Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Circ Res ; 134(10): 1379-1397, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38723031

ABSTRACT

Chagas cardiomyopathy caused by infection with the intracellular parasite Trypanosoma cruzi is the most common and severe expression of human Chagas disease. Heart failure, systemic and pulmonary thromboembolism, arrhythmia, and sudden cardiac death are the principal clinical manifestations of Chagas cardiomyopathy. Ventricular arrhythmias contribute significantly to morbidity and mortality and are the major cause of sudden cardiac death. Significant gaps still exist in the understanding of the pathogenesis mechanisms underlying the arrhythmogenic manifestations of Chagas cardiomyopathy. This article will review the data from experimental studies and translate those findings to draw hypotheses about clinical observations. Human- and animal-based studies at molecular, cellular, tissue, and organ levels suggest 5 main pillars of remodeling caused by the interaction of host and parasite: immunologic, electrical, autonomic, microvascular, and contractile. Integrating these 5 remodeling processes will bring insights into the current knowledge in the field, highlighting some key features for future management of this arrhythmogenic disease.


Subject(s)
Arrhythmias, Cardiac , Chagas Cardiomyopathy , Humans , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/parasitology , Arrhythmias, Cardiac/physiopathology , Chagas Cardiomyopathy/parasitology , Trypanosoma cruzi/pathogenicity , Chagas Disease/complications , Chagas Disease/parasitology , Chagas Disease/immunology
2.
J Bioenerg Biomembr ; 56(4): 373-387, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38869808

ABSTRACT

Hypercholesterolemia is one of the most important risk factors for cardiovascular diseases. However, it is mostly associated with vascular dysfunction and atherosclerotic lesions, while evidence of direct effects of hypercholesterolemia on cardiomyocytes and heart function is still incomplete and controversial. In this study, we assessed the direct effects of hypercholesterolemia on heart function and the electro-contractile properties of isolated cardiomyocytes. After 5 weeks, male Swiss mice fed with AIN-93 diet added with 1.25% cholesterol (CHO), developed an increase in total serum cholesterol levels and cardiomyocytes cholesterol content. These changes led to altered electrocardiographic records, with a shortening of the QT interval. Isolated cardiomyocytes displayed a shortening of the action potential duration with increased rate of depolarization, which was explained by increased IK, reduced ICa.L and altered INa voltage-dependent inactivation. Also, reduced diastolic [Ca2+]i was found with preserved adrenergic response and cellular contraction function. However, contraction of isolated hearts is impaired in isolated CHO hearts, before and after ischemia/reperfusion, although CHO heart was less susceptible to arrhythmic contractions. Overall, our results demonstrate that early hypercholesterolemia-driven increase in cellular cholesterol content is associated with direct modulation of the heart and cardiomyocytes' excitability, Ca2+ handling, and contraction.


Subject(s)
Hypercholesterolemia , Myocytes, Cardiac , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Hypercholesterolemia/physiopathology , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Mice , Male
3.
Clin Exp Pharmacol Physiol ; 51(10): e13915, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39227010

ABSTRACT

S-Limonene (s-Lim) is a monocyclic monoterpene found in a variety of plants and has been shown to present antioxidant and cardioprotective activity in experimental models of myocardial infarction. The aim of this study was to evaluate the potential mechanism by which s-Lim exerts its antiarrhythmic effect, focusing on the blockade of ß-adrenoceptor (ß-AR) and its effects on various in vivo and in vitro parameters, including electrocardiogram (ECG) measurements, left ventricular developed pressure (LVDP), the ß-adrenergic pathway, sarcomeric shortening and L-type calcium current (ICa,L). In isolated hearts, 10 µM of s-Lim did not alter the ECG profile or LVPD. s-Lim increased the heart rate corrected QT interval (QTc) (10.8%) at 50 µM and reduced heart rate at the concentrations of 30 (12.4%) and 50 µM (16.6%). s-Lim (10 µM) also inhibited the adrenergic response evoked by isoproterenol (ISO) (1 µM) reducing the increased of heart rate, LVDP and ECG changes. In ventricular cardiomyocyte, s-Lim antagonized the effect of dobutamine by preventing the increase of sarcomeric shortening, demonstrating a similar effect to atenolol (blocker ß1-AR). In vivo, s-Lim antagonized the effect of ISO (agonists ß1-AR), presenting a similar effect to propranolol (a non-selective blocker ß-AR). In ventricular cardiomyocyte, s-Lim did not alter the voltage dependence for ICa,L activation or the ICa,L density. In addition, s-Lim did not affect changes in the ECG effect mediated by 5 µM forskolin (an activator of adenylate cyclase). In an in vivo caffeine/ISO-induced arrhythmia model, s-Lim (1 mg/kg) presented antiarrhythmic action verified by a reduced arrhythmia score, heart rate, and occurrence of ventricular premature beats and inappropriate sinus tachycardia. These findings indicate that the antiarrhythmic activity of s-Lim is related to blockade of ß-AR in the heart.


Subject(s)
Anti-Arrhythmia Agents , Limonene , Rats, Wistar , Receptors, Adrenergic, beta , Signal Transduction , Animals , Rats , Anti-Arrhythmia Agents/pharmacology , Male , Receptors, Adrenergic, beta/metabolism , Limonene/pharmacology , Signal Transduction/drug effects , Terpenes/pharmacology , Heart/drug effects , Heart Rate/drug effects , Cyclohexenes/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism
4.
Toxicol Appl Pharmacol ; 474: 116609, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37392997

ABSTRACT

BACKGROUND: Arrhythmias are one manifestation of the cardiotoxicity that results from doxorubicin (Doxo) administration. Although cardiotoxicity is an anticipated outcome in anticancer therapies, there is still a lack of treatment options available for its effective management. This study sought to evaluate the possible cardioprotective effect of complex d-limonene (DL) plus hydroxypropyl-ß-cyclodextrin (HßDL) during treatment with Doxo, focusing on the arrhythmic feature. METHODS: Cardiotoxicity was induced in Swiss mice with Doxo 20 mg/kg, with 10 mg/kg of HßDL being administered 30 min before the Doxo. Plasma CK-MB and LDH levels were analyzed. Cellular excitability and susceptibility to cardiac and cardiomyocyte arrhythmias were evaluated using in vivo (pharmacological cardiac stress) and in vitro (burst pacing) ECG protocols. Ca2+ dynamics were also investigated. The expression of CaMKII and its activation by phosphorylation and oxidation were evaluated by western blot, and molecular docking was used to analyze the possible interaction between DL and CaMKII. RESULTS: Electrocardiograms showed that administration of 10 mg/kg of HßDL prevented Doxo-induced widening of the QRS complex and QT interval. HßDL also prevented cardiomyocyte electrophysiological changes that trigger cellular arrhythmias, such as increases in action potential duration and variability; decreased the occurrence of delayed afterdepolarizations (DADs) and triggered activities (TAs), and reduced the incidence of arrhythmia in vivo. Ca2+ waves and CaMKII overactivation caused by phosphorylation and oxidation were also decreased. In the in silico study, DL showed potential inhibitory interaction with CaMKII. CONCLUSION: Our results show that 10 mg/kg of ßDL protects the heart against Doxo-induced cardiotoxicity arrhythmias, and that this is probably due to its inhibitory effect on CaMKII hyperactivation.


Subject(s)
Calcium , Cyclodextrins , Mice , Animals , Limonene/adverse effects , Limonene/metabolism , Calcium/metabolism , Cardiotoxicity/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Molecular Docking Simulation , Doxorubicin/adverse effects , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/prevention & control , Arrhythmias, Cardiac/metabolism , Myocytes, Cardiac
5.
Clin Exp Pharmacol Physiol ; 50(11): 893-902, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37610053

ABSTRACT

Atrial arrhythmias (AA) are common in pulmonary hypertension (PH) and are closely associated with poor clinical outcomes. One of the most studied models to investigate PH is the rat model of monocrotaline (MCT) induced PH (MCT-PH). To date, little is known about right atrium (RA) function in the MCT-PH model and the propensity of RA to develop arrhythmias. Therefore, the aim of the study was to evaluate the function of the RA of control (CTRL) and MCT treated rats, and the ability of amiodarone, a classical antiarrhythmic, to prevent the occurrence of AA in the RA in MCT-PH rats. RA function was studied in MCT-PH rats 20 days after a single subcutaneous injection of MCT 50 mg/kg. The histological results indicated the presence of RA and right ventricular hypertrophy. Surface electrocardiogram demonstrated increased P wave duration, PR wave duration and QT interval in MCT rats. RA from MCT rats were more susceptible to develop ex vivo burst pacing arrhythmias when compared to CTRL. Intriguingly, amiodarone in clinical relevant concentration was not able to prevent the occurrence arrhythmias in RA from MCT-PH animals. Hence, we conclude that the rat model of MCT-PH impairs RA structure and function, and acute exposure of RA to amiodarone in clinical relevant concentration is not able to attenuate the onset of arrhythmias in the ex vivo RA preparation.

6.
Phytother Res ; 37(6): 2531-2551, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37115723

ABSTRACT

Sepsis is a host's dysregulated immune response to an infection associated with systemic inflammation and excessive oxidative stress, which can cause multiple organ failure and death. The literature suggests that flavonoids, a broad class of secondary plant metabolites, have numerous biological activities which can be valuable in the treatment of sepsis. This study aimed to review the effects of flavonoids on experimental sepsis, focusing mainly on survival rate, and also summarizing information on its mechanisms of action. We searched in the main databases up to November 2022 using relevant keywords, and data were extracted and analyzed qualitatively and quantitatively. Thirty-two articles met the study criteria for review and 29 for meta-analysis. Overall, 30 different flavonoids were used in the studies. The flavonoids were able to strongly inhibit inflammatory response by reducing the levels of important pro-inflammatory mediators, for example, tumor necrosis factor-alpha and interleukin-1ß, oxidative stress, and showed antibacterial and anti-apoptotic actions. The meta-analysis found an increase of 50% in survival rate of the animals treated with flavonoids. They appear to act as multi-target drugs and may be an excellent therapeutic alternative to reduce a number of the complications caused by sepsis, and consequently, to improve survival rate.


Subject(s)
Flavonoids , Sepsis , Animals , Flavonoids/pharmacology , Flavonoids/therapeutic use , Inflammation/drug therapy , Oxidative Stress , Tumor Necrosis Factor-alpha/pharmacology , Sepsis/drug therapy
7.
PLoS Pathog ; 16(3): e1008379, 2020 03.
Article in English | MEDLINE | ID: mdl-32160269

ABSTRACT

Chagas Disease (CD) is one of the leading causes of heart failure and sudden death in Latin America. Treatments with antioxidants have provided promising alternatives to ameliorate CD. However, the specific roles of major reactive oxygen species (ROS) sources, including NADPH-oxidase 2 (NOX2), mitochondrial-derived ROS and nitric oxide (NO) in the progression or resolution of CD are yet to be elucidated. We used C57BL/6 (WT) and a gp91PHOX knockout mice (PHOX-/-), lacking functional NOX2, to investigate the effects of ablation of NOX2-derived ROS production on the outcome of acute chagasic cardiomyopathy. Infected PHOX-/- cardiomyocytes displayed an overall pro-arrhythmic phenotype, notably with higher arrhythmia incidence on ECG that was followed by higher number of early afterdepolarizations (EAD) and 2.5-fold increase in action potential (AP) duration alternans, compared to AP from infected WT mice. Furthermore, infected PHOX-/- cardiomyocytes display increased diastolic [Ca2+], aberrant Ca2+ transient and reduced Ca2+ transient amplitude. Cardiomyocyte contraction is reduced in infected WT and PHOX-/- mice, to a similar extent. Nevertheless, only infected PHOX-/- isolated cardiomyocytes displayed significant increase in non-triggered extra contractions (appearing in ~75% of cells). Electro-mechanical remodeling of infected PHOX-/-cardiomyocytes is associated with increase in NO and mitochondria-derived ROS production. Notably, EADs, AP duration alternans and in vivo arrhythmias were reverted by pre-incubation with nitric oxide synthase inhibitor L-NAME. Overall our data show for the first time that lack of NOX2-derived ROS promoted a pro-arrhythmic phenotype in the heart, in which the crosstalk between ROS and NO could play an important role in regulating cardiomyocyte electro-mechanical function during acute CD. Future studies designed to evaluate the potential role of NOX2-derived ROS in the chronic phase of CD could open new and more specific therapeutic strategies to treat CD and prevent deaths due to heart complications.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Signaling , Chagas Cardiomyopathy/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Acute Disease , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium/metabolism , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/pathology , Chagas Cardiomyopathy/physiopathology , Disease Models, Animal , Male , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism
9.
Clin Sci (Lond) ; 136(5): 329-343, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35190819

ABSTRACT

ß-Cyfluthrin, a class II Pyrethroid, is an insecticide used worldwide in agriculture, horticulture (field and protected crops), viticulture, and domestic applications. ß-Cyfluthrin may impair the function of biological systems; however, little information is available about its potential cardiotoxic effect. Here, we explored the acute toxicity of ß-Cyfluthrin in isolated heart preparations and its cellular basis, using isolated cardiomyocytes. Moreover, ß-Cyfluthrin effects on the sodium current, especially late sodium current (INa-L), were investigated using human embryonic kidney cells (HEK-293) cells transiently expressing human NaV1.5 channels. We report that ß-Cyfluthrin raised INa-L in a dose-dependent manner. ß-Cyfluthrin prolonged the repolarization of the action potential (AP) and triggered oscillations on its duration. Cardiomyocytes contraction and calcium dynamics were disrupted by the pesticide with a marked incidence of non-electronic-stimulated contractions. The antiarrhythmic drug Ranolazine was able to reverse most of the phenotypes observed in isolated cells. Lastly, ventricular premature beats (VPBs) and long QT intervals were found during ß-Cyfluthrin exposure, and Ranolazine was able to attenuate them. Overall, we demonstrated that ß-Cyfluthrin can cause significant cardiac alterations and Ranolazine ameliorated the phenotype. Understanding the insecticides' impacts upon electromechanical properties of the heart is important for the development of therapeutic approaches to treat cases of pesticides intoxication.


Subject(s)
Insecticides , Pyrethrins , Action Potentials , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , HEK293 Cells , Humans , Insecticides/toxicity , Myocytes, Cardiac , Nitriles , Phenotype , Pyrethrins/pharmacology , Ranolazine/pharmacology , Sodium , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use
10.
FASEB J ; 35(10): e21901, 2021 10.
Article in English | MEDLINE | ID: mdl-34569665

ABSTRACT

Chagasic cardiomyopathy (CCC) is one of the main causes of heart failure and sudden death in Latin America. To date, there is no available medication to prevent or reverse the onset of cardiac symptoms. CCC occurs in a scenario of disrupted calcium dynamics and enhanced oxidative stress, which combined, may favor the hyper activation of calcium/calmodulin (Ca2+ /CaM)-calcium/calmodulin-dependent protein kinase II (CaMKII) (Ca2+ /CaM-CaMKII) pathway, which is fundamental for heart physiology and it is implicated in other cardiac diseases. Here, we evaluated the association between Ca2+ /CaM-CaMKII in the electro-mechanical (dys)function of the heart in the early stage of chronic experimental Trypanosoma cruzi infection. We observed that in vitro and ex vivo inhibition of Ca2+ /CaM-CaMKII reversed the arrhythmic profile of isolated hearts and isolated left-ventricles cardiomyocytes. The benefits of the limited Ca2+ /CaM-CaMKII activation to cardiomyocytes' electrical properties are partially related to the restoration of Ca2+ dynamics in a damaged cellular environment created after T. cruzi infection. Moreover, Ca2+ /CaM-CaMKII inhibition prevented the onset of arrhythmic contractions on isolated heart preparations of chagasic mice and restored the responsiveness to the increase in the left-ventricle pre-load. Taken together, our data provide the first experimental evidence for the potential of targeting Ca2+ /CaM-CaMKII pathway as a novel therapeutic target to treat CCC.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Calmodulin/metabolism , Chagas Cardiomyopathy/metabolism , Trypanosoma cruzi/metabolism , Animals , Arrhythmias, Cardiac/parasitology , Chagas Cardiomyopathy/parasitology , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C
11.
Mol Pharmacol ; 99(6): 448-459, 2021 06.
Article in English | MEDLINE | ID: mdl-33824187

ABSTRACT

Nav1.5-derived Na+ current (INa) exerts a pivotal role in the depolarization phase of cardiomyocytes' action potential, and, therefore, changes in INa can contribute to fatal arrhythmias. Nav1.5 displays naturally occurring ethnicity-related polymorphisms, which might alter the functioning and pharmacology of the channel. Some studies have shown how single-nucleotide polymorphism can change the response to antiarrhythmic drugs. Investigations on the role of Nav1.5 in arrhythmogenesis associated with its functional polymorphisms are currently growing as well as the possible variability in the antiarrhythmic pharmacotherapy among ethnic groups. The influence of the ethnicity-related polymorphisms (S524Y, S1103Y, R1193Q, V1951L) on the responsiveness, selectivity, and pharmacological efficacy of the clinically used antiarrhythmic amiodarone (AMIO) is not completely known. Our objectives were to analyze biophysical and pharmacological aspects of four ethnicity-related polymorphisms before and after exposure to AMIO. Polymorphisms caused reduced AMIO potency compared with wild type (WT), which can vary by up to 4× between them. AMIO shifted the voltage dependency for current inactivation without significant effect in voltage-dependent activation to a similar extent in WT and polymorphisms. The recovery from inactivation was altered between the polymorphisms when compared with WT. Finally, the use dependency of AMIO differed between studied groups, especially at a more depolarized cell membrane. Thus, our work may guide future studies focusing on the efficiency of AMIO in treating different arrhythmias and establish more individualized guidelines for its use depending on the Nav1.5 polymorphism after validating our findings using in vivo studies. SIGNIFICANCE STATEMENT: Sodium voltage-gated channel α subunit 5 (SCN5A) gene encodes the α subunit of Nav1.5, the main cardiac voltage-gated Na+ channel. Interestingly, ethnicity-related polymorphisms are found in SCN5A. Amiodarone is used in clinical practice, and some of its effects are attributed to interaction with Nav1.5. Important, amiodarone efficacy is variable among patients. Here we show that ethnicity-related SCN5A polymorphisms lead to altered Nav1.5-amiodarone interaction, which may be the cause for the variable efficacy observed in clinical usage of amiodarone.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Ethnicity/genetics , NAV1.5 Voltage-Gated Sodium Channel/drug effects , NAV1.5 Voltage-Gated Sodium Channel/genetics , Polymorphism, Single Nucleotide , HEK293 Cells , Humans
12.
Am J Physiol Cell Physiol ; 312(4): C478-C486, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28122728

ABSTRACT

Little is known regarding the role of suppressor of cytokine signaling (SOCS) in the control of cytokine signaling in cardiomyocytes. We investigated the consequences of SOCS2 ablation for leukemia inhibitory factor (LIF)-induced enhancement of intracellular Ca2+ ([Ca2+]i) transient by performing experiments with cardiomyocytes from SOCS2-knockout (ko) mice. Similar levels of SOCS3 transcripts were seen in cardiomyocytes from wild-type and SOCS2-ko mice, while SOCS1 mRNA was reduced in SOCS2-ko. Immunoprecipitation experiments showed increased SOCS3 association with gp130 receptor in SOCS2-ko myocytes. Measurements of Ca2+ in wild-type myocytes exposed to LIF showed a significant increase in the magnitude of the Ca2+ transient. This change was absent in LIF-treated SOCS2-ko cells. LIF activation of ERK and STAT3 was observed in both wild-type and SOCS2-ko cells, indicating that in SOCS2-ko, LIF receptors were functional, despite the lack of effect in the Ca2+ transient. In wild-type cells, LIF-induced increase in [Ca2+]i and phospholamban Thr17 [PLN(Thr17)] phosphorylation was inhibited by KN-93, indicating a role for CaMKII in LIF-induced Ca2+ raise. LIF-induced phosphorylation of PLN(Thr17) was abrogated in SOCS2-ko myocytes. In wild-type cardiomyocytes, LIF treatment increased L-type Ca2+ current (ICa,L), a key activator of CaMKII in response to LIF. Conversely, SOCS2-ko myocytes failed to activate ICa,L in response to LIF, providing a rationale for the lack of LIF effect on Ca2+ transient. Our data show that absence of SOCS2 turns cardiomyocytes unresponsive to LIF-induced [Ca2+] raise, indicating that endogenous levels of SOCS2 are crucial for full activation of LIF signaling in the heart.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Leukemia Inhibitory Factor/metabolism , Myocytes, Cardiac/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Line , Gene Expression Regulation/physiology , Male , Mice , Mice, Knockout , Suppressor of Cytokine Signaling Proteins/genetics
13.
Int J Sports Med ; 38(12): 949-953, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28922681

ABSTRACT

We investigated the effects of palmitate, a high saturated fat, on Ca2+, action potential and reactive oxygen species dynamics in cardiomyocytes from untrained and trained mice. Male mice were subjected to moderate intensity exercise training on a treadmill. Cardiomyocytes of untrained and trained mice were isolated, treated for 30 min with palmitate and intracellular calcium transient and action potential duration were recorded. Additionally, we assessed reactive oxygen species generation. Treatment of cardiomyocytes from untrained mice with palmitate induced a significant decrease in Ca2+ transient magnitude by 34%. Exercise training did not change cardiomyocyte Ca2+ dynamics in the control group. However, trained cardiomyocytes were protected from deleterious effects of palmitate. Action potential duration was not altered by palmitate in either untrained or trained cardiomyocytes. Moreover, palmitate treatment increased reactive oxygen species generation in both untrained and trained cardiomyocytes. Nevertheless, the levels of reactive oxygen species in trained cardiomyocytes treated with palmitate were still 27% lower than those seen at basal conditions in untrained cardiomyocytes. Taken together, these findings indicate that exercise training protects cardiomyocytes from deleterious effects of palmitate possibly by inhibiting exacerbated ROS production.


Subject(s)
Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Palmitic Acid/toxicity , Physical Conditioning, Animal , Action Potentials/drug effects , Animals , Body Weight , Heart/anatomy & histology , Male , Mice, Inbred C57BL , Organ Size , Reactive Oxygen Species/metabolism
14.
N Engl J Med ; 369(4): 351-361, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23883380

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension is a devastating disease with high mortality. Familial cases of pulmonary arterial hypertension are usually characterized by autosomal dominant transmission with reduced penetrance, and some familial cases have unknown genetic causes. METHODS: We studied a family in which multiple members had pulmonary arterial hypertension without identifiable mutations in any of the genes known to be associated with the disease, including BMPR2, ALK1, ENG, SMAD9, and CAV1. Three family members were studied with whole-exome sequencing. Additional patients with familial or idiopathic pulmonary arterial hypertension were screened for the mutations in the gene that was identified on whole-exome sequencing. All variants were expressed in COS-7 cells, and channel function was studied by means of patch-clamp analysis. RESULTS: We identified a novel heterozygous missense variant c.608 G→A (G203D) in KCNK3 (the gene encoding potassium channel subfamily K, member 3) as a disease-causing candidate gene in the family. Five additional heterozygous missense variants in KCNK3 were independently identified in 92 unrelated patients with familial pulmonary arterial hypertension and 230 patients with idiopathic pulmonary arterial hypertension. We used in silico bioinformatic tools to predict that all six novel variants would be damaging. Electrophysiological studies of the channel indicated that all these missense mutations resulted in loss of function, and the reduction in the potassium-channel current was remedied by the application of the phospholipase inhibitor ONO-RS-082. CONCLUSIONS: Our study identified the association of a novel gene, KCNK3, with familial and idiopathic pulmonary arterial hypertension. Mutations in this gene produced reduced potassium-channel current, which was successfully remedied by pharmacologic manipulation. (Funded by the National Institutes of Health.)


Subject(s)
Channelopathies/genetics , Hypertension, Pulmonary/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Amino Acid Sequence , Channelopathies/drug therapy , Chlorobenzoates/therapeutic use , Cinnamates/therapeutic use , Exome , Familial Primary Pulmonary Hypertension , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Hypertension, Pulmonary/drug therapy , Lung/pathology , Male , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Pedigree , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/physiology , Sequence Analysis, DNA , ortho-Aminobenzoates/therapeutic use
15.
Pharmacol Rep ; 76(3): 585-599, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38619735

ABSTRACT

BACKGROUND: Amiodarone (AMIO) is an antiarrhythmic drug with the pKa in the physiological range. Here, we explored how mild extracellular pH (pHe) changes shape the interaction of AMIO with atrial tissue and impact its pharmacological properties in the classical model of sea anemone sodium channel neurotoxin type 2 (ATX) induced late sodium current (INa-Late) and arrhythmias. METHOD: Isolated atrial cardiomyocytes from male Wistar rats and human embryonic kidney cells expressing SCN5A Na+ channels were used for patch-clamp experiments. Isolated right atria (RA) and left atria (LA) tissue were used for bath organ experiments. RESULTS: A more acidophilic pHe caused negative inotropic effects on isolated RA and LA atrial tissue, without modification of the pharmacological properties of AMIO. A pHe of 7.0 changed the sodium current (INa) related components of the action potential (AP), which was enhanced in the presence of AMIO. ATXinduced arrhythmias in isolated RA and LA. Also, ATX prolonged the AP duration and enhanced repolarization dispersion in isolated cardiomyocytes in both pHe 7.4 and pHe 7.0. Pre-incubation of the isolated RA and LA and isolated atrial cardiomyocytes with AMIO prevented arrhythmias induced by ATX only at a pHe of 7.0. Moreover, AMIO was able to block INa-Late induced by ATX only at a pHe of 7.0. CONCLUSION: The pharmacological properties of AMIO concerning healthy rat atrial tissue are not dependent on pHe. However, the prevention of arrhythmias induced by INa-Late is pHe-dependent. The development of drugs analogous to AMIO with charge stabilization may help to create more effective drugs to treat arrhythmias related to the INa-Late.


Subject(s)
Action Potentials , Amiodarone , Anti-Arrhythmia Agents , Arrhythmias, Cardiac , Heart Atria , Myocytes, Cardiac , Rats, Wistar , Animals , Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Male , Humans , Rats , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Heart Atria/drug effects , Heart Atria/metabolism , Hydrogen-Ion Concentration , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced , NAV1.5 Voltage-Gated Sodium Channel/metabolism , HEK293 Cells , Sodium/metabolism , Patch-Clamp Techniques , Cnidarian Venoms/pharmacology
16.
Eur J Pharmacol ; : 176980, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39241944

ABSTRACT

Dronedarone (DRN) is a clinically used drug to mitigate arrhythmias with multichannel block properties, including the sodium channel Nav1.5. Extracellular acidification is known to change the pharmacological properties of several antiarrhythmic drugs. Here, we explore how modification in extracellular pH (pHe) shapes the pharmacological profile of DRN upon Nav1.5 sodium current (INa) and in the ex vivo heart preparation. Embryonic human kidney cells (HEK293T/17) were used to transiently express the Nav1.5 α-subunit. Patch-Clamp technique was employed to study INa. Neurotoxin-II (ATX-II) was used to induce the late sodium current (INaLate). Additionally, ex vivo Wistar male rat preparations in the Langendorff system were utilized to study electrocardiogram (ECG) waves. DRN preferentially binds to the closed state inactivation mode of Nav1.5 at pHe 7.0. The recovery from INa inactivation was delayed in the presence of DRN in both pHe 7.0 and 7.4, and the use-dependent properties were distinct at pHe 7.0 and 7.4. However, the potency of DRN upon the peak INa, the voltage dependence for activation, and the steady-state inactivation curves were not altered in both pHe tested. Also, the pHe did not change the ability of DRN to block INaLate. Lastly, DRN in a concentration and pH dependent manner modulated the QRS complex, QT and RR interval in clinically relevant concentration. Thus, the pharmacological properties of DRN upon Nav1.5 and ex vivo heart preparation partially depend on the pHe. The pHe changed the biological effect of DRN in the heart electrical function in relevant clinical concentration.

17.
Food Chem Toxicol ; 187: 114596, 2024 May.
Article in English | MEDLINE | ID: mdl-38556154

ABSTRACT

Tebuconazole (TEB), a widely used pesticide in agriculture to combat fungal infections, is commonly detected in global food, potable water, groundwater, and human urine samples. Despite its known in vivo toxicity, its impact on heart function remains unclear. In a 28-day study on male Wistar rats (approximately 100 g), administering 10 mg/kg/day TEB or a vehicle (control) revealed no effect on body weight gain or heart weight, but an increase in the infarct area in TEB-treated animals. Notably, TEB induced time-dependent changes in in vivo electrocardiograms, particularly prolonging the QT interval after 28 days of administration. Isolated left ventricular cardiomyocytes exposed to TEB exhibited lengthened action potentials and reduced transient outward potassium current. TEB also increased reactive oxygen species (ROS) production in these cardiomyocytes, a phenomenon reversed by N-acetylcysteine (NAC). Furthermore, TEB-treated animals, when subjected to an in vivo dobutamine (Dob) and caffeine (Caf) challenge, displayed heightened susceptibility to severe arrhythmias, a phenotype prevented by NAC. In conclusion, TEB at the no observed adverse effect level (NOAEL) dose adversely affects heart electrical function, increases arrhythmic susceptibility, partially through ROS overproduction, and this phenotype is reversible by scavenging ROS with NAC.


Subject(s)
Arrhythmias, Cardiac , Dobutamine , Triazoles , Humans , Rats , Animals , Male , Reactive Oxygen Species , Rats, Wistar , Arrhythmias, Cardiac/chemically induced , Acetylcysteine , Myocytes, Cardiac
18.
Am J Pathol ; 181(1): 130-40, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22658486

ABSTRACT

Infection with Trypanosoma cruzi induces inflammation, which limits parasite proliferation but may result in chagasic heart disease. Suppressor of cytokine signaling 2 (SOCS2) is a regulator of immune responses and may therefore participate in the pathogenesis of T. cruzi infection. SOCS2 is expressed during T. cruzi infection, and its expression is partially reduced in infected 5-lipoxygenase-deficient [knockout (KO)] mice. In SOCS2 KO mice, there was a reduction in both parasitemia and the expression of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), IL-6, IL-10, SOCS1, and SOCS3 in the spleen. Expression of IFN-γ, TNF-α, SOCS1, and SOCS3 was also reduced in the hearts of infected SOCS2 KO mice. There was an increase in the generation and expansion of T regulatory (Treg) cells and a decrease in the number of memory cells in T. cruzi-infected SOCS2 KO mice. Levels of lipoxinA(4) (LXA(4)) increased in these mice. Echocardiography studies demonstrated an impairment of cardiac function in T. cruzi-infected SOCS2 KO mice. There were also changes in calcium handling and in action potential waveforms, and reduced outward potassium currents in isolated cardiac myocytes. Our data suggest that reductions of inflammation and parasitemia in infected SOCS2-deficient mice may be secondary to the increases in Treg cells and LXA(4) levels. This occurs at the cost of greater infection-associated heart dysfunction, highlighting the relevance of balanced inflammatory and immune responses in preventing severe T. cruzi-induced disease.


Subject(s)
Chagas Cardiomyopathy/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Acute Disease , Animals , Arachidonate 5-Lipoxygenase/physiology , Cells, Cultured , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/pathology , Chagas Cardiomyopathy/physiopathology , Cytokines/biosynthesis , Disease Models, Animal , Heart/parasitology , Lipoxins/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/immunology , Parasite Load , Parasitemia/immunology , Patch-Clamp Techniques , Suppressor of Cytokine Signaling Proteins/deficiency , T-Lymphocyte Subsets/immunology , Trypanosoma cruzi/isolation & purification
19.
Proc Natl Acad Sci U S A ; 107(9): 4442-7, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20147620

ABSTRACT

Morphine is one of the most prescribed and effective drugs used for the treatment of acute and chronic pain conditions. In addition to its central effects, morphine can also produce peripheral analgesia. However, the mechanisms underlying this peripheral action of morphine have not yet been fully elucidated. Here, we show that the peripheral antinociceptive effect of morphine is lost in neuronal nitric-oxide synthase null mice and that morphine induces the production of nitric oxide in primary nociceptive neurons. The activation of the nitric-oxide pathway by morphine was dependent on an initial stimulation of PI3Kgamma/AKT protein kinase B (AKT) and culminated in increased activation of K(ATP) channels. In the latter, this intracellular signaling pathway might cause a hyperpolarization of nociceptive neurons, and it is fundamental for the direct blockade of inflammatory pain by morphine. This understanding offers new targets for analgesic drug development.


Subject(s)
KATP Channels/metabolism , Morphine/therapeutic use , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Pain/drug therapy , Pain/enzymology , Pain/metabolism , Rats , Rats, Wistar
20.
Mem Inst Oswaldo Cruz ; 108(2): 243-5, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23579807

ABSTRACT

Chagas disease, which is caused by the parasite Trypanosoma cruzi, is an important cause of heart failure. We investigated modifications in the cellular electrophysiological and calcium-handling characteristics of an infected mouse heart during the chronic phase of the disease. The patch-clamp technique was used to record action potentials (APs) and L-type Ca2+ and transient outward K+ currents. [Ca2+]i changes were determined using confocal microscopy. Infected ventricular cells showed prolonged APs, reduced transient outward K+ and L-type Ca2+ currents and reduced Ca2+ release from the sarcoplasmic reticulum. Thus, the chronic phase of Chagas disease is characterised by cardiomyocyte dysfunction, which could lead to heart failure.


Subject(s)
Action Potentials/physiology , Chagas Disease/physiopathology , Myocytes, Cardiac/physiology , Sodium-Calcium Exchanger/physiology , Animals , Chronic Disease , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL