Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
Int J Mol Sci ; 24(14)2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37511269

ABSTRACT

The first conotoxin affecting the voltage-gated potassium channels of the EAG family was identified and characterized from the venom of the vermivorous species Conus spurius from the Gulf of Mexico. This conopeptide, initially named Cs68 and later designated κO-SrVIA, is extremely hydrophobic and comprises 31 amino acid residues, including six Cysteines in the framework VI/VII, and a free C-terminus. It inhibits the currents mediated by two human EAG subtypes, Kv10.1 (IC50 = 1.88 ± 1.08 µM) and Kv11.1 (IC50 = 2.44 ± 1.06 µM), and also the human subtype Kv1.6 (IC50 = 3.6 ± 1.04 µM). Despite its clear effects on potassium channels, it shares a high sequence identity with δ-like-AtVIA and δ-TsVIA. Also, κO-SrVIA is the third conopeptide from the venom of C. spurius with effects on potassium channels, and the seventh conotoxin that blocks Kv1.6 channels.


Subject(s)
Conotoxins , Conus Snail , Ether-A-Go-Go Potassium Channels , Animals , Humans , Amino Acid Sequence , Conotoxins/pharmacology , Conotoxins/chemistry , Conus Snail/chemistry , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/toxicity , Peptides/chemistry
2.
Drug Chem Toxicol ; 46(3): 430-440, 2023 May.
Article in English | MEDLINE | ID: mdl-35296205

ABSTRACT

Two synthetic phenylethylamines, N-methyl-1-(naphthalen-2-yl)propan-2-amine (MNA) and 1-phenyl-2-pyrrolidinylpentane (prolintane), are being abused by people seeking hallucinogens for pleasure. These new psychotropic substances may provoke problems because there is no existing information about their toxicity and pharmacological behaviors. Therefore, we evaluated the safety of nerves and cardiovascular systems by determining toxicity after MNA and prolintane drugs administrations to mice and rat. Consequently, side effects such as increased spontaneous motion and body temperature were observed in oral administration of MNA. In addition, both substances reduced motor coordination levels. The IHC tests were conducted to see whether the immune response also shows abnormalities in brain tissue compared to the control group. It has been confirmed that the length of allograft inflammatory factor 1(IBA-1), an immune antibody known as microglia marker, has been shortened. We identified that a problem with the contact between synapses and neurons might be possibly produced. In the assessment of the cardiac toxicity harmfulness, no substances have been confirmed to be toxic to myocardial cells, but at certain concentrations, they have caused the QT prolongation, an indicator of ventricular arrhythmia. In addition, the hERG potassium channel, the biomarker of the QT prolongation, has been checked for inhibition. The results revealed that the possibility of QT prolongation through the hERG channel could not be excluded, and the two substances can be considered toxic that may cause ventricular arrhythmia. In sum, this study demonstrated that the possibility of toxicity in MNA and prolintane compounds might bring many harmful effects on nerves and hearts.


Subject(s)
Cardiotoxicity , Hallucinogens , Long QT Syndrome , Neurotoxicity Syndromes , Phenethylamines , Animals , Mice , Rats , Cardiotoxicity/etiology , Ether-A-Go-Go Potassium Channels/drug effects , Hallucinogens/toxicity , Long QT Syndrome/chemically induced , Myocytes, Cardiac/drug effects , Neurotoxicity Syndromes/etiology , Phenethylamines/toxicity
3.
Toxicology ; 458: 152822, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34058295

ABSTRACT

Hydroxychloroquine (HCQ) was noted to produce severe cardiac arrhythmia, an adverse effect as its use against severe acute respiratory syndrome caused by coronavirus 2 (SAES-CoV-2). HCQ is an antimalarial drug with quinoline structure. Some other quinoline compounds, such as fluoroquinolone antibiotics (FQs), also lead to arrhythmias characterized by QT prolongation. QT prolongation is usually related to the human ether-a-go-go-related gene (hERG) potassium channel inhibitory activity of most drugs. In this research, molecular docking was used to study the potential inhibitory activities of HCQ as well as other quinolines derivatives and hERG potassium channel protein. The possible causes of these QT prolongation effects were revealed. Molecular docking and patch clamp experiments showed that HCQ could bind to hERG and inhibit the efflux of potassium ion preferentially in the repolarization stage. The IC50 of HCQ was 8.6 µM ± 0.8 µM. FQs, which are quinoline derivatives, could also bind to hERG molecules. The binding energies of FQs varied according to their molecular polarity. It was found that drugs with a quinoline structure, particularly with high molecular polarity, can exert a significant potential hERG inhibitory activity. The potential side effects of QT prolongation during the development and use of quinolines should be carefully considered.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Hydroxychloroquine/pharmacology , Amino Acids/chemistry , Computational Biology , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/drug effects , HEK293 Cells , Humans , Hydroxychloroquine/chemistry , Inhibitory Concentration 50 , Molecular Docking Simulation , Patch-Clamp Techniques
4.
Int J Mol Sci ; 22(1)2020 Dec 27.
Article in English | MEDLINE | ID: mdl-33375447

ABSTRACT

Neurological disorders including depression, anxiety, post-traumatic stress disorder (PTSD), schizophrenia, autism and epilepsy are associated with an increased incidence of cardiovascular disorders and susceptibility to heart failure. The underlying molecular mechanisms that link neurological disorders and adverse cardiac function are poorly understood. Further, a lack of progress is likely due to a paucity of studies that investigate the relationship between neurological disorders and cardiac electrical activity in health and disease. Therefore, there is an important need to understand the spatiotemporal behavior of neurocardiac mechanisms. This can be advanced through the identification and validation of neurological and cardiac signaling pathways that may be adversely regulated. In this review we highlight how dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis, autonomic nervous system (ANS) activity and inflammation, predispose to psychiatric disorders and cardiac dysfunction. Moreover, antipsychotic and antidepressant medications increase the risk for adverse cardiac events, mostly through the block of the human ether-a-go-go-related gene (hERG), which plays a critical role in cardiac repolarization. Therefore, understanding how neurological disorders lead to adverse cardiac ion channel remodeling is likely to have significant implications for the development of effective therapeutic interventions and helps improve the rational development of targeted therapeutics with significant clinical implications.


Subject(s)
Antipsychotic Agents/adverse effects , Cardiovascular Diseases/complications , Cardiovascular Diseases/metabolism , Ion Channels/drug effects , Mental Disorders/drug therapy , Antipsychotic Agents/therapeutic use , Arrhythmias, Cardiac/complications , Arrhythmias, Cardiac/metabolism , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , Mental Disorders/complications , Risk Factors
5.
Sci Rep ; 10(1): 16262, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004839

ABSTRACT

Off-target interactions of drugs with the human ether-à-go-go related gene 1 (hERG1) channel have been associated with severe cardiotoxic conditions leading to the withdrawal of many drugs from the market over the last decades. Consequently, predicting drug-induced hERG-liability is now a prerequisite in any drug discovery campaign. Understanding the atomic level interactions of drug with the channel is essential to guide the efficient development of safe drugs. Here we utilize the recent cryo-EM structure of the hERG channel and describe an integrated computational workflow to characterize different drug-hERG interactions. The workflow employs various structure-based approaches and provides qualitative and quantitative insights into drug binding to hERG. Our protocol accurately differentiated the strong blockers from weak and revealed three potential anchoring sites in hERG. Drugs engaging in all these sites tend to have high affinity towards hERG. Our results were cross-validated using a fluorescence polarization kit binding assay and with electrophysiology measurements on the wild-type (WT-hERG) and on the two hERG mutants (Y652A-hERG and F656A-hERG), using the patch clamp technique on HEK293 cells. Finally, our analyses show that drugs binding to hERG disrupt and hijack certain native-structural networks in the channel, thereby, gaining more affinity towards hERG.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Computational Biology/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/genetics , HEK293 Cells , Humans , Patch-Clamp Techniques , Structure-Activity Relationship
6.
Neurobiol Dis ; 143: 104977, 2020 09.
Article in English | MEDLINE | ID: mdl-32553709

ABSTRACT

Environmental toxicants have the potential to contribute to the pathophysiology of multiple complex diseases, but the underlying mechanisms remain obscure. One such toxicant is the widely used fungicide ziram, a dithiocarbamate known to have neurotoxic effects and to increase the risk of Parkinson's disease. We have used Drosophila melanogaster as an unbiased discovery tool to identify novel molecular pathways by which ziram may disrupt neuronal function. Consistent with previous results in mammalian cells, we find that ziram increases the probability of synaptic vesicle release by dysregulation of the ubiquitin signaling system. In addition, we find that ziram increases neuronal excitability. Using a combination of live imaging and electrophysiology, we find that ziram increases excitability in both aminergic and glutamatergic neurons. This increased excitability is phenocopied and occluded by null mutant animals of the ether a-go-go (eag) potassium channel. A pharmacological inhibitor of the temperature sensitive hERG (human ether-a-go-go related gene) phenocopies the excitability effects of ziram but only at elevated temperatures. seizure (sei), a fly ortholog of hERG, is thus another candidate target of ziram. Taken together, the eag family of potassium channels emerges as a candidate for mediating some of the toxic effects of ziram. We propose that ziram may contribute to the risk of complex human diseases by blockade of human eag and sei orthologs, such as hERG.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Fungicides, Industrial/toxicity , Neurons/drug effects , Synaptic Vesicles/drug effects , Ziram/toxicity , Animals , Drosophila melanogaster , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Neurons/metabolism , Neurotransmitter Agents/metabolism , Synaptic Vesicles/metabolism
7.
Toxicol Pathol ; 48(3): 465-480, 2020 04.
Article in English | MEDLINE | ID: mdl-32124659

ABSTRACT

Cyclic adenosine monophosphate-response element (CREB)-binding protein (CBP) and EP300E1A-binding protein (p300) are members of the bromodomain and extraterminal motif (BET) family. These highly homologous proteins have a key role in modulating transcription, including altering the status of chromatin or through interactions with or posttranslational modifications of transcription factors. As CBP and p300 have known roles for stimulating c-Myc oncogenic activity, a small-molecule inhibitor, GNE-781, was developed to selectively and potently inhibit the CBP/p300 bromodomains (BRDs). Genetic models have been challenging to develop due to embryonic lethality arising from germline homozygous mutations in either CBP or P300. Hence, the purpose of this study was to characterize the role of dual inhibition of these proteins in adult rats and dogs. Repeat dose toxicity studies were conducted, and toxicologic and pathologic end points were assessed. GNE-781 was generally tolerated; however, marked effects on thrombopoiesis occurred in both species. Evidence of inhibition of erythroid, granulocytic, and lymphoid cell differentiation was also present, as well as deleterious changes in gastrointestinal and reproductive tissues. These findings are consistent with many preclinical (and clinical) effects reported with BET inhibitors targeting BRD proteins; thus, the current study findings indicate a likely important role for CBP/p300 in stem cell differentiation.


Subject(s)
Pyrazoles/pharmacology , Pyridines/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Animals , Dogs , Drug Evaluation, Preclinical/methods , Ether-A-Go-Go Potassium Channels/drug effects , Female , Humans , Male , Rats , Rats, Sprague-Dawley
8.
Cell Death Dis ; 11(3): 161, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123164

ABSTRACT

We have studied how the macrolide antibiotic Clarithromycin (Cla) regulates autophagy, which sustains cell survival and resistance to chemotherapy in cancer. We found Cla to inhibit the growth of human colorectal cancer (CRC) cells, by modulating the autophagic flux and triggering apoptosis. The accumulation of cytosolic autophagosomes accompanied by the modulation of autophagic markers LC3-II and p62/SQSTM1, points to autophagy exhaustion. Because Cla is known to bind human Ether-à-go-go Related Gene 1 (hERG1) K+ channels, we studied if its effects depended on hERG1 and its conformational states. By availing of hERG1 mutants with different gating properties, we found that fluorescently labelled Cla preferentially bound to the closed channels. Furthermore, by sequestering the channel in the closed conformation, Cla inhibited the formation of a macromolecular complex between hERG1 and the p85 subunit of PI3K. This strongly reduced Akt phosphorylation, and stimulated the p53-dependent cell apoptosis, as witnessed by late caspase activation. Finally, Cla enhanced the cytotoxic effect of 5-fluorouracil (5-FU), the main chemotherapeutic agent in CRC, in vitro and in a xenograft CRC model. We conclude that Cla affects the autophagic flux by impairing the signaling pathway linking hERG1 and PI3K. Combining Cla with 5-FU might be a novel therapeutic option in CRC.


Subject(s)
Autophagy/drug effects , Clarithromycin/pharmacology , Colonic Neoplasms/drug therapy , Ether-A-Go-Go Potassium Channels/drug effects , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism
9.
Ann Noninvasive Electrocardiol ; 24(6): e12679, 2019 11.
Article in English | MEDLINE | ID: mdl-31347753

ABSTRACT

BACKGROUND: Human ether-à-go-go-related gene (hERG) potassium-channel block represents a harmful side effect of drug therapy that may cause torsade de pointes (TdP). Analysis of ventricular repolarization through electrocardiographic T-wave features represents a noninvasive way to accurately evaluate the TdP risk in drug-safety studies. This study proposes an artificial neural network (ANN) for noninvasive electrocardiography-based classification of the hERG potassium-channel block. METHODS: The data were taken from the "ECG Effects of Ranolazine, Dofetilide, Verapamil, and Quinidine in Healthy Subjects" Physionet database; they consisted of median vector magnitude (VM) beats of 22 healthy subjects receiving a single 500 µg dose of dofetilide. Fourteen VM beats were considered for each subject, relative to time-points ranging from 0.5 hr before to 14.0 hr after dofetilide administration. For each VM, changes in two indexes accounting for the early and the late phases of repolarization, ΔERD30% and ΔTS/A , respectively, were computed as difference between values at each postdose time-point and the predose time-point. Thus, the dataset contained 286 ΔERD30% -ΔTS/A pairs, partitioned into training, validation, and test sets (114, 29, and 143 pairs, respectively) and used as inputs of a two-layer feedforward ANN with two target classes: high block (HB) and low block (LB). Optimal ANN (OANN) was identified using the training and validation sets and tested on the test set. RESULTS: Test set area under the receiver operating characteristic was 0.91; sensitivity, specificity, accuracy, and precision were 0.93, 0.83, 0.92, and 0.96, respectively. CONCLUSION: OANN represents a reliable tool for noninvasive assessment of the hERG potassium-channel block.


Subject(s)
Electrocardiography/methods , Ether-A-Go-Go Potassium Channels/drug effects , Neural Networks, Computer , Phenethylamines/administration & dosage , Potassium Channel Blockers/administration & dosage , Sulfonamides/administration & dosage , Humans
10.
Elife ; 82019 04 29.
Article in English | MEDLINE | ID: mdl-31032798

ABSTRACT

Most neurons do not simply convert inputs into firing rates. Instead, moment-to-moment firing rates reflect interactions between synaptic inputs and intrinsic currents. Few studies investigated how intrinsic currents function together to modulate output discharges and which of the currents attenuated by synthetic cholinergic ligands are actually modulated by endogenous acetylcholine (ACh). In this study we optogenetically stimulated cholinergic fibers in rat neocortex and find that ACh enhances excitability by reducing Ether-à-go-go Related Gene (ERG) K+ current. We find ERG mediates the late phase of spike-frequency adaptation in pyramidal cells and is recruited later than both SK and M currents. Attenuation of ERG during coincident depolarization and ACh release leads to reduced late phase spike-frequency adaptation and persistent firing. In neuronal ensembles, attenuating ERG enhanced signal-to-noise ratios and reduced signal correlation, suggesting that these two hallmarks of cholinergic function in vivo may result from modulation of intrinsic properties.


Subject(s)
Acetylcholine/physiology , Adaptation, Physiological , Ether-A-Go-Go Potassium Channels/physiology , Neocortex/physiology , Action Potentials/physiology , Animals , Ether-A-Go-Go Potassium Channels/drug effects , Female , Kinetics , Male , Membrane Potentials , Neurons , Potassium Channel Blockers/pharmacology , Pyramidal Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/metabolism
11.
Mol Pharm ; 16(4): 1477-1488, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30807184

ABSTRACT

The hERG potassium channel (IKr) encoded by human ether-a-go-go-related gene plays an important role in cardiac repolarization. Decreased IKr may lead to long QT syndrome, which subsequently causes torsade de pointes and sudden cardiac death. Previous studies have shown that statins inhibit IKr and are more potent in inhibiting hERG currents when combined with other drugs. Since chemical structure of rosuvastatin is similar to that of several IKr blockers (ibutilide and E-4031), the present study aimed to reveal the mechanism that underlies rosuvastatin-induced hERG current reduction and to evaluate the possibility of cardiac toxicity. The results showed that rosuvastatin reduced hERG currents by accelerating the inactivation and prolonged action potential duration (APD) in hiPSC-CMs. Meanwhile, it was observed that rosuvastatin reduced the expression of the mature hERG. Transcription factor Sp1 was involved in hERG protein downregulation induced by rosuvastatin, and the result was verified by Sp1 siRNA and Sp1 agonist epicatechin. These results indicated that rosuvastatin could potentially inhibit transcription and reduce hERG mRNA expression. The interaction between hERG and heat shock protein was evaluated to study the mechanism of trafficking inhibition through co-immunoprecipitation. We found that rosuvastatin reduces the interaction of heat shock protein 70 (Hsp70) with the hERG protein, thereby affecting the folding of the hERG channel. Additionally, rosuvastatin significantly activates ATF6, which plays a key role in the activation of the unfolded protein response (UPR) pathway. Increased expression of the molecular chaperone calnexin and calreticulin, which are activated by ATF6 to help channel folding, further confirmed UPR activation. Meanwhile, the degradation of the hERG channel was mediated by lysosomes and proteasomes. In conclusion, Rosuvastatin reduced the expression of hERG plasma membrane by two pathways, the first is to disrupt the transport of immature hERG channels to the membrane, and the second is to increase the degradation of mature hERG channels. In addition, Rosuvastatin potently blocked hERG current, delayed cardiac repolarization, and thereby prolonged APDs and QTc intervals. Therefore, caution should be taken when rosuvastatin is used in the treatment of hyperlipidemia, especially when combined with drugs that can prolong the QT interval.


Subject(s)
Anticholesteremic Agents/pharmacology , Cell Membrane/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Proteolysis/drug effects , Rosuvastatin Calcium/pharmacology , Action Potentials , Cell Membrane/drug effects , Ether-A-Go-Go Potassium Channels/drug effects , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Protein Transport , Unfolded Protein Response
12.
Biochem Biophys Res Commun ; 503(4): 2531-2535, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30208521

ABSTRACT

The ether-à-go-go1 (EAG1, Kv10.1) K+ channel is a member of the voltage-gated K+ channel family mainly expressed in the central nervous system and cancer cells. Membrane lipids regulate several voltage-gated K+ channels but their influence on EAG1 channels has been poorly explored. Here we have studied the regulation of hEAG1 channels by phosphatidylinositol 4,5-bisfofate (PIP2) by using different strategies to manipulate the levels of this lipid, and the patch clamp technique. We found that depletion of endogenous PIP2 by activation of the voltage-sensing phosphatase from Danio rerio (Dr-VSP) or the human muscarinic type-1 receptor (hM1R) inhibits hEAG1 currents; however, the application of exogenous PIP2 to increase the level of this lipid on the plasma membrane, also induced an inhibition of hEAG1. In summary, our results indicate that PIP2 have dual effects on hEAG1 channels and its action as activator or inhibitor depends on its initial level on the plasma membrane.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Animals , Humans , Patch-Clamp Techniques , Phosphoric Monoester Hydrolases , Receptors, Muscarinic , Zebrafish
13.
J Membr Biol ; 251(5-6): 711-722, 2018 12.
Article in English | MEDLINE | ID: mdl-30206647

ABSTRACT

ERG (ether-a-go-go-related gene) channels are the members of the voltage-dependent potassium channel family, which have three subtypes, as ERG1 (Kv 11.1), ERG2 (Kv 11.2), and ERG3 (Kv11.3). There is no information on ERG channels in the cochlear nucleus (CN) neurons, which is the first relay station of the auditory pathway. As occur in some of congenital long QT Syndromes (LQTS), mutation of the KCNQ11 genes for ERG channel has been reported to be accompanied by hearing loss. For that reason, we aimed to study biophysical properties and physiological importance, and contribution of ERG K+ currents to the formation of action potentials in the stellate and bushy neurons of the ventral cochlear nucleus (VCN). A total of 70 mice at 14-17 days old were used for this study. Electrophysiological characterization of ERG channels was performed using patch-clamp technique in the CN slices. In current clamp, ERG channel blockers, terfenadine (10 µM) and E-4031 (10 µM), were applied in both cell types. The activation, inactivation, and deactivation kinetics of the ERG channels were determined by voltage clamp. In conclusion, the findings obtained in the present study suggest that stellate and bushy neurons express ERG channels and ERG channels appear to contribute to setting action potential (AP) frequency, threshold for AP induction, and, possibly, resting membrane potentials in this cells.


Subject(s)
Cochlear Nucleus/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Auditory Pathways/drug effects , Auditory Pathways/physiology , Cochlear Nucleus/drug effects , Electrophysiology , Ether-A-Go-Go Potassium Channels/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Piperidines/pharmacology , Pyridines/pharmacology , Stellate Ganglion/drug effects , Stellate Ganglion/metabolism , Terfenadine/pharmacology
14.
J Med Chem ; 60(19): 8145-8159, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28880552

ABSTRACT

Our previous work ( Wang et al. J. Med. Chem. 2016 , 59 , 4831 - 4848 ) revealed that effective benzocycloalkane-derived staphyloxanthin inhibitors against methicillin-resistant Staphylococcus aureus (S. aureus) infections were accompanied by poor water solubility and high hERG inhibition and dosages (preadministration). In this study, 92 chroman and coumaran derivatives as novel inhibitors have been addressed for overcoming deficiencies above. Derivatives 69 and 105 displayed excellent pigment inhibitory activities and low hERG inhibition, along with improvement of solubility by salt type selection. The broad and significantly potent antibacterial spectra of 69 and 105 were displayed first with normal administration in the livers and hearts in mice against pigmented S. aureus Newman, Mu50 (vancomycin-intermediate S. aureus), and NRS271 (linezolid-resistant S. aureus), compared with linezolid and vancomycin. In summary, both 69 and 105 have the potential to be developed as good antibacterial candidates targeting virulence factors.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Linezolid/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Vancomycin/pharmacology , Xanthophylls/antagonists & inhibitors , Animals , Anti-Bacterial Agents/pharmacokinetics , Antifungal Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Ether-A-Go-Go Potassium Channels/drug effects , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/microbiology , Structure-Activity Relationship , Vancomycin Resistance/drug effects
15.
J Med Chem ; 60(19): 7984-7999, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28892629

ABSTRACT

Inhibition of lysine specific demethylase 1 (LSD1) has been shown to induce the differentiation of leukemia stem cells in acute myeloid leukemia (AML). Irreversible inhibitors developed from the nonspecific inhibitor tranylcypromine have entered clinical trials; however, the development of effective reversible inhibitors has proved more challenging. Herein, we describe our efforts to identify reversible inhibitors of LSD1 from a high throughput screen and subsequent in silico modeling approaches. From a single hit (12) validated by biochemical and biophysical assays, we describe our efforts to develop acyclic scaffold-hops from GSK-690 (1). A further scaffold modification to a (4-cyanophenyl)glycinamide (e.g., 29a) led to the development of compound 32, with a Kd value of 32 nM and an EC50 value of 0.67 µM in a surrogate cellular biomarker assay. Moreover, this derivative does not display the same level of hERG liability as observed with 1 and represents a promising lead for further development.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glycine/analogs & derivatives , Histone Demethylases/antagonists & inhibitors , Leukemia/drug therapy , Spiro Compounds/pharmacology , Biomarkers , Cell Line, Tumor , Computer Simulation , Drug Design , Drug Discovery , Ether-A-Go-Go Potassium Channels/drug effects , Glycine/chemical synthesis , Glycine/pharmacology , High-Throughput Screening Assays , Humans , Models, Molecular , Molecular Docking Simulation , Spiro Compounds/chemical synthesis , Structure-Activity Relationship , Tranylcypromine/analogs & derivatives , Tranylcypromine/chemistry , Tranylcypromine/pharmacology
16.
Nat Prod Rep ; 34(8): 957-980, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28497823

ABSTRACT

Covering: 1996-December 2016The human Ether-à-go-go Related Gene (hERG) channel is a voltage-gated potassium channel playing an essential role in the normal electrical activity in the heart. It is involved in the repolarization and termination of action potentials in excitable cardiac cells. Mutations in the hERG gene and hERG channel blockage by small molecules are associated with increased risk of fatal arrhythmias. Several drugs have been withdrawn from the market due to hERG channel-related cardiotoxicity. Moreover, as a result of its notorious ligand promiscuity, this ion channel has emerged as an important antitarget in early drug discovery and development. Surprisingly, the hERG channel blocking profile of natural compounds present in frequently consumed botanicals (i.e. dietary supplements, spices, and herbal medicinal products) is not routinely assessed. This comprehensive review will address these issues and provide a critical compilation of hERG channel data for isolated natural products and extracts over the past two decades (1996-2016). In addition, the review will provide (i) a solid basis for the molecular understanding of the physiological functions of the hERG channel, (ii) the translational potential of in vitro/in vivo results to cardiotoxicity in humans, (iii) approaches for the identification of hERG channel blockers from natural sources, (iv) future perspectives for cardiac safety guidelines and their applications within phytopharmaceuticals and dietary supplements, and (v) novel applications of hERG channel modulation (e.g. as a drug target).


Subject(s)
Alkaloids , Arrhythmias, Cardiac/physiopathology , Biological Products , Ether-A-Go-Go Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/genetics , Heart/physiology , Alkaloids/chemistry , Alkaloids/isolation & purification , Alkaloids/pharmacology , Biological Products/chemistry , Biological Products/isolation & purification , Biological Products/pharmacology , Drug Discovery , Humans , Molecular Structure , Myocytes, Cardiac/physiology , Plants, Medicinal/chemistry
17.
Circ Arrhythm Electrophysiol ; 10(2): e004628, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28202629

ABSTRACT

BACKGROUND: The current proarrhythmia safety testing paradigm, although highly efficient in preventing new torsadogenic drugs from entering the market, has important limitations that can restrict the development and use of valuable new therapeutics. The CiPA (Comprehensive in vitro Proarrhythmia Assay) proposes to overcome these limitations by evaluating drug effects on multiple cardiac ion channels in vitro and using these data in a predictive in silico model of the adult human ventricular myocyte. A set of drugs with known clinical torsade de pointes risk was selected to develop and calibrate the in silico model. METHODS AND RESULTS: Manual patch-clamp data assessing drug effects on expressed cardiac ion channels were integrated into the O'Hara-Rudy myocyte model modified to include dynamic drug-hERG channel (human Ether-à-go-go-Related Gene) interactions. Together with multichannel pharmacology data, this model predicts that compounds with high torsadogenic risk are more likely to be trapped within the hERG channel and show stronger reverse use dependency of action potential prolongation. Furthermore, drug-induced changes in the amount of electronic charge carried by the late sodium and L-type calcium currents was evaluated as a potential metric for assigning torsadogenic risk. CONCLUSIONS: Modeling dynamic drug-hERG channel interactions and multi-ion channel pharmacology improves the prediction of torsadogenic risk. With further development, these methods have the potential to improve the regulatory assessment of drug safety models under the CiPA paradigm.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Long QT Syndrome/chemically induced , Torsades de Pointes/chemically induced , Biomarkers/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Ion Channels/drug effects , Kinetics , Long QT Syndrome/physiopathology , Membrane Potentials/drug effects , Patch-Clamp Techniques , Risk Assessment , Torsades de Pointes/physiopathology
18.
J Pharmacol Toxicol Methods ; 81: 151-60, 2016.
Article in English | MEDLINE | ID: mdl-27374776

ABSTRACT

In the present study, we investigated an impact of the stimulation rate on the detection of the proarrhythmic potential of 10 reference compounds with effects on different cardiac ion channels in the isolated arterially-perfused rabbit left ventricular wedge preparation. The compounds were tested in the wedge model using two distinct protocols; including baseline stimulation at 1-Hz followed by a brief period at 0.5-Hz, either without an additional brief period of 2-Hz stimulation (i.e. Protocol 1) or with 2-Hz stimulation (i.e. Protocol 2). As expected, QT-prolonging drugs (ibutilide and quinidine) prolonged the QT interval, similarly increased the Torsades de Pointes (TdP) score, and elicited early afterdepolarizations (EADs) in both protocols. HMR1556 and JNJ-303 (IKs blockers) also prolonged the QT interval up to 1µM similarly in both protocols. Nifedipine (Ca(2+) antagonist) shortened the QT interval, and reduced force of contraction similarly in both protocols. However, Na(+) channel blockers (Ia, Ib, Ic) widened the QRS duration more in Protocol 2 than in Protocol 1. Furthermore, it was only possible to detect non-TdP-like ventricular tachycardia/fibrillation (VT/VF) induced by Na(+) blockers and by QT-shortening drugs (levcromakalim and mallotoxin) using the 2-Hz stimulation (Protocol 2). Our data suggest that the inclusion of a brief period of fast stimulation at 2Hz is critical for detecting drug-induced slowing of conduction (QRS widening), QT shortening and associated (non-TdP-like) VT/VF, which are distinct from the QT prolongation/TdP proarrhythmia in isolated, arterially-perfused rabbit left ventricular wedges.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Animals , Disease Models, Animal , Electrocardiography/drug effects , Ether-A-Go-Go Potassium Channels/drug effects , Female , Heart Conduction System/drug effects , In Vitro Techniques , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Male , Rabbits , Sodium Channel Blockers/pharmacology , Tachycardia, Ventricular/chemically induced , Tachycardia, Ventricular/physiopathology , Torsades de Pointes/chemically induced , Torsades de Pointes/physiopathology
19.
Article in English | MEDLINE | ID: mdl-27178106

ABSTRACT

INTRODUCTION: Current regulatory guidelines for assessing the risk of QT prolongation include in vitro assays assessing drug effects on the human ether-à-go-go-related (hERG; also known as Kv11.1) channel expressed in cell lines. These assays are typically conducted at room temperature to promote the ease and stability of recording hERG currents. However, the new Comprehensive in vitro Proarrhythmia Assay (CiPA) paradigm proposes to use an in silico model of the human ventricular myocyte to assess risk, requiring as input hERG channel pharmacology data obtained at physiological temperatures. To accommodate current industry safety pharmacology practices for measuring hERG channel activity, an in silico model of hERG channel that allows for the extrapolation of hERG assay data across different temperatures is desired. Because temperature may have an effect on both channel gating and drug binding rate, such models may need to have two components: a base model dealing with temperature-dependent gating changes without drug, and a pharmacodynamic component simulating temperature-dependent drug binding kinetics. As a first step, a base mode that can capture temperature effects on hERG channel gating without drug is needed. METHODS AND RESULTS: To meet this need for a temperature-dependent base model, a Markov model of the hERG channel with state transition rates explicitly dependent on temperature was developed and calibrated using data from a variety of published experiments conducted over a range of temperatures. The model was able to reproduce observed temperature-dependent changes in key channel gating properties and also to predict the results obtained in independent sets of new experiments. DISCUSSION: This new temperature-sensitive model of hERG gating represents an attempt to improve the predictivity of safety pharmacology testing by enabling the translation of room temperature hERG assay data to more physiological conditions. With further development, this model can be incorporated into the CiPA paradigm and also be used as a tool for developing insights into the thermodynamics of hERG channel gating mechanisms and the temperature-dependence of hERG channel block by drugs.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Algorithms , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Calibration , Computer Simulation , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Kinetics , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Markov Chains , Membrane Potentials/drug effects , Potassium Channel Blockers/pharmacology , Safety , Temperature
20.
Curr Protoc Toxicol ; 68: 22.4.1-22.4.32, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27145112

ABSTRACT

More relevant and reliable preclinical cardiotoxicity tests are required to improve drug safety and reduce the cost of drug development. Human stem cell-derived cardiomyocytes (hSC-CMs) provide a potential model for the development of superior assays for preclinical drug safety screening. One such hSC-CM assay that has shown significant potential for enabling more predictive drug cardiac risk assessment is the MEA assay. The Multi-electrode Array (MEA) assay is an electrophysiology-based technique that uses microelectrodes embedded in the culture surface of each well to measure fluctuations in extracellular field potential (FP) generated from spontaneously beating hSC-CMs. Perturbations to the recorded FP waveform can be used as an unbiased method of predicting the identity of ion channel(s) impacted on drug exposure. Here, a higher throughput MEA assay using hSC-CMs in 48-well MEA plates is described for profiling compound-induced effects on cardiomyocyte electrophysiology. Techniques for preparing hSC-CM monolayers in MEA plates and methods to contextualize MEA assay experimental results are also covered. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Cardiotoxins/toxicity , Electrophysiological Phenomena/drug effects , Myocytes, Cardiac/drug effects , Stem Cells/drug effects , Toxicology/methods , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Electrodes , Ether-A-Go-Go Potassium Channels/drug effects , Extracellular Space , Heart Rate/drug effects , Humans , Ion Channels/drug effects , Microelectrodes , Predictive Value of Tests , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...