Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26.829
Filter
1.
Bull Exp Biol Med ; 177(1): 57-62, 2024 May.
Article in English | MEDLINE | ID: mdl-38954299

ABSTRACT

We studied the effect of Refralon on the electrophysiological properties of the supraventricular myocardium against the background of adrenergic (epinephrine) influence in the zone of the pulmonary veins, the area where 50-90% of atrial arrhythmias is triggered. The experiments were carried out on isolated tissue preparations of Wistar rats. The multichannel microelectrode array technique was used to record action potentials simultaneously in the atrium and in the ostium and distal parts of the pulmonary veins. Epinephrine application (12-50 nM) led to depolarization of the resting potential and the conduction block in the distal part of the pulmonary veins. Refralon (30 µg/kg) restored the resting potential in the distal part of the pulmonary veins. Against the background of epinephrine, Refralon did not significantly change the duration of the action potential at 90% repolarization in comparison with control. At the same time, the comparison drug E-4031 against the background of epinephrine significantly increased the duration of action potential in the atrium and in the ostium of the pulmonary veins, and sotalol increased it only in the ostium. Neither E-4031, nor sotalol restored conduction in their distal part. Refralon has a biphasic effect under conditions of adrenergic stimulation: the fast component is responsible for stabilizing the resting potential in the pulmonary vein and reduces the dispersion of action potential duration in the atrium and pulmonary vein and is also quickly washed away, and the slow component is responsible for the increase of the action potential duration and is slowly washed away.


Subject(s)
Action Potentials , Anti-Arrhythmia Agents , Epinephrine , Heart Atria , Pulmonary Veins , Rats, Wistar , Animals , Rats , Epinephrine/pharmacology , Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Pulmonary Veins/drug effects , Male , Heart Atria/drug effects , Heart Atria/physiopathology , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/drug therapy
4.
Sci Rep ; 14(1): 15683, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977794

ABSTRACT

We explored physiological effects of the sodium-glucose co-transporter-2 inhibitor empagliflozin on intact experimentally hypertrophic murine hearts following transverse aortic constriction (TAC). Postoperative drug (2-6 weeks) challenge resulted in reduced late Na+ currents, and increased phosphorylated (p-)CaMK-II and Nav1.5 but not total (t)-CaMK-II, and Na+/Ca2+ exchanger expression, confirming previous cardiomyocyte-level reports. It rescued TAC-induced reductions in echocardiographic ejection fraction and fractional shortening, and diastolic anterior and posterior wall thickening. Dual voltage- and Ca2+-optical mapping of Langendorff-perfused hearts demonstrated that empagliflozin rescued TAC-induced increases in action potential durations at 80% recovery (APD80), Ca2+ transient peak signals and durations at 80% recovery (CaTD80), times to peak Ca2+ (TTP100) and Ca2+ decay constants (Decay30-90) during regular 10-Hz stimulation, and Ca2+ transient alternans with shortening cycle length. Isoproterenol shortened APD80 in sham-operated and TAC-only hearts, shortening CaTD80 and Decay30-90 but sparing TTP100 and Ca2+ transient alternans in all groups. All groups showed similar APD80, and TAC-only hearts showed greater CaTD80, heterogeneities following isoproterenol challenge. Empagliflozin abolished or reduced ventricular tachycardia and premature ventricular contractions and associated re-entrant conduction patterns, in isoproterenol-challenged TAC-operated hearts following successive burst pacing episodes. Empagliflozin thus rescues TAC-induced ventricular hypertrophy and systolic functional, Ca2+ homeostatic, and pro-arrhythmogenic changes in intact hearts.


Subject(s)
Benzhydryl Compounds , Calcium , Glucosides , Homeostasis , Animals , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Mice , Calcium/metabolism , Homeostasis/drug effects , Male , Action Potentials/drug effects , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium-Calcium Exchanger/metabolism , Aorta/drug effects , Aorta/metabolism , Aorta/surgery , Mice, Inbred C57BL , Isoproterenol/pharmacology , Disease Models, Animal
5.
ASN Neuro ; 16(1): 2371160, 2024.
Article in English | MEDLINE | ID: mdl-39024573

ABSTRACT

Promising new pharmacological strategies for the enhancement of cognition target either nicotinic acetylcholine receptors (nAChR) or N-methyl-D-aspartate receptors (NMDAR). There is also an increasing interest in low-dose combination therapies co-targeting the above neurotransmitter systems to reach greater efficacy over the monotreatments and to reduce possible side effects of high-dose monotreatments. In the present study, we assessed modulatory effects of the α7 nAChR-selective agonist PHA-543613 (PHA), a novel α7 nAChR positive allosteric modulator compound (CompoundX) and the NMDAR antagonist memantine on the in vivo firing activity of CA1 pyramidal neurons in the rat hippocampus. Three different test conditions were applied: spontaneous firing activity, NMDA-evoked firing activity and ACh-evoked firing activity. Results showed that high but not low doses of memantine decreased NMDA-evoked firing activity, and low doses increased the spontaneous and ACh-evoked firing activity. Systemically applied PHA robustly potentiated ACh-evoked firing activity with having no effect on NMDA-evoked activity. In addition, CompoundX increased both NMDA- and ACh-evoked firing activity, having no effects on spontaneous firing of the neurons. A combination of low doses of memantine and PHA increased firing activity in all test conditions and similar effects were observed with memantine and CompoundX but without spontaneous firing activity increasing effects. Our present results demonstrate that α7 nAChR agents beneficially interact with Alzheimer's disease medication memantine. Moreover, positive allosteric modulators potentiate memantine effects on the right time and the right place without affecting spontaneous firing activity. All these data confirm previous behavioral evidence for the viability of combination therapies for cognitive enhancement.


Subject(s)
Hippocampus , Memantine , alpha7 Nicotinic Acetylcholine Receptor , Animals , Memantine/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Hippocampus/drug effects , Male , Rats , Neurons/drug effects , Neurons/physiology , Action Potentials/drug effects , Action Potentials/physiology , Dose-Response Relationship, Drug , Cognition/drug effects , Cognition/physiology , Excitatory Amino Acid Antagonists/pharmacology , Nootropic Agents/pharmacology , Rats, Wistar , Ligands , Nicotinic Agonists/pharmacology
6.
Cereb Cortex ; 34(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39042031

ABSTRACT

Interhemispheric inhibition of the homotopic motor cortex is believed to be effective for accurate unilateral motor function. However, the cellular mechanisms underlying interhemispheric inhibition during unilateral motor behavior remain unclear. Furthermore, the impact of the neuromodulator acetylcholine on interhemispheric inhibition and the associated cellular mechanisms are not well understood. To address this knowledge gap, we conducted recordings of neuronal activity from the bilateral motor cortex of mice during the paw-reaching task. Subsequently, we analyzed interhemispheric spike correlation at the cell-pair level, classifying putative cell types to explore the underlying cellular circuitry mechanisms of interhemispheric inhibition. We found a cell-type pair-specific enhancement of the interhemispheric spike correlation when the mice were engaged in the reaching task. We also found that the interhemispheric spike correlation was modulated by pharmacological acetylcholine manipulation. The local field responses to contralateral excitation differed along the cortical depths, and muscarinic receptor antagonism enhanced the inhibitory component of the field response in deep layers. The muscarinic subtype M2 receptor is predominantly expressed in deep cortical neurons, including GABAergic interneurons. These results suggest that GABAergic interneurons expressing muscarinic receptors in deep layers mediate the neuromodulation of interhemispheric inhibition in the homotopic motor cortex.


Subject(s)
Acetylcholine , Motor Cortex , Neural Inhibition , Animals , Motor Cortex/physiology , Motor Cortex/drug effects , Acetylcholine/metabolism , Mice , Male , Neural Inhibition/physiology , Neural Inhibition/drug effects , Functional Laterality/physiology , Mice, Inbred C57BL , Interneurons/physiology , Interneurons/drug effects , Muscarinic Antagonists/pharmacology , Receptor, Muscarinic M2/antagonists & inhibitors , Receptor, Muscarinic M2/metabolism , GABAergic Neurons/physiology , GABAergic Neurons/drug effects , Action Potentials/physiology , Action Potentials/drug effects
7.
Cardiovasc Diabetol ; 23(1): 252, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010053

ABSTRACT

Sodium-glucose cotransporter type 2 inhibitors (SGLT2i) were initially recommended as oral anti-diabetic drugs to treat type 2 diabetes (T2D), by inhibiting SGLT2 in proximal tubule and reduce renal reabsorption of sodium and glucose. While many clinical trials demonstrated the tremendous potential of SGLT2i for cardiovascular diseases. 2022 AHA/ACC/HFSA guideline first emphasized that SGLT2i were the only drug class that can cover the entire management of heart failure (HF) from prevention to treatment. Subsequently, the antiarrhythmic properties of SGLT2i have also attracted attention. Although there are currently no prospective studies specifically on the anti-arrhythmic effects of SGLT2i. We provide clues from clinical and fundamental researches to identify its antiarrhythmic effects, reviewing the evidences and mechanism for the SGLT2i antiarrhythmic effects and establishing a novel paradigm involving intracellular sodium, metabolism and autophagy to investigate the potential mechanisms of SGLT2i in mitigating arrhythmias.


Subject(s)
Anti-Arrhythmia Agents , Arrhythmias, Cardiac , Diabetes Mellitus, Type 2 , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Humans , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/diagnosis , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Arrhythmias, Cardiac/metabolism , Treatment Outcome , Heart Rate/drug effects , Autophagy/drug effects , Sodium-Glucose Transporter 2/metabolism , Action Potentials/drug effects , Sodium/metabolism
8.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-39079743

ABSTRACT

Brain activity implies the orchestrated functioning of interconnected brain regions. Typical in vitro models aim to mimic the brain using single human pluripotent stem cell-derived neuronal networks. However, the field is constantly evolving to model brain functions more accurately through the use of new paradigms, e.g., brain-on-a-chip models with compartmentalized structures and integrated sensors. These methods create novel data requiring more complex analysis approaches. The previously introduced circular tripartite network concept models the connectivity between spatially diverse neuronal structures. The model consists of a microfluidic device allowing axonal connectivity between separated neuronal networks with an embedded microelectrode array to record both local and global electrophysiological activity patterns in the closed circuitry. The existing tools are suboptimal for the analysis of the data produced with this model. Here, we introduce advanced tools for synchronization and functional connectivity assessment. We used our custom-designed analysis to assess the interrelations between the kainic acid (KA)-exposed proximal compartment and its nonexposed distal neighbors before and after KA. Novel multilevel circuitry bursting patterns were detected and analyzed in parallel with the inter- and intracompartmental functional connectivity. The effect of KA on the proximal compartment was captured, and the spread of this effect to the nonexposed distal compartments was revealed. KA induced divergent changes in bursting behaviors, which may be explained by distinct baseline activity and varied intra- and intercompartmental connectivity strengths. The circular tripartite network concept combined with our developed analysis advances importantly both face and construct validity in modeling human epilepsy in vitro.


Subject(s)
Kainic Acid , Nerve Net , Kainic Acid/pharmacology , Nerve Net/drug effects , Nerve Net/physiology , Models, Neurological , Animals , Excitatory Amino Acid Agonists/pharmacology , Humans , Neurons/drug effects , Neurons/physiology , Action Potentials/drug effects , Action Potentials/physiology , Lab-On-A-Chip Devices
9.
Sci Rep ; 14(1): 17360, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075089

ABSTRACT

Prostaglandin E2 (PGE2) is a major contributor to inflammatory pain hyperalgesia, however, the extent to which it modulates the activity of nociceptive axons is incompletely understood. We developed and characterized a microfluidic cell culture model to investigate sensitisation of the axons of dorsal root ganglia neurons. We show that application of PGE2 to fluidically isolated axons leads to sensitisation of their responses to depolarising stimuli. Interestingly the application of PGE2 to the DRG axons elicited a direct and persistent spiking activity propagated to the soma. Both the persistent activity and the membrane depolarisation in the axons are abolished by the EP4 receptor inhibitor and a blocker of cAMP synthesis. Further investigated into the mechanisms of the spiking activity showed that the PGE2 evoked depolarisation was inhibited by Nav1.8 sodium channel blockers but was refractory to the application of TTX or zatebradine. Interestingly, the depolarisation of axons was blocked by blocking ANO1 channels with T16Ainh-A01. We further show that PGE2-elicited axonal responses are altered by the changes in chloride gradient within the axons following treatment with bumetanide a Na-K-2Cl cotransporter NKCC1 inhibitor, but not by VU01240551 an inhibitor of potassium-chloride transporter KCC2. Our data demonstrate a novel role for PGE2/EP4/cAMP pathway which culminates in a sustained depolarisation of sensory axons mediated by a chloride current through ANO1 channels. Therefore, using a microfluidic culture model, we provide evidence for a potential dual function of PGE2 in inflammatory pain: it sensitises depolarisation-evoked responses in nociceptive axons and directly triggers action potentials by activating ANO1 and Nav1.8 channels.


Subject(s)
Anoctamin-1 , Axons , Dinoprostone , Ganglia, Spinal , NAV1.8 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Animals , Dinoprostone/pharmacology , Dinoprostone/metabolism , Axons/metabolism , Axons/drug effects , Axons/physiology , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Rats , Anoctamin-1/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Action Potentials/drug effects , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Rats, Sprague-Dawley , Cells, Cultured , Solute Carrier Family 12, Member 2/metabolism , Cyclic AMP/metabolism
10.
Commun Biol ; 7(1): 893, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075164

ABSTRACT

Maintenance of autonomic homeostasis is continuously calibrated by sensory fibers of the vagus nerve and sympathetic chain that convey compound action potentials (CAPs) to the central nervous system. Lipopolysaccharide (LPS) intravenous challenge reliably elicits a robust inflammatory response that can resemble systemic inflammation and acute endotoxemia. Here, we administered LPS intravenously in nine healthy subjects while recording ventral cervical magnetoneurography (vcMNG)-derived CAPs at the rostral Right Nodose Ganglion (RNG) and the caudal Right Carotid Artery (RCA) with optically pumped magnetometers (OPM). We observed vcMNG RNG and RCA neural firing rates that tracked changes in TNF-α levels in the systemic circulation. Further, endotype subgroups based on high and low IL-6 responders segregate RNG CAP frequency (at 30-120 min) and based on high and low IL-10 response discriminate RCA CAP frequency (at 0-30 min). These vcMNG tools may enhance understanding and management of the neuroimmune axis that can guide personalized treatment based on an individual's distinct endophenotype.


Subject(s)
Inflammation , Lipopolysaccharides , Humans , Male , Female , Inflammation/chemically induced , Adult , Action Potentials/drug effects , Young Adult , Carotid Arteries , Magnetometry/methods
11.
Curr Protoc ; 4(7): e1101, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38980221

ABSTRACT

Cardiovascular diseases have emerged as one of the leading causes of human mortality, but the discovery of new drugs has been hindered by the absence of suitable in vitro platforms. In recent decades, continuously refined protocols for differentiating human induced pluripotent stem cells (hiPSCs) into hiPSC-derived cardiomyocytes (hiPSC-CMs) have significantly advanced disease modeling and drug screening; however, this has led to an increasing need to monitor the function of hiPSC-CMs. The precise regulation of action potentials (APs) and intracellular calcium (Ca2+) transients is critical for proper excitation-contraction coupling and cardiomyocyte function. These important parameters are usually adversely affected in cardiovascular diseases or under cardiotoxic conditions and can be measured using optical imaging-based techniques. However, this procedure is complex and technologically challenging. We have adapted the IonOptix system to simultaneously measure APs and Ca2+ transients in hiPSC-CMs loaded with the fluorescent dyes FluoVolt and Rhod 2, respectively. This system serves as a powerful high-throughput platform to facilitate the discovery of new compounds to treat cardiovascular diseases with the cellular phenotypes of abnormal APs and Ca2+ handling. Here, we present a comprehensive protocol for hiPSC-CM preparation, device setup, optical imaging, and data analysis. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Maintenance and seeding of hiPSC-CMs Basic Protocol 2: Simultaneous detection of action potentials and Ca2+ transients in hiPSC-CMs.


Subject(s)
Action Potentials , Calcium , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Optical Imaging , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Calcium/metabolism , Optical Imaging/methods , Cell Differentiation/drug effects
12.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38928302

ABSTRACT

An accumulation of reactive oxygen species (ROS) in cardiomyocytes can induce pro-arrhythmogenic late Na+ currents by removing the inactivation of voltage-gated Na+ channels including the tetrodotoxin (TTX)-resistant cardiac α-subunit Nav1.5 as well as TTX-sensitive α-subunits like Nav1.2 and Nav1.3. Here, we explored oxidant-induced late Na+ currents in mouse cardiomyocytes and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as well as in HEK 293 cells expressing Nav1.2, Nav1.3, or Nav1.5. Na+ currents in mouse cardiomyocytes and hiPSC-CMs treated with the oxidant chloramine T (ChT) developed a moderate reduction in peak current amplitudes accompanied by large late Na+ currents. While ChT induced a strong reduction in peak current amplitudes but only small persistent currents on Nav1.5, both Nav1.2 and Nav1.3 produced increased peak current amplitudes and large persistent currents following oxidation. TTX (300 nM) blocked ChT-induced late Na+ currents significantly stronger as compared to peak Na+ currents in both mouse cardiomyocytes and hiPSC-CMs. Similar differences between Nav1.2, Nav1.3, and Nav1.5 regarding ROS sensitivity were also evident when oxidation was induced with UVA-light (380 nm) or the cysteine-selective oxidant nitroxyl (HNO). To conclude, our data on TTX-sensitive Na+ channels expressed in cardiomyocytes may be relevant for the generation of late Na+ currents following oxidative stress.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Oxidation-Reduction , Tetrodotoxin , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Humans , Animals , Tetrodotoxin/pharmacology , Mice , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , HEK293 Cells , Chloramines/pharmacology , Reactive Oxygen Species/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium/metabolism , Action Potentials/drug effects , Tosyl Compounds
13.
Cardiovasc Diabetol ; 23(1): 221, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926835

ABSTRACT

BACKGROUND: The incidence of myocardial infarction (MI) and sudden cardiac death (SCD) is significantly higher in individuals with Type 2 Diabetes Mellitus (T2DM) than in the general population. Strategies for the prevention of fatal arrhythmias are often insufficient, highlighting the need for additional non-invasive diagnostic tools. The T-wave heterogeneity (TWH) index measures variations in ventricular repolarization and has emerged as a promising predictor for severe ventricular arrhythmias. Although the EMPA-REG trial reported reduced cardiovascular mortality with empagliflozin, the underlying mechanisms remain unclear. This study investigates the potential of empagliflozin in mitigating cardiac electrical instability in patients with T2DM and coronary heart disease (CHD) by examining changes in TWH. METHODS: Participants were adult outpatients with T2DM and CHD who exhibited TWH > 80 µV at baseline. They received a 25 mg daily dose of empagliflozin and were evaluated clinically including electrocardiogram (ECG) measurements at baseline and after 4 weeks. TWH was computed from leads V4, V5, and V6 using a validated technique. The primary study outcome was a significant (p < 0.05) change in TWH following empagliflozin administration. RESULTS: An initial review of 6,000 medical records pinpointed 800 patients for TWH evaluation. Of these, 412 exhibited TWH above 80 µV, with 97 completing clinical assessments and 90 meeting the criteria for high cardiovascular risk enrollment. Empagliflozin adherence exceeded 80%, resulting in notable reductions in blood pressure without affecting heart rate. Side effects were generally mild, with 13.3% experiencing Level 1 hypoglycemia, alongside infrequent urinary and genital infections. The treatment consistently reduced mean TWH from 116 to 103 µV (p = 0.01). CONCLUSIONS: The EMPATHY-HEART trial preliminarily suggests that empagliflozin decreases heterogeneity in ventricular repolarization among patients with T2DM and CHD. This reduction in TWH may provide insight into the mechanism behind the decreased cardiovascular mortality observed in previous trials, potentially offering a therapeutic pathway to mitigate the risk of severe arrhythmias in this population. TRIAL REGISTRATION: NCT: 04117763.


Subject(s)
Benzhydryl Compounds , Diabetes Mellitus, Type 2 , Glucosides , Sodium-Glucose Transporter 2 Inhibitors , Humans , Benzhydryl Compounds/therapeutic use , Benzhydryl Compounds/adverse effects , Glucosides/therapeutic use , Glucosides/adverse effects , Male , Female , Middle Aged , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Aged , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Treatment Outcome , Time Factors , Action Potentials/drug effects , Arrhythmias, Cardiac/mortality , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/physiopathology , Heart Rate/drug effects , Coronary Disease/mortality , Coronary Disease/physiopathology , Coronary Disease/drug therapy , Coronary Disease/diagnosis , Electrocardiography , Risk Factors
14.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38834302

ABSTRACT

Linked rhythmic behaviors, such as respiration/locomotion or swallowing/chewing, often require coordination for proper function. Despite its prevalence, the cellular mechanisms controlling coordination of the underlying neural networks remain undetermined in most systems. We use the stomatogastric nervous system of the crab Cancer borealis to investigate mechanisms of internetwork coordination, due to its small, well-characterized feeding-related networks (gastric mill [chewing, ∼0.1 Hz]; pyloric [filtering food, ∼1 Hz]). Here, we investigate coordination between these networks during the Gly1-SIFamide neuropeptide modulatory state. Gly1-SIFamide activates a unique triphasic gastric mill rhythm in which the typically pyloric-only LPG neuron generates dual pyloric-plus gastric mill-timed oscillations. Additionally, the pyloric rhythm exhibits shorter cycles during gastric mill rhythm-timed LPG bursts, and longer cycles during IC, or IC plus LG gastric mill neuron bursts. Photoinactivation revealed that LPG is necessary to shorten pyloric cycle period, likely through its rectified electrical coupling to pyloric pacemaker neurons. Hyperpolarizing current injections demonstrated that although LG bursting enables IC bursts, only gastric mill rhythm bursts in IC are necessary to prolong the pyloric cycle period. Surprisingly, LPG photoinactivation also eliminated prolonged pyloric cycles, without changing IC firing frequency or gastric mill burst duration, suggesting that pyloric cycles are prolonged via IC synaptic inhibition of LPG, which indirectly slows the pyloric pacemakers via electrical coupling. Thus, the same dual-network neuron directly conveys excitation from its endogenous bursting and indirectly funnels synaptic inhibition to enable one network to alternately decrease and increase the cycle period of a related network.


Subject(s)
Brachyura , Ganglia, Invertebrate , Neurons , Neuropeptides , Animals , Brachyura/physiology , Neuropeptides/pharmacology , Neuropeptides/metabolism , Neurons/physiology , Neurons/drug effects , Ganglia, Invertebrate/physiology , Ganglia, Invertebrate/drug effects , Action Potentials/physiology , Action Potentials/drug effects , Nerve Net/physiology , Nerve Net/drug effects , Male , Feeding Behavior/physiology , Feeding Behavior/drug effects , Pylorus/physiology , Pylorus/drug effects , Periodicity
15.
Cardiovasc Toxicol ; 24(7): 656-666, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38851664

ABSTRACT

Antiviral therapies for treatment of COVID-19 may be associated with significant proarrhythmic potential. In the present study, the potential cardiotoxic side effects of these therapies were evaluated using a Langendorff model of the isolated rabbit heart. 51 hearts of female rabbits were retrogradely perfused, employing a Langendorff-setup. Eight catheters were placed endo- and epicardially to perform an electrophysiology study, thus obtaining cycle length-dependent action potential duration at 90% of repolarization (APD90), QT intervals and dispersion of repolarization. After generating baseline data, the hearts were assigned to four groups: In group 1 (HXC), hearts were treated with 1 µM hydroxychloroquine. Thereafter, 3 µM hydroxychloroquine were infused additionally. Group 2 (HXC + AZI) was perfused with 3 µM hydroxychloroquine followed by 150 µM azithromycin. In group 3 (LOP) the hearts were perfused with 3 µM lopinavir followed by 5 µM and 10 µM lopinavir. Group 4 (REM) was perfused with 1 µM remdesivir followed by 5 µM and 10 µM remdesivir. Hydroxychloroquine- and azithromycin-based therapies have a significant proarrhythmic potential mediated by action potential prolongation and an increase in dispersion. Lopinavir and remdesivir showed overall significantly less pronounced changes in electrophysiology. In accordance with the reported bradycardic events under remdesivir, it significantly reduced the rate of the ventricular escape rhythm.


Subject(s)
Action Potentials , Antiviral Agents , Isolated Heart Preparation , Animals , Rabbits , Female , Antiviral Agents/pharmacology , Antiviral Agents/toxicity , Action Potentials/drug effects , COVID-19 Drug Treatment , Hydroxychloroquine/toxicity , Hydroxychloroquine/pharmacology , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Cardiotoxicity , Alanine/analogs & derivatives , Alanine/pharmacology , Heart Rate/drug effects , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/toxicity , Adenosine Monophosphate/pharmacology , Heart/drug effects
16.
Cell Mol Life Sci ; 81(1): 268, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884814

ABSTRACT

It has been recently established that GPR158, a class C orphan G protein-coupled receptor, serves as a metabotropic glycine receptor. GPR158 is highly expressed in the nucleus accumbens (NAc), a major input structure of the basal ganglia that integrates information from cortical and subcortical structures to mediate goal-directed behaviors. However, whether glycine modulates neuronal activity in the NAc through GPR158 activation has not been investigated yet. Using whole-cell patch-clamp recordings, we found that glycine-dependent activation of GPR158 increased the firing rate of NAc medium spiny neurons (MSNs) while it failed to significantly affect the excitability of cholinergic interneurons (CIN). In MSNs GPR158 activation reduced the latency to fire, increased the action potential half-width, and reduced action potential afterhyperpolarization, effects that are all consistent with negative modulation of potassium M-currents, that in the central nervous system are mainly carried out by Kv7/KCNQ-channels. Indeed, we found that the GPR158-induced increase in MSN excitability was associated with decreased M-current amplitude, and selective pharmacological inhibition of the M-current mimicked and occluded the effects of GPR158 activation. In addition, when the protein kinase A (PKA) or extracellular signal-regulated kinase (ERK) signaling was pharmacologically blocked, modulation of MSN excitability by GPR158 activation was suppressed. Moreover, GPR158 activation increased the phosphorylation of ERK and Kv7.2 serine residues. Collectively, our findings suggest that GPR158/PKA/ERK signaling controls MSN excitability via Kv7.2 modulation. Glycine-dependent activation of GPR158 may significantly affect MSN firing in vivo, thus potentially mediating specific aspects of goal-induced behaviors.


Subject(s)
Action Potentials , Glycine , Neurons , Nucleus Accumbens , Receptors, G-Protein-Coupled , Animals , Glycine/pharmacology , Glycine/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/cytology , Neurons/metabolism , Neurons/drug effects , Receptors, G-Protein-Coupled/metabolism , Male , Action Potentials/drug effects , Mice , Mice, Inbred C57BL , Receptors, Glycine/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Medium Spiny Neurons
17.
Eur J Pharmacol ; 977: 176675, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38825303

ABSTRACT

BACKGROUND: Ibrutinib, a Bruton's tyrosine kinase inhibitor used in cancer therapy, exerts ventricular proarrhythmic effects; however, the underlying mechanisms remain unclear. Excitation-contraction coupling (E-C) disorders are pivotal for the genesis of ventricular arrhythmias (VAs), which arise mainly from the right ventricular outflow tract (RVOT). In this study, we aimed to comprehensively investigate whether ibrutinib regulates the electromechanical activities of the RVOT, leading to enhanced arrhythmogenesis, and explore the underlying mechanisms. METHODS: We utilized conventional microelectrodes to synchronously record electrical and mechanical responses in rabbit RVOT tissue preparations before and after treatment with ibrutinib (10, 50, and 100 nM) and investigated their electromechanical interactions and arrhythmogenesis during programmed electrical stimulation. The fluorometric ratio technique was used to measure intracellular calcium concentration in isolated RVOT myocytes. RESULTS: Ibrutinib (10-100 nM) shortened the action potential duration. Ibrutinib at 100 nM significantly increased pacing-induced ventricular tachycardia (VT) (from 0% to 62.5%, n = 8, p = 0.025). Comparisons between pacing-induced VT and non-VT episodes demonstrated that VT episodes had a greater increase in contractility than that of non-VT episodes (402.1 ± 41.4% vs. 232.4 ± 29.2%, p = 0.003). The pretreatment of ranolazine (10 µM, a late sodium current blocker) prevented the occurrence of ibrutinib-induced VAs. Ibrutinib (100 nM) increased late sodium current, reduced intracellular calcium transients, and enhanced calcium leakage in RVOT myocytes. CONCLUSION: Ibrutinib increased the risk of VAs in the RVOT due to dysregulated electromechanical responses, which can be attenuated by ranolazine or apamin.


Subject(s)
Action Potentials , Adenine , Agammaglobulinaemia Tyrosine Kinase , Piperidines , Protein Kinase Inhibitors , Animals , Piperidines/pharmacology , Rabbits , Adenine/analogs & derivatives , Adenine/pharmacology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/adverse effects , Action Potentials/drug effects , Pyrimidines/pharmacology , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Male , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Calcium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Tachycardia, Ventricular/physiopathology , Pyrazoles/pharmacology , Excitation Contraction Coupling/drug effects
18.
Bull Exp Biol Med ; 176(6): 761-766, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38896318

ABSTRACT

A number of pharmacological drugs have side effects that contribute to the occurrence of atrial fibrillation, the most common type of cardiac rhythm disorders. The clinical use of antihistamines is widespread; however, information regarding their anti- and/or proarrhythmic effects is contradictory. In this work, we studied the effects and mechanisms of the potential proarrhythmic action of the first-generation antihistamine chloropyramine (Suprastin) in the atrial myocardium and pulmonary vein (PV) myocardial tissue. In PV, chloropyramine caused depolarization of the resting potential and led to reduction of excitation wave conduction. These effects are likely due to suppression of the inward rectifier potassium current (IK1). In presence of epinephrine, chloropyramine induced spontaneous automaticity in the PV and could not be suppressed by atrial pacing. Chloropyramine change functional characteristics of PV and contribute to occurrence of atrial fibrillation. It should be noted that chloropyramine does not provoke atrial tachyarrhythmias, but create conditions for their occurrence during physical exercise and sympathetic stimulation.


Subject(s)
Atrial Fibrillation , Pulmonary Veins , Pulmonary Veins/drug effects , Pulmonary Veins/physiopathology , Animals , Atrial Fibrillation/physiopathology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/chemically induced , Heart Atria/drug effects , Heart Atria/physiopathology , Chlorpheniramine/pharmacology , Epinephrine/pharmacology , Histamine H1 Antagonists/pharmacology , Myocardium/metabolism , Myocardium/pathology , Male , Action Potentials/drug effects , Heart Conduction System/drug effects , Heart Conduction System/physiopathology
19.
J Neurosci ; 44(30)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38937101

ABSTRACT

Many neurons including vasopressin (VP) magnocellular neurosecretory cells (MNCs) of the hypothalamic supraoptic nucleus (SON) generate afterhyperpolarizations (AHPs) during spiking to slow firing, a phenomenon known as spike frequency adaptation. The AHP is underlain by Ca2+-activated K+ currents, and while slow component (sAHP) features are well described, its mechanism remains poorly understood. Previous work demonstrated that Ca2+ influx through N-type Ca2+ channels is a primary source of sAHP activation in SON oxytocin neurons, but no obvious channel coupling was described for VP neurons. Given this, we tested the possibility of an intracellular source of sAHP activation, namely, the Ca2+-handling organelles endoplasmic reticulum (ER) and mitochondria in male and female Wistar rats. We demonstrate that ER Ca2+ depletion greatly inhibits sAHPs without a corresponding decrease in Ca2+ signal. Caffeine sensitized AHP activation by Ca2+ In contrast to ER, disabling mitochondria with CCCP or blocking mitochondria Ca2+ uniporters (MCUs) enhanced sAHP amplitude and duration, implicating mitochondria as a vital buffer for sAHP-activating Ca2+ Block of mitochondria Na+-dependent Ca2+ release via triphenylphosphonium (TPP+) failed to affect sAHPs, indicating that mitochondria Ca2+ does not contribute to sAHP activation. Together, our results suggests that ER Ca2+-induced Ca2+ release activates sAHPs and mitochondria shape the spatiotemporal trajectory of the sAHP via Ca2+ buffering in VP neurons. Overall, this implicates organelle Ca2+, and specifically ER-mitochondria-associated membrane contacts, as an important site of Ca2+ microdomain activity that regulates sAHP signaling pathways. Thus, this site plays a major role in influencing VP firing activity and systemic hormonal release.


Subject(s)
Calcium , Endoplasmic Reticulum , Mitochondria , Neurons , Rats, Wistar , Supraoptic Nucleus , Vasopressins , Animals , Rats , Vasopressins/metabolism , Male , Female , Neurons/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Calcium/metabolism , Supraoptic Nucleus/metabolism , Action Potentials/physiology , Action Potentials/drug effects , Calcium Signaling/physiology
20.
Neuropharmacology ; 257: 110030, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38851531

ABSTRACT

Administration or consumption of classic psychedelics (CPs) leads to profound changes in experience which are often described as highly novel and meaningful. They have shown substantial promise in treating depressive symptoms and may be therapeutic in other situations. Although research suggests that the therapeutic response is correlated with the intensity of the experience, the neural circuit basis for the alterations in experience caused by CPs requires further study. The medial prefrontal cortex (mPFC), where CPs have been shown to induce rapid, 5-HT2A receptor-dependent structural and neurophysiological changes, is believed to be a key site of action. To investigate the acute neural circuit changes induced by CPs, we recorded single neurons and local field potentials in the mPFC of freely behaving male mice after administration of the 5-HT2A/2C receptor-selective CP, 2,5-Dimethoxy-4-iodoamphetamine (DOI). We segregated recordings into active and rest periods in order to examine cortical activity during desynchronized (active) and synchronized (rest) states. We found that DOI induced a robust decrease in low frequency power when animals were at rest, attenuating the usual synchronization that occurs during less active behavioral states. DOI also increased broadband gamma power and suppressed activity in fast-spiking neurons in both active and rest periods. Together, these results suggest that the CP DOI induces persistent desynchronization in mPFC, including during rest when mPFC typically exhibits more synchronized activity. This shift in cortical dynamics may in part underlie the longer-lasting effects of CPs on plasticity, and may be critical to their therapeutic properties.


Subject(s)
Amphetamines , Hallucinogens , Prefrontal Cortex , Animals , Male , Hallucinogens/pharmacology , Hallucinogens/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Mice , Amphetamines/pharmacology , Amphetamines/administration & dosage , Neurons/drug effects , Neurons/physiology , Mice, Inbred C57BL , Behavior, Animal/drug effects , Action Potentials/drug effects , Action Potentials/physiology
SELECTION OF CITATIONS
SEARCH DETAIL