodeling in G(αq) transgenic mice. In conclusion, unlike other established models of cardiomyopathy, disrupting AC(V) expression in the G(αq) mouse model is insufficient to overcome several parallel pathophysiological processes leading to progressive heart failure.


Asunto(s)
Adenilil Ciclasas/metabolismo , Cardiomiopatías/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Adenilil Ciclasas/genética , Animales , Bradicardia/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnicas Electrofisiológicas Cardíacas , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Proteína Quinasa C/metabolismo
16.
Life Sci ; 83(13-14): 490-5, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18713640

RESUMEN

AIMS: To evaluate vagal stimulation-mediated myocardial protection against ischemia and reperfusion in in vivo ischemic myocardium. MAIN METHODS: We measured myocardial interstitial myoglobin levels in the ischemic region using a cardiac microdialysis technique in anesthetized and vagotomized cats. We occluded the left anterior descending coronary artery (LAD) for 60 min and reperfused it for 60 min (VX group, n = 6). The effects of bilateral vagal stimulation (10 V, 5 Hz, 1-ms pulse duration), initiated immediately after LAD occlusion, were examined (VS group, n = 6). To examine the involvement of phosphatidylinositol 3-kinase (PI3K), vagal stimulation was performed after pretreatment with a PI3K inhibitor wortmannin (0.6 mg/kg, i.v.) (VS-W group, n = 6). To examine the contribution of bradycardia, vagal stimulation was performed with fixed-rate ventricular pacing (VS-P group, n = 6). KEY FINDINGS: The average myoglobin level during the ischemic period was 1170+/-141 in VX (in ng/ml, mean+/-SE), which was significantly attenuated in VS (466+/-87, P<0.05) and VS-W (613+/-124, P<0.05) but not in VS-P (953+/-203). Reperfusion increased the myoglobin level to 2500+/-544 in VX, whereas it was suppressed in VS (824+/-213, P<0.05) and VS-W (948+/-315, P<0.05) but not in VS-P (1710+/-253). SIGNIFICANCE: Vagal stimulation, initiated immediately after LAD occlusion, attenuated the myocardial injury. Moreover, bradycardia, independent of PI3K pathway, plays a significant role in vagally induced cardioprotection during acute myocardial ischemia.


Asunto(s)
Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Mioglobina/metabolismo , Nervio Vago/metabolismo , Androstadienos/farmacología , Animales , Bradicardia/metabolismo , Bradicardia/fisiopatología , Estimulación Cardíaca Artificial , Gatos , Modelos Animales de Enfermedad , Terapia por Estimulación Eléctrica , Corazón/efectos de los fármacos , Microdiálisis , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Nervio Vago/efectos de los fármacos , Wortmanina
17.
J Cardiovasc Electrophysiol ; 15(11): 1310-6, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15574183

RESUMEN

INTRODUCTION: The sterol carrier protein-2 gene encodes two functionally distinct proteins: sterol carrier protein-2 (SCP2, a peroxisomal lipid carrier) and sterol carrier protein-x (SCPx, a peroxisomal thiolase known as peroxisomal thiolase-2), which is involved in peroxisomal metabolism of bile acids and branched-chain fatty acids. We show in this study that mice deficient in SCP2 and SCPx (SCP2null) develop a cardiac phenotype leading to a high sudden cardiac death rate if mice are maintained on diets enriched for phytol (a metabolic precursor of branched-chain fatty acids). METHODS AND RESULTS: In 210 surface and 305 telemetric ECGs recorded in wild-type (C57BL/6; wt; n = 40) and SCP2 null mice (n = 40), no difference was observed at baseline. However, on diet, cycle lengths were prolonged in SCP2 null mice (262.9 +/- 190 vs 146.3 +/- 43 msec), AV conduction was prolonged (58.3 +/- 17 vs 42.6 +/- 4 ms), and QRS complexes were wider (19.1 +/- 5 vs 14.0 +/- 4 ms). In 11 gene-targeted Langendorff-perfused hearts isolated from SCP2 null mice after dietary challenge, complete AV blocks (n = 5/11) or impaired AV conduction (Wenckebach point 132 +/- 27 vs 92 +/- 10 msec; P < 0.05) could be confirmed. Monophasic action potentials were not different between the two genotypes. Left ventricular function studied by echocardiography was similar in both strains. Phytanic acid but not pristanic acid accumulated in the phospholipid fraction of myocardial membranes isolated from SCP2 null mice. CONCLUSION: Accumulation of phytanic acid in myocardial phospholipid membranes is associated with bradycardia and impaired AV nodal and intraventricular impulse conduction, which could provide an explanation for sudden cardiac death in this model.


Asunto(s)
Proteínas Portadoras/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Ácido Fitánico/metabolismo , Animales , Bradicardia/metabolismo , Proteínas Portadoras/genética , Muerte Súbita Cardíaca , Dieta , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Ácido Fitánico/efectos adversos , Factores de Tiempo
18.
In. Melo, Celso Salgado de; Greco, Oswaldo Tadeu; Mateos, José Carlos Pachón. Temas de marcapasso. São Paulo, Lemos, 2; 2004. p.69-85, ilus.
Monografía en Portugués | LILACS, SES-SP, SESSP-IDPCPROD, SES-SP | ID: biblio-1069607

RESUMEN

Considera-se bradirritmia uma fequência cardíaca menor que 50 bpm. Evidentemente, esse conceito é relativo à situação metabólica e à idade do paciente, mesmo porque uma frequência cardíaca de 45bpm ou menos pode ser normal durante o sono em pacientes idosos, assim como uma frequência de 70 bpm constituti bradicardia em recém-nascidos ou em pacientes com febre. Isso demonstra que o diagnóstico depende do quadro clínico do paciente.


Asunto(s)
Masculino , Femenino , Recién Nacido , Adulto , Persona de Mediana Edad , Anciano , Humanos , Arritmia Sinusal/fisiopatología , Bloqueo Cardíaco/clasificación , Bloqueo Cardíaco/fisiopatología , Bradicardia/metabolismo , Diagnóstico Clínico , Frecuencia Cardíaca/fisiología
19.
Brain Res ; 991(1-2): 133-41, 2003 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-14575885

RESUMEN

Hypocretin-1 (hcrt-1)-containing axons have been shown to have an extensive distribution within the central nervous system, although the total number of hypothalamic hcrt-1 neurons has been shown to be small. This suggests that hcrt-1 neurons may innervate central structures with similar function through collateral axonal projections. Retrograde tract-tracing techniques combined with immunohistochemistry were used in this study to investigate whether hypothalamic hcrt-1-containing neurons send collateral axonal projections to cardiovascular sites in the nucleus of the solitary tract (NTS) and in the nucleus ambiguus (Amb) in the rat. Fluorogold- (FG) and/or rhodamine (Rd)-labeled latex microspheres were microinjected into either the NTS or Amb at sites that elicited bardycardia responses (L-glutamate; 0.25 M; 10 nl). After a survival period of 10-15 days, the rats were sacrificed and tissue sections of the hypothalamus were processed immunohistochemically for the identification of hcrt-1-containing cell bodies. After injection of the tract-tracers into the NTS or Amb, retrogradely labeled neurons were observed within several hypothalamic regions; the paraventricular hypothalamic nucleus, lateral hypothalamic area, perifornical hypothalamic area, and posterior hypothalamus, bilaterally, but with an ipsilateral predominance. In addition, after NTS injections, retrogradely labeled neurons were found within the ipsilateral caudal arcuate nucleus. Of the total number (1107+/-97) of hcrt-1-immunoreactive neurons found bilaterally within the lateral and perifornical hypothalamic nuclei, 7.9+/-1.4% were found to be retrogradely labeled from the NTS, 16.4+/-1.8% from the Amb, and 3.1+/-0.5% from both medullary sites. Hcrt-1 neurons projecting to the NTS were found mainly in and around the perifornical hypothalamic region, with a smaller number in the caudal lateral hypothalamic area. On the other hand, those innervating the Amb were primarily observed within the caudal lateral hypothalamic area, with a smaller number in the perifornical hypothalamic area. Neurons with collateral axonal projections to NTS and Amb were observed within two specific hypothalamic areas: one group of neurons was found in the perifornical hypothalamic area, and the other was observed in the lateral hypothalamic region just dorsal to the retrochiasmatic component of the supraoptic nucleus. These data indicate that axons from hcrt-1 neurons bifurcate to innervate functionally similar cardiovascular-responsive sites in the NTS and Amb. Although the function of these hcrt-1-containing hypothalamic-medullary pathways is not known, they likely represent the anatomical substrate by which the lateral hypothalamic hcrt-1 neurons simultaneously coordinate autonomic-cardiovascular responses to different behaviors.


Asunto(s)
Axones/fisiología , Sistema Cardiovascular/inervación , Proteínas Portadoras/metabolismo , Hipotálamo/anatomía & histología , Péptidos y Proteínas de Señalización Intracelular , Neuropéptidos/metabolismo , Núcleo Solitario/anatomía & histología , Animales , Bradicardia/metabolismo , Inmunohistoquímica , Masculino , Bulbo Raquídeo/anatomía & histología , Vías Nerviosas , Orexinas , Ratas
20.
Jpn J Pharmacol ; 69(2): 177-80, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8569056

RESUMEN

In anesthetized rats, intravenous administration of dl-tetrahydropalmatine (dl-THP, 1-10 mg/kg) elicited proportional hypotension, bradycardia and decreases in hypothalamic serotonin (5-HT) release (measured by carbon-fiber electrodes in combination with voltammetry). In addition, postsynaptic blockade of 5-HT2 receptors with cyproheptadine (2-5 mg/kg, i.v.) or ketanserin (2-5 mg/kg, i.v.) produced both hypotension and bradycardia, while stimulation of 5-HT2 receptors with 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) (10-250 mg/kg, i.v.) produced both hypertension and tachycardia. The dl-THP-induced hypotension and bradycardia could be reversed by DOI treatment. The data indicate that dl-THP decreases both arterial pressure and heart rate through a serotonergic release process in the hypothalamus.


Asunto(s)
Adrenérgicos/farmacología , Alcaloides de Berberina/farmacología , Bradicardia/metabolismo , Hipotensión/metabolismo , Hipotálamo/efectos de los fármacos , Serotonina/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA