Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
Cell Tissue Res ; 391(2): 375-391, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36422735

ABSTRACT

Bepridil is a commonly used medication for arrhythmia and heart failure. It primarily exerts hemodynamic effects by inhibiting Na+/K+ movement and regulating the Na+/Ca2+ exchange. In comparison to other Ca2+ inhibitors, bepridil has a long half-life and a complex pharmacology. Additionally, it is widely used in antiviral research and the treatment of various diseases. However, the toxicity of this compound and its other possible effects on embryonic development are unknown. In this study, we investigated the toxicity of bepridil on rat myocardial H9c2 cells. After treatment with bepridil, the cells became overloaded with Ca2+ and entered a state of cytoplasmic vacuolization and nuclear abnormality. Bepridil treatment resulted in several morphological abnormalities in zebrafish embryo models, including pericardium enlargement, yolk sac swelling, and growth stunting. The hemodynamic effects on fetal development resulted in abnormal cardiovascular circulation and myocardial weakness. After inhibiting the Ca2+ transmembrane, the liver of zebrafish larvae also displayed an ectopic and deficient spatial location. Additionally, the results of the RNA-seq analysis revealed the detailed gene expression profiles and metabolic responses to bepridil treatment in zebrafish embryonic development. Taken together, our study provides an important evaluation of antiarrhythmic agents for clinical use in prenatal heart patients.


Subject(s)
Bepridil , Zebrafish , Animals , Rats , Bepridil/metabolism , Bepridil/pharmacology , Anti-Arrhythmia Agents/metabolism , Anti-Arrhythmia Agents/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/metabolism
2.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33597253

ABSTRACT

Guided by a computational docking analysis, about 30 Food and Drug Administration/European Medicines Agency (FDA/EMA)-approved small-molecule medicines were characterized on their inhibition of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro). Of these small molecules tested, six displayed a concentration that inhibits response by 50% (IC50) value below 100 µM in inhibiting Mpro, and, importantly, three, that is, pimozide, ebastine, and bepridil, are basic molecules that potentiate dual functions by both raising endosomal pH to interfere with SARS-CoV-2 entry into the human cell host and inhibiting Mpro in infected cells. A live virus-based modified microneutralization assay revealed that bepridil possesses significant anti-SARS-CoV-2 activity in both Vero E6 and A459/ACE2 cells in a dose-dependent manner with low micromolar effective concentration, 50% (EC50) values. Therefore, the current study urges serious considerations of using bepridil in COVID-19 clinical tests.


Subject(s)
Antiviral Agents/pharmacology , Bepridil/pharmacology , Drug Discovery , SARS-CoV-2/drug effects , A549 Cells , Animals , Chlorocebus aethiops , Humans , Molecular Docking Simulation , Molecular Structure , Small Molecule Libraries , Vero Cells
3.
J Pharmacol Sci ; 143(1): 56-59, 2020 May.
Article in English | MEDLINE | ID: mdl-32144028

ABSTRACT

Since microminipig is becoming attractive model for various cardiac electropharmacological applications, which may meet consideration of 3Rs. We characterized microminipigs by analyzing how multi-ionic channel inhibitor bepridil may affect their in situ hearts in comparison with dogs. Bepridil in doses of 0.3 and 3.0 mg/kg were intravenously administered over 10 min under halothane anesthesia (n = 4). Microminipigs may be less sensitive for ICaT inhibition of bepridil, whereas they are more responsive to INa, IKr and IKs suppression than dogs. This information would help predict cardiovascular effects of a drug in patients with the remodeled hearts having similar electrophysiological profile to microminipigs.


Subject(s)
Animals, Laboratory , Bepridil/pharmacology , Calcium Channel Blockers/pharmacology , Cardiovascular System/drug effects , Disease Models, Animal , Swine, Miniature , Animals , Bepridil/administration & dosage , Calcium Channel Blockers/administration & dosage , Dogs , Dose-Response Relationship, Drug , Infusions, Intravenous , Swine
4.
Int Heart J ; 61(2): 338-346, 2020 Mar 28.
Article in English | MEDLINE | ID: mdl-32173709

ABSTRACT

Sympathetic nerve activity has arrhythmogenic potential for ventricular arrhythmias associated with structural heart diseases. However, a sufficient amount of beta-blockers occasionally cannot be prescribed in some patients.An experimental study was performed to clarify the therapeutic effects of bepridil, a multiple ionic current inhibitor that does not affect beta-adrenergic receptors, for premature beats occurring during enhanced sympathetic nerve activity. Cardio-sympathetic nerve activity was augmented via stellate-ganglion (SG) stimulation in a canine model (n = 8), and the arrhythmogenic potential and anti-arrhythmic effects of bepridil (2 and 4 mg/kg intravenously) were assessed. For safe use, vagal-stimulation-induced slow HR and programmed electrical stimulation were applied to evaluate possible pro-arrhythmic effects of the drug. Heart rate variability (HRV) indexes were used to estimate cardio-autonomic nerve activity.Either side of the SG-stimulation increased BP and HR. Premature beats were induced in 10/16 SG-stimulations and it was more frequent in left (8/8) rather than right stimulation (2/8). Following 2 mg/kg drug administration, premature beats were still inducible in 8/16 stimulations (7/8 in left and 1/8 in right), but burden of the premature beats decreased from 87.1 ± 46.8 to 62.1 ± 42.6 beats. After 4 mg/kg administration, premature beats were inducible in one SG-stimulation. Proarrhythmic effects were not observed in all experiments. Steady-state HRV indexes and percent increases in SG-stimulation-induced BP-elevation and HR-acceleration were similar among the 3 periods (before, 2 and 4 mg/kg of the drug).Bepridil may be an option for ventricular arrhythmias developed during enhanced cardio-sympathetic nerve activity with minimal effect on autonomic nerve responses.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Bepridil/therapeutic use , Ventricular Premature Complexes/drug therapy , Animals , Anti-Arrhythmia Agents/pharmacology , Bepridil/pharmacology , Blood Pressure/drug effects , Dogs , Drug Evaluation, Preclinical , Electric Stimulation , Electrocardiography/drug effects , Heart Rate/drug effects , Stellate Ganglion
5.
J Pharmacol Sci ; 141(4): 153-159, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31757741

ABSTRACT

Experimental evidence regarding the risk of proarrhythmic potential of acehytisine is limited. We assessed its electropharmacological effect together with proarrhythmic potential at intravenous doses of 4 and 10 mg/kg (n = 6) using isoflurane-anesthetized guinea pigs in comparison with that of bepridil at 1 and 3 mg/kg, intravenously (n = 6). Acehytisine at therapeutic dose (4 mg/kg) decreased the heart rate, prolonged P wave duration, QRS width, QT interval, QTc, MAP90(sinus), MAP90(CL300) and MAP90(CL250). At supratherapeutic dose (10 mg/kg), it prolonged the PR interval besides enhancing the changes induced by the therapeutic dose. Quantitative assessment showed that peak changes in P wave duration by acehytisine at 10 mg/kg were 1.7 times longer than bepridil, and in MAP90(sinus), MAP90(CL300) and MAP90(CL250) by acehytisine were 1.9, 1.5 and 1.5 times shorter than bepridil, respectively. Importantly, qualitative assessment indicated that bepridil increased beat-to-beat variability and J-Tpeakc in a dose-related manner, confirming a higher proarrhythmic risk, whereas such dose-related responses were not observed in acehytisine, suggesting a lower proarrhythmic risk. These results suggest that acehytisine exhibits favorable pharmacological characters, i.e. potent atrial inhibition and lower proarrhythmic toxicity compared with bepridil, being a promising candidate for the treatment of paroxysmal supraventricular tachycardia.


Subject(s)
Anti-Arrhythmia Agents/metabolism , Heart Atria/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Sodium Channel Blockers/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Bepridil/metabolism , Bepridil/pharmacology , Electrocardiography/methods , Guinea Pigs , Heart Rate/drug effects , Heterocyclic Compounds, 4 or More Rings/metabolism , Isoflurane/pharmacology , Male , Sodium Channel Blockers/pharmacology
6.
Br J Pharmacol ; 176(15): 2691-2707, 2019 08.
Article in English | MEDLINE | ID: mdl-31034096

ABSTRACT

BACKGROUND AND PURPOSE: The Na+ /Ca2+ exchanger (NCX) working in either forward or reverse mode participates in maintaining intracellular Ca2+ ([Ca2+ ]i ) homeostasis, which is essential for determining cell fate. Previously, numerous blockers targeting reverse or forward NCX have been developed and studied in ischaemic tissue injury but barely examined in glioblastoma for the purpose of anti-tumour therapy. We assessed the effect of NCX blockers on glioblastoma growth and whether NCX can become a therapeutic target. EXPERIMENTAL APPROACH: Patch-clamp recording, Ca2+ imaging, flow cytometry, and Western blot were used to study the effects of specific and non-specific NCX blockers on cultured glioblastoma cells. In vivo bioluminescent imaging was used to measure effects on grafted glioblastoma. KEY RESULTS: Selectively blocking the reverse NCX with SEA0400, SN-6, and YM-244769 did not affect tumour cell viability. Blocking the forward NCX with bepridil, CB-DMB, or KB-R7943 elevated [Ca2+ ]i and killed glioblastoma cells. Bepridil and CB-DMB caused Ca2+ -dependent cell cycle arrest together with apoptosis, which were all attenuated by a Ca2+ chelator BAPTA-AM. Systemic administration of bepridil inhibited growth of brain-grafted glioblastoma. Bepridil did not appear to have a cytotoxic effect on human astrocytes, which have higher functional expression of NCX than glioblastoma cells. CONCLUSIONS AND IMPLICATIONS: Low expression of the NCX makes glioblastoma cells sensitive to disturbance of [Ca2+ ]i . Interventions designed to block the forward NCX can cause Ca2+ -mediated injury to glioblastoma thus having therapeutic potential. Bepridil could be a lead compound for developing new anti-tumour drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bepridil/pharmacology , Bepridil/therapeutic use , Calcium/metabolism , Glioblastoma/drug therapy , Sodium-Calcium Exchanger/antagonists & inhibitors , Amiloride/analogs & derivatives , Amiloride/pharmacology , Aniline Compounds/pharmacology , Animals , Astrocytes/drug effects , Benzyl Compounds/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glioblastoma/metabolism , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenyl Ethers/pharmacology , Sodium-Calcium Exchanger/physiology , Thiazolidines/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology
7.
Oncol Rep ; 41(4): 2241-2253, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816547

ABSTRACT

Systemic treatment options for soft tissue sarcomas (STSs) have remained unchanged despite the need for novel drug candidates to improve STS outcomes. Drug repurposing involves the application of clinical drugs to different diseases, reducing development time, and cost. It has also become a fast and effective way to identify drug candidates. The present study used a computational method to screen three drug­gene interaction databases for novel drug candidates for the treatment of several common STS histologic subtypes through drug repurposing. STS survival­associated genes were generated by conducting a univariate cox regression analysis using The Cancer Genome Atlas survival data. These genes were then applied to three databases (the Connectivity Map, the Drug Gene Interaction Database and the L1000 Fireworks Display) to identify drug candidates for STS treatment. Additionally, pathway analysis and molecular docking were conducted to evaluate the molecular mechanisms of the candidate drug. Bepridil was identified as a potential candidate for several STS histologic subtype treatments by overlapping the screening results from three drug­gene interaction databases. The pathway analysis with the Kyoto Encyclopedia of Genes and Genomes predicted that Bepridil may target CRK, fibroblast growth factor receptor 4 (FGFR4), laminin subunit ß1 (LAMB1), phosphoinositide­3­kinase regulatory subunit 2 (PIK3R2), WNT5A, cluster of differentiation 47 (CD47), elastase, neutrophil expressed (ELANE), 15­hydroxyprostaglandin dehydrogenase (HPGD) and protein kinase cß (PRKCB) to suppress STS development. Further molecular docking simulation suggested a relatively stable binding selectivity between Bepridil and eight proteins (CRK, FGFR4, LAMB1, PIK3R2, CD47, ELANE, HPGD, and PRKCB). In conclusion, a computational method was used to identify Bepridil as a potential candidate for the treatment of several common STS histologic subtypes. Experimental validation of these in silico results is necessary before clinical translation can occur.


Subject(s)
Antineoplastic Agents/pharmacology , Bepridil/pharmacology , Drug Repositioning/methods , Gene Regulatory Networks/genetics , Sarcoma/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Bepridil/chemistry , Bepridil/therapeutic use , Computational Biology , Databases, Genetic/statistics & numerical data , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Molecular Docking Simulation , Protein Binding , Sarcoma/genetics , Sarcoma/mortality , Sarcoma/pathology , Survival Analysis
8.
Plant Physiol ; 180(1): 480-496, 2019 05.
Article in English | MEDLINE | ID: mdl-30737267

ABSTRACT

Many signal perception mechanisms are connected to Ca2+-based second messenger signaling to modulate specific cellular responses. The well-characterized plant hormone auxin elicits a very rapid Ca2+ signal. However, the cellular targets of auxin-induced Ca2+ are largely unknown. Here, we screened a biologically annotated chemical library for inhibitors of auxin-induced Ca2+ entry in plant cell suspensions to better understand the molecular mechanism of auxin-induced Ca2+ and to explore the physiological relevance of Ca2+ in auxin signal transduction. Using this approach, we defined a set of diverse, small molecules that interfere with auxin-induced Ca2+ entry. Based on annotated biological activities of the hit molecules, we found that auxin-induced Ca2+ signaling is, among others, highly sensitive to disruption of membrane proton gradients and the mammalian Ca2+ channel inhibitor bepridil. Whereas protonophores nonselectively inhibited auxin-induced and osmotic stress-induced Ca2+ signals, bepridil specifically inhibited auxin-induced Ca2+ We found evidence that bepridil severely alters vacuolar morphology and antagonized auxin-induced vacuolar remodeling. Further exploration of this plant-tailored collection of inhibitors will lead to a better understanding of auxin-induced Ca2+ entry and its relevance for auxin responses.


Subject(s)
Arabidopsis/drug effects , Calcium Signaling/drug effects , Indoleacetic Acids/metabolism , Nicotiana/drug effects , Small Molecule Libraries/pharmacology , 2,4-Dichlorophenoxyacetic Acid/pharmacology , Arabidopsis/genetics , Bacterial Proteins/genetics , Bepridil/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling/physiology , Cell Line , Drug Evaluation, Preclinical/methods , Fenamates/pharmacology , Indoleacetic Acids/antagonists & inhibitors , Luminescent Measurements , Luminescent Proteins/genetics , Niclosamide/pharmacology , Plant Cells/drug effects , Plant Cells/metabolism , Plant Growth Regulators/metabolism , Plant Roots/drug effects , Plants, Genetically Modified , Nicotiana/genetics , Vacuoles/drug effects , Vacuoles/metabolism
9.
Anat Sci Int ; 94(2): 163-171, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30353456

ABSTRACT

Nisin is a food preservative produced by Lactococcus lactis subsp. lactis. Previous blood biochemical research revealed that nisin has physiological effects in mammals; although the site of action has yet to be identified, keratinocytes have been proposed as a possible target. In this study, we investigated whether nisin affects keratinocytes by examining the effects on eukaryotic intermediate filaments in HaCaT human keratinocytes. Treatment with 93 µg/ml nisin for 24 h decreased the localization of the intermediate filament proteins cytokeratin (CK)5 and CK17 at the cell periphery, which were distributed in a limited area in a ring- or net-like shape. However, this was not observed upon treatment for 6 h. The results of a serial dilution assay revealed that the effect on CK17 localization depends on the nisin concentration and were observed at ≥47 µg/ml. Moreover, this effect was partially blocked by treatment with the calcium channel blocker bepridil. Thus, despite the long history of nisin as being safe for humans, it has measurable effects on the keratinocyte cytoskeleton. Our findings also indicate that CK5 and CK17 can serve as markers for evaluating the effects of nisin on keratinocytes.


Subject(s)
Food Preservatives/pharmacology , Intermediate Filament Proteins/metabolism , Keratinocytes/metabolism , Nisin/pharmacology , Bepridil/pharmacology , Calcium Channel Blockers/pharmacology , Cells, Cultured , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Humans , Keratin-17/metabolism , Keratin-5/metabolism , Keratinocytes/cytology , Lactococcus lactis/metabolism , Nisin/antagonists & inhibitors , Nisin/biosynthesis
10.
J Gen Physiol ; 151(1): 9-17, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30442775

ABSTRACT

Despite extensive efforts spanning multiple decades, the development of highly effective Ca2+ sensitizers for the heart remains an elusive goal. Existing Ca2+ sensitizers have other targets in addition to cardiac troponin (cTn), which can lead to adverse side effects, such as hypotension or arrhythmias. Thus, there is a need to design Ca2+-sensitizing drugs with higher affinity and selectivity for cTn. Previously, we determined that many compounds based on diphenylamine (DPA) were able to bind to a cTnC-cTnI chimera with moderate affinity (Kd ∼10-120 µM). Of these compounds, 3-chlorodiphenylamine (3-Cl-DPA) bound most tightly (Kd of 10 µM). Here, we investigate 3-Cl-DPA further and find that it increases the Ca2+ sensitivity of force development in skinned cardiac muscle. Using NMR, we show that, like the known Ca2+ sensitizers, trifluoperazine (TFP) and bepridil, 3-Cl-DPA is able to bind to the isolated N-terminal domain (N-domain) of cTnC (Kd of 6 µM). However, while the bulky molecules of TFP and bepridil stabilize the open state of the N-domain of cTnC, the small and flexible 3-Cl-DPA molecule is able to bind without stabilizing this open state. Thus, unlike TFP, which drastically slows the rate of Ca2+ dissociation from the N-domain of isolated cTnC in a dose-dependent manner, 3-Cl-DPA has no effect on the rate of Ca2+ dissociation. On the other hand, the affinity of 3-Cl-DPA for a cTnC-TnI chimera is at least an order of magnitude higher than that of TFP or bepridil, likely because 3-Cl-DPA is less disruptive of cTnI binding to cTnC. Therefore, 3-Cl-DPA has a bigger effect on the rate of Ca2+ dissociation from the entire cTn complex than TFP and bepridil. Our data suggest that 3-Cl-DPA activates the cTn complex via a unique mechanism and could be a suitable scaffold for the development of novel treatments for systolic heart failure.


Subject(s)
Bepridil/pharmacology , Diphenylamine/pharmacology , Heart/drug effects , Trifluoperazine/pharmacology , Troponin C/metabolism , Troponin I/metabolism , Animals , Calcium/metabolism , Female , Humans , Myocardium/metabolism , Rats , Rats, Wistar
11.
J Pharmacol Sci ; 137(4): 372-378, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30126708

ABSTRACT

We examined electrophysiological indices of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) sheets in order to quantitatively estimate Na+, K+ and Ca2+ channel blocking actions of bepridil and amiodarone using microelectrode array system in comparison with that of E-4031. We analyzed the field potential duration, effective refractory period, current threshold and conduction property using a programmed electrical stimulation protocol to obtain the post repolarization refractoriness and coefficient a of the relationship between the pacing cycle length and field potential duration. Electropharmacological profile of each drug was successfully characterized; namely, 1) the changes in the current threshold and conduction property provided basic information of Na+ channel blocking kinetics, 2) the relationship between pacing cycle length and field potential duration reflected drug-induced inhibition of human ether-à-go-go-related gene (hERG) K+ channel, 3) the post repolarization refractoriness indicated the relative contribution of these drugs to Na+ and K+ channel blockade, and 4) L-type Ca2+ channel blocking action was more obvious in the field potential waveform of the hiPSC-CMs sheets than that expected in the electrocardiogram in humans. Thus, this information may help to better utilize the hiPSC-CMs sheets for grasping the properties and net effects of drug-induced Na+, Ca2+ and K+ channel blockade.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents , Bepridil/pharmacology , Calcium Channel Blockers/pharmacology , Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells , Microelectrodes , Myocytes, Cardiac/physiology , Potassium Channel Blockers/pharmacology , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Cells, Cultured , Electrophysiological Phenomena/drug effects , Humans , Piperidines/pharmacology , Pyridines/pharmacology
12.
J Neurochem ; 147(2): 256-274, 2018 10.
Article in English | MEDLINE | ID: mdl-29804308

ABSTRACT

The initial step in the amyloidogenic cascade of amyloid precursor protein (APP) processing is catalyzed by beta-site APP-cleaving enzyme (BACE), and this protease has increased activities in affected areas of Alzheimer's disease brains. We hypothesized that altered APP processing, because of augmented BACE activity, would affect the actions of direct and indirect BACE inhibitors. We therefore compared post-mitotic human neurons (LUHMES) with their BACE-overexpressing counterparts (BLUHMES). Although ß-cleavage of APP was strongly increased in BLUHMES, they produced less full-length and truncated amyloid beta (Aß) than LUHMES. Moreover, low concentrations of BACE inhibitors decreased cellular BACE activity as expected, but increased Aß1-40 levels. Several other approaches to modulate BACE activity led to a similar, apparently paradoxical, behavior. For instance, reduction in intracellular acidification by bepridil increased Aß production in parallel with decreased BACE activity. In contrast to BLUHMES, the respective control cells (LUHMES or BLUHMES with catalytically inactive BACE) showed conventional pharmacological responses. Other non-canonical neurochemical responses (so-called 'rebound effects') are well-documented for the Aß pathway, especially for γ-secretase: a partial block of its activity leads to an increased Aß secretion by some cell types. We therefore compared LUHMES and BLUHMES regarding rebound effects of γ-secretase inhibitors and found an Aß rise in LUHMES but not in BLUHMES. Thus, different cellular factors are responsible for the γ-secretase- versus BACE-related Aß rebound. We conclude that increased BACE activity, possibly accompanied by an altered cellular localization pattern, can dramatically influence Aß generation in human neurons and affect pharmacological responses to secretase inhibitors. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Neurons/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Bepridil/pharmacology , Calcium Channel Blockers/pharmacology , Cell Differentiation/physiology , Cell Survival , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans
13.
Biol Pharm Bull ; 41(7): 1049-1061, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29769465

ABSTRACT

Ethanol (EtOH) dosage, frequency, and paired associative learning affect the risk of alcoholism. Recently, Spanagel et al. reported that acamprosate calcium (Acam Ca) prescribed for alcoholism exerts an anti-relapse effect via Ca. Ca is contained in foods, sometimes consumed with alcohol. Therefore, we investigated the association among oral Ca ingestion, EtOH-induced locomotor sensitization, and plasma Ca levels on how to consume Ca for moderate drinking. We used DBA/2 CrSlc mice, and CaCl2 as water-soluble Ca salts. For pre-administration, elemental Ca (50, 75, 100, or 150 mg/kg, per os (p.o.)) or water for control was administered 1 h before EtOH (2 g/kg, 20 v/v (%) EtOH in saline) administration intraperitoneal (i.p.) for locomotor sensitization or for plasma Ca level changes. For post-administration, elemental Ca (100 mg/kg) was administered 1 h after EtOH. Moreover, we employed bepridil and the dopamine D1 antagonist, SCH-23390 to further examine the mechanism of EtOH-induced sensitization. The locomotor sensitization segmentalized for 300 s had two peaks (0-90 s and 180-300 s). Pre-administration of Ca (50, 75, and 100 mg/kg) significantly reduced the 0-90-s peak, selectively blocked by SCH-23390, but "non-dose dependently" as Ca 150 mg/kg did not have this effect. Bepridil blocked the suppressive effect of pre-administration of Ca (100 mg/kg). The effective pre-doses of Ca (50-100 mg/kg) maintained plasma Ca basal levels against EtOH-induced decrease of Ca. On the contrary, post-administration of Ca inversely led to significant promotion of sensitization of both locomotor peaks. Oral Ca intake had diverse effects on EtOH-induced sensitization depending on Ca dosage and timing.


Subject(s)
Alcoholism/drug therapy , Calcium, Dietary/pharmacology , Ethanol/pharmacology , Locomotion/drug effects , Administration, Oral , Alcoholism/blood , Animals , Benzazepines/pharmacology , Bepridil/pharmacology , Calcium, Dietary/blood , Calcium, Dietary/therapeutic use , Conditioning, Psychological/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred DBA , Motor Activity/drug effects , Time Factors , Treatment Outcome
14.
Int J Cancer ; 143(4): 958-970, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29508386

ABSTRACT

Dysregulated NOTCH1 signaling, by either gene mutations or microenvironment interactions, has been increasingly linked to chronic lymphocytic leukemia (CLL). Thus, inhibiting NOTCH1 activity represents a potential therapeutic opportunity for this disease. Using gene expression-based screening, we identified the calcium channel modulator bepridil as a new NOTCH1 pathway inhibitor. In primary CLL cells, bepridil induced selective apoptosis even in the presence of the protective stroma. Cytotoxic effects of bepridil were independent of NOTCH1 mutation and other prognostic markers. The antitumor efficacy of bepridil was associated with inhibition of NOTCH1 activity through a decrement in trans-membrane and activated NOTCH1 protein levels with unchanged NOTCH2 protein levels. In a CLL xenotransplant model, bepridil significantly reduced the percentage of leukemic cells infiltrating the spleen via enhanced apoptosis and decreased NOTCH1 activation. In conclusion, we report in vitro and in vivo anti-leukemic activity of bepridil associated with inhibition of the NOTCH1 pathway in CLL. These data provide a rationale for the clinical development of bepridil as anti-NOTCH1 targeted therapy for CLL patients.


Subject(s)
Antineoplastic Agents/pharmacology , Bepridil/pharmacology , Calcium Channel Blockers/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Chemotaxis/drug effects , Drug Screening Assays, Antitumor , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mutation , Prognosis , Receptor, Notch1/genetics , Tumor Microenvironment , Xenograft Model Antitumor Assays
15.
ChemMedChem ; 13(7): 754-761, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29359495

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF1) is a toxin produced by pathogenic strains of Escherichia coli responsible for extra-intestinal infections. CNF1 deamidates Rac1, thereby triggering its permanent activation and worsening inflammatory reactions. Activated Rac1 is prone to proteasomal degradation. There is no targeted therapy against CNF1, despite its clinical relevance. In this work we developed a fluorescent cell-based immunoassay to screen for inhibitors of CNF1-induced Rac1 degradation among 1120 mostly approved drugs. Eleven compounds were found to prevent CNF1-induced Rac1 degradation, and five also showed a protective effect against CNF1-induced multinucleation. Finally, lasalocid, monensin, bepridil, and amodiaquine protected cells from both diphtheria toxin and CNF1 challenges. These data highlight the potential for drug repurposing to fight several bacterial infections and Rac1-based diseases.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Escherichia coli Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , rac1 GTP-Binding Protein/metabolism , Amodiaquine/pharmacology , Bacterial Toxins/adverse effects , Bacterial Toxins/metabolism , Bepridil/pharmacology , Diphtheria Toxin/adverse effects , Drug Repositioning , Escherichia coli/chemistry , Escherichia coli Proteins/adverse effects , Escherichia coli Proteins/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunoassay , Lasalocid/pharmacology , Monensin/pharmacology , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/immunology
16.
Eur J Neurosci ; 46(5): 2108-2120, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28715135

ABSTRACT

The Na+ /Ca2+ exchanger (NCX) is a bidirectional plasma membrane antiporter involved in Ca2+ homeostasis in eukaryotes. NCX has three isoforms, NCX1-3, and all of them are expressed in the cerebellum. Immunostaining on cerebellar slice cultures indicates that NCX is widely expressed in the cerebellum, including expression in Purkinje cells. The pharmacological blockade of the forward mode of NCX (Ca2+ efflux mode) by bepridil moderately inhibited growth and development of Purkinje cell dendritic arbor in cerebellar slice cultures. However, the blockade of the reverse mode (Ca2+ influx mode) by KB-R7943 severely reduced the dendritic arbor and induced a morphological change with thickened distal dendrites. The effect of KB-R7943 on dendritic growth was unrelated to the activity of voltage-gated calcium channels and was also apparent in the absence of bioelectrical activity indicating that it was mediated by NCX expressed in Purkinje cells. We have used additional NCX inhibitors including CB-DMB, ORM-10103, SEA0400, YM-244769, and SN-6 which have higher specificity for NCX isoforms and target either the forward, reverse, or both modes. These inhibitors caused a strong dendritic reduction similar to that seen with KB-R7943, but did not elicit thickening of distal dendrites. Our findings indicate that disturbance of the NCX-dependent calcium transport in Purkinje cells induces a reduction of dendritic arbor, which is presumably caused by changes in the calcium handling, and underline the importance of the calcium equilibrium for the dendritic development in cerebellar Purkinje cells.


Subject(s)
Dendrites/drug effects , Neuronal Plasticity/drug effects , Purkinje Cells/drug effects , Sodium-Calcium Exchanger/antagonists & inhibitors , Animals , Bepridil/pharmacology , Calcium Channels/metabolism , Central Nervous System Agents/pharmacology , Dendrites/metabolism , Dendrites/pathology , Immunohistochemistry , Mice , Microscopy, Confocal , Neuronal Plasticity/physiology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Purkinje Cells/metabolism , Purkinje Cells/pathology , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Sodium-Calcium Exchanger/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology , Tissue Culture Techniques
17.
Physiol Res ; 66(3): 459-465, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28248537

ABSTRACT

Various types of mechanosensitive ion channels, including cationic stretch-activated channels (SAC(NS)) and stretch-activated BKca (SAKca) channels, modulate heart rhythm. Bepridil has been used as an antiarrhythmic drug with multiple pharmacological effects; however, whether it is effective for mechanically induced arrhythmia has not been well investigated. To test the effects of Bepridil on SAKca channels activity, cultured chick embryonic ventricular myocytes were used for single-channel recordings. Bepridil significantly reduced the open probability of the SAKca channel (P(O)). Next, to test the effects of bepridil on stretch-induced extrasystoles (SIE), we used an isolated 2-week-old Langendorff-perfused chick heart. The left ventricle (LV) volume was rapidly changed, and the probability of SIE was calculated in the presence and absence of bepridil, and the effect of the drug was compared with that of Gadolinium (Gd(3+)). Bepridil decreased the probability of SIE despite its suppressive effects on SAKca channel activity. The effects of Gd(3+), which blocks both SAKca and SAC(NS), on the probability of SIE were the same as those of bepridil. Our results suggest that bepridil blocks not only SAKca channels but possible also blocks SAC(NS), and thus decreases the stretch-induced cation influx (stabilizing membrane potential) to compensate and override the effects of the decrease in outward SAKca current (destabilizing membrane potential).


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Bepridil/pharmacology , Isolated Heart Preparation , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/physiology , Ventricular Premature Complexes/physiopathology , Animals , Anti-Arrhythmia Agents/therapeutic use , Bepridil/therapeutic use , Cells, Cultured , Chick Embryo , Electrocardiography/drug effects , Electrocardiography/methods , Heart , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Ventricular Premature Complexes/drug therapy
18.
Oncotarget ; 7(27): 42110-42125, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27283899

ABSTRACT

Triple-negative breast cancer (TNBC) is the most lethal form of breast cancer. Lacking effective therapeutic options hinders treatment of TNBC. Here, we show that bepridil (BPD) and trifluoperazine (TFP), which are FDA-approved drugs for treatment of schizophrenia and angina respectively, inhibit Akt-pS473 phosphorylation and promote FOXO3 nuclear localization and activation in TNBC cells. BPD and TFP inhibit survival and proliferation in TNBC cells and suppress the growth of TNBC tumors, whereas silencing FOXO3 reduces the BPD- and TFP-mediated suppression of survival in TNBC cells. While BPD and TFP decrease the expression of oncogenic c-Myc, KLF5, and dopamine receptor DRD2 in TNBC cells, silencing FOXO3 diminishes BPD- and TFP-mediated repression of the expression of these proteins in TNBC cells. Since c-Myc, KLF5, and DRD2 have been suggested to increase cancer stem cell-like populations in various tumors, reducing these proteins in response to BPD and TFP suggests a novel FOXO3-dependent mechanism underlying BPD- and TFP-induced apoptosis in TNBC cells.


Subject(s)
Breast Neoplasms/drug therapy , Forkhead Box Protein O3/metabolism , Gene Silencing , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis , Bepridil/pharmacology , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Dopamine/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Transcription Factors/metabolism , MCF-7 Cells , Mice , Mice, Nude , Neoplastic Stem Cells , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Receptors, Dopamine D2/metabolism , Trifluoperazine/pharmacology , Triple Negative Breast Neoplasms/metabolism
19.
J Pharmacol Sci ; 130(3): 170-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27032905

ABSTRACT

Electrical remodeling plays a pivotal role in maintaining the reentry during atrial fibrillation. In this study, we assessed influence of electrical remodeling on pharmacological manipulation of the atrial refractoriness in rabbits. We used an atrial electrical remodeling model of the rabbit, subjected to rapid atrial pacing (RAP; 600 beats/min) for 2-4 weeks, leading to shortening of atrial effective refractory period (AERP). Intravenous administration of dl-sotalol (6 mg/kg), bepridil (1 mg/kg), amiodarone (10 mg/kg) or vernakalant (3 mg/kg) significantly prolonged the AERP both in the control and RAP rabbits. The extents in the RAP rabbits were similar to those in the control animals. On the other hand, prolonging effects of intravenously administered ranolazine (10 mg/kg) or tertiapin-Q (0.03 mg/kg) on the AERP in the RAP rabbits were more potent than those in the control animals. These results suggest that rapid pacing-induced electrical remodeling effectively modified the prolonging effects of ranolazine and tertiapin-Q on the AERP in contrast to those of clinically available antiarrhythmic drugs, dl-sotalol, bepridil amiodarone and vernakalant.


Subject(s)
Amiodarone/pharmacology , Anisoles/pharmacology , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Atrial Remodeling/drug effects , Atrial Remodeling/physiology , Bepridil/pharmacology , Pyrrolidines/pharmacology , Sotalol/pharmacology , Amiodarone/administration & dosage , Animals , Anisoles/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Bepridil/administration & dosage , Cardiac Pacing, Artificial , Disease Models, Animal , Infusions, Intravenous , Male , Pyrrolidines/administration & dosage , Rabbits , Sotalol/administration & dosage
20.
Heart Vessels ; 31(7): 1176-84, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26498939

ABSTRACT

Bepridil is an effective antiarrhythmic drug on supraventricular and ventricular arrhythmias, and inhibitor of calmodulin. Recent investigations have been elucidating that bepridil exerts antiarrhythmic effects through its acute and chronic application for patients. The aim of this study was to identify the efficacy and the potential mechanism of bepridil on the inward-rectifier potassium channel in neonatal rat cardiomyocytes in acute- and long-term conditions. Bepridil inhibited inward-rectifier potassium current (I K1) as a short-term effect with IC50 of 17 µM. Bepridil also reduced I K1 of neonatal cardiomyocytes when applied for 24 h in the culture medium with IC50 of 2.7 µM. Both a calmodulin inhibitor (W-7) and an inhibitor of calmodulin-kinase II (KN93) reduced I K1 when applied for 24 h as a long-term effect in the same fashion, suggesting that the long-term application of bepridil inhibits I K1 more potently than that of the short-term application through the inhibition of calmodulin kinase II pathway in cardiomyocytes.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Bepridil/pharmacology , Myocytes, Cardiac/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Animals , Animals, Newborn , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Cells, Cultured , Membrane Potentials , Myocytes, Cardiac/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Protein Kinase Inhibitors/pharmacology , Rats, Wistar , Sulfonamides/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL