Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 235
Filter
1.
J Pharmacol Toxicol Methods ; 128: 107527, 2024.
Article in English | MEDLINE | ID: mdl-38852685

ABSTRACT

INTRODUCTION: Cardiovascular safety and the risk of developing the potentially fatal ventricular tachyarrhythmia, Torsades de Pointes (TdP), have long been major concerns of drug development. TdP is associated with a delayed ventricular repolarization represented by QT interval prolongation in the electrocardiogram (ECG), typically due to block of the potassium channel encoded by the human ether-a-go-go related gene (hERG). Importantly however, not all drugs that prolong the QT interval are torsadagenic and not all hERG blockers prolong the QT interval. Recent clinical reports suggest that partitioning the QT interval into early (J to T peak; JTp) and late repolarization (T peak to T end; TpTe) components may be valuable for distinguishing low-risk mixed ion channel blockers (hERG plus calcium and/or late sodium currents) from high-risk pure hERG channel blockers. This strategy, if true for nonclinical animal models, could be used to de-risk QT prolonging compounds earlier in the drug development process. METHODS: To explore this, we investigated JTp and TpTe in ECG data collected from telemetered dogs and/or monkeys administered moxifloxacin or amiodarone at doses targeting relevant clinical exposures. An optimized placement of the Tpeak fiducial mark was utilized, and all intervals were corrected for heart rate (QTc, JTpc, TpTec). RESULTS: Increases in QTc and JTpc intervals with administration of the pure hERG blocker moxifloxacin and an initial QTc and JTpc shortening followed by prolongation with the mixed ion channel blocker amiodarone were detected as expected, aligning with clinical data. However, anticipated increases in TpTec by both standard agents were not detected. DISCUSSION: The inability to detect changes in TpTec reduces the utility of these subintervals for prediction of arrhythmias using continuous single­lead ECGs collected from freely moving dogs and monkeys.


Subject(s)
Amiodarone , Electrocardiography , Long QT Syndrome , Moxifloxacin , Torsades de Pointes , Animals , Moxifloxacin/administration & dosage , Moxifloxacin/pharmacology , Dogs , Amiodarone/administration & dosage , Amiodarone/pharmacology , Electrocardiography/drug effects , Electrocardiography/methods , Torsades de Pointes/chemically induced , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Male , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Female , Macaca fascicularis , Fluoroquinolones/administration & dosage , Fluoroquinolones/pharmacology , Heart Rate/drug effects , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/pharmacology , ERG1 Potassium Channel/antagonists & inhibitors , ERG1 Potassium Channel/metabolism
2.
Acta Neurol Belg ; 124(4): 1289-1294, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38582798

ABSTRACT

INTRODUCTION: Fatigue is a highly prevalent debilitating symptom among patients with multiple sclerosis (PwMS), which markedly affects the quality of life. The present study aimed to evaluate the effect of extended-release fampridine on fatigue in PwMS. METHODS: This was a randomized, double-blind clinical trial on 77 PwMS with a complaint of fatigue, aged over 18 years old, randomized to extended-release fampridine (n = 44) or placebo (n = 35) for 12 weeks. Fatigue and motor function were assessed at baseline and end point. RESULTS: A total of 88 patients were recruited, of whom 77 were analyzed. 80.5% were female, with a median age of 38. 87% were diagnosed with relapsing-remitting MS (RRMS) with a median disease duration of 96 months. Fingolimod (37.7%) was considered the most frequently used DMT, followed by ani-CD20s (32.5%). The total median MFIS score was 43.5 and 37 in the fampridine and placebo groups which were not significantly different (p > 0.05). After 12 weeks, the total MFIS improved in both groups compared to the baseline, which was significant in the active group (p = 0.04). However, the final end point total MFIS was still comparable between the two groups (p = 0.11). CONCLUSION: The present study revealed a positive short-term effect of extended-release fampridine on MFIS in PwMS. However, this effect was not significantly superior to the placebo.


Subject(s)
4-Aminopyridine , Fatigue , Multiple Sclerosis , Humans , Female , Male , 4-Aminopyridine/therapeutic use , 4-Aminopyridine/administration & dosage , Adult , Double-Blind Method , Middle Aged , Fatigue/drug therapy , Fatigue/etiology , Multiple Sclerosis/complications , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/therapeutic use , Potassium Channel Blockers/administration & dosage , Treatment Outcome , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/complications
3.
CNS Drugs ; 35(9): 1009-1022, 2021 09.
Article in English | MEDLINE | ID: mdl-34322853

ABSTRACT

BACKGROUND: Prolonged-release fampridine (PR-FAM) 10-mg tablet twice daily is the only approved pharmacological treatment for improvement of walking ability in adults with multiple sclerosis (MS). LIBERATE assessed the safety/effectiveness of PR-FAM in the real-world. OBJECTIVES: The aim of this study was to collect additional safety data, including the incidence rate of seizures and other adverse events (AEs) of interest, from patients with MS taking PR-FAM in routine clinical practice (including patients aged ≥ 65 years and those with pre-existing cardiovascular risk factors). Other objectives included change over time in patient-reported evaluation of physical and psychological impact of MS while taking PR-FAM, and change over time in physician-reported assessment of walking ability in MS patients taking PR-FAM. METHODS: Patients with MS newly prescribed PR-FAM were recruited (201 sites, 13 countries). Demographic/safety data were collected at enrolment through 12 months. Physician-rated Clinical Global Impression of Improvement (CGI-I) scores for walking ability, and Multiple Sclerosis Impact Scale-29 (MSIS-29) were assessed. RESULTS: Safety analysis included 4646 patients with 3534.8 patient-years of exposure; median (range) age, 52.6 (21-85) years, 87.3% < 65 years, and 65.7% women. Treatment-emergent AEs (TEAEs) were reported in 2448 (52.7%) patients, and serious TEAEs were reported in 279 (6.0%) patients, of whom 37 (< 1%) experienced treatment-emergent serious AEs (TESAEs) considered related to PR-FAM. AEs of special interest (AESI) occurred in 1799 (38.7%) patients, and serious AESI in 128 (2.8%) patients. Seventeen (< 1%) patients experienced actual events of seizure. Overall, 1158 (24.9%) patients discontinued treatment due to lack of efficacy. At 12 months, a greater proportion of patients on-treatment had improvement from baseline in CGI-I for walking ability versus those who discontinued (61% vs. 11%; p <  0.001). MSIS-29 physical impact score improved significantly for patients on-treatment for 12 months versus those who discontinued (mean change, baseline to 12 months: - 9.99 vs. - 0.34 points; p <  0.001). Results were similar for MSIS-29 psychological impact. CONCLUSION: No new safety concerns were identified in this real-world study, suggesting that routine risk-minimization measures are effective. CGI-I and MSIS-29 scores after 12 months treatment with PR-FAM treatment show clinical benefits consistent with those previously reported. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01480063.


Subject(s)
4-Aminopyridine/administration & dosage , Multiple Sclerosis/drug therapy , Patient Reported Outcome Measures , Physicians , Potassium Channel Blockers/administration & dosage , Walking/physiology , Adult , Aged , Aged, 80 and over , Delayed-Action Preparations/administration & dosage , Female , Humans , Male , Middle Aged , Multiple Sclerosis/diagnosis , Multiple Sclerosis/physiopathology , Prospective Studies , Treatment Outcome , Young Adult
4.
Pharmacol Rep ; 73(6): 1744-1753, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34213738

ABSTRACT

BACKGROUND: Ion channels have been proposed as therapeutic targets for different types of malignancies. One of the most studied ion channels in cancer is the voltage-gated potassium channel ether-à-go-go 1 or Kv10.1. Various studies have shown that Kv10.1 expression induces the proliferation of several cancer cell lines and in vivo tumor models, while blocking or silencing inhibits proliferation. Kv10.1 is a promising target for drug discovery modulators that could be used in cancer treatment. This work aimed to screen for new Kv10.1 channel modulators using a thallium influx-based assay. METHODS: Pharmacological effects of small molecules on Kv10.1 channel activity were studied using a thallium-based fluorescent assay and patch-clamp electrophysiological recordings, both performed in HEK293 stably expressing the human Kv10.1 potassium channel. RESULTS: In thallium-sensitive fluorescent assays, we found that the small molecules loperamide and amitriptyline exert a potent inhibition on the activity of the oncogenic potassium channel Kv10.1. These results were confirmed by electrophysiological recordings, which showed that loperamide and amitriptyline decreased the amplitude of Kv10.1 currents in a dose-dependent manner. Both drugs could be promising tools for further studies. CONCLUSIONS: Thallium-sensitive fluorescent assay represents a reliable methodological tool for the primary screening of different molecules with potential activity on Kv10.1 channels or other K+ channels.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Loperamide/pharmacology , Potassium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Fluorescence , HEK293 Cells , Humans , Loperamide/administration & dosage , Patch-Clamp Techniques , Potassium Channel Blockers/administration & dosage , Reproducibility of Results , Thallium/metabolism
5.
Pharmacol Rep ; 73(6): 1724-1733, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34146337

ABSTRACT

BACKGROUND: Olanzapine, an FDA-approved atypical antipsychotic, is widely used to treat schizophrenia and bipolar disorder. In this study, the inhibitory effect of olanzapine on voltage-dependent K+ (Kv) channels in rabbit coronary arterial smooth muscle cells was investigated. METHODS: Electrophysiological recordings were performed in freshly isolated coronary arterial smooth muscle cells. RESULTS: Olanzapine inhibited the Kv channels in a concentration-dependent manner with an IC50 value of 7.76 ± 1.80 µM and a Hill coefficient of 0.82 ± 0.09. Although olanzapine did not change the steady-state activation curve, it shifted the inactivation curve to a more negative potential, suggesting that it inhibited Kv currents by affecting the voltage sensor of the Kv channel. Application of 1 or 2 Hz train pulses did not affect the olanzapine-induced inhibition of Kv channels, suggesting that its effect on Kv channels occurs in a use (state)-independent manner. Pretreatment with DPO-1 (Kv1.5 subtype inhibitor) reduced the olanzapine-induced inhibition of Kv currents. In addition, pretreatment with guangxitoxin (Kv2.1 subtype inhibitor) and linopirdine (Kv7 subtype inhibitor) partially decreased the degree of Kv current inhibition. Olanzapine induced membrane depolarization. CONCLUSION: From these results, we suggest that olanzapine inhibits the Kv channels in a concentration-dependent, but state-independent, manner by affecting the gating properties of Kv channels. The primary Kv channel target of olanzapine is the Kv1.5 subtype.


Subject(s)
Antipsychotic Agents/pharmacology , Kv1.5 Potassium Channel/antagonists & inhibitors , Olanzapine/pharmacology , Potassium Channel Blockers/pharmacology , Animals , Antipsychotic Agents/administration & dosage , Coronary Vessels/cytology , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Olanzapine/administration & dosage , Potassium Channel Blockers/administration & dosage , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Rabbits
6.
Physiol Res ; 70(4): 649-953, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34062081

ABSTRACT

Previously, we demonstrated that supplementation of resuscitation fluids with the Kv7 voltage-activated potassium channel inhibitor linopirdine reduces fluid resuscitation requirements and stabilizes hemodynamics in various rat models of hemorrhagic shock. To further evaluate the therapeutic potential of linopirdine, we tested the effects of linopirdine-supplemented resuscitation fluids in a rat model of ischemia-reperfusion injury-induced acute respiratory distress syndrome (ARDS). Ventilated rats underwent unilateral lung ischemia from t=0-75 min, followed by lung reperfusion and fluid resuscitation to a mean arterial blood pressure of 60 mmHg with normal saline (NS, n=9) or NS supplemented with 50 µg/ml linopridine (NS-L), n=7) until t=360 min. As compared with NS, fluid resuscitation with NS-L stabilized blood pressure and reduced fluid requirements by 40% (p<0.05 vs. NS at t=240-360 min). While NS-L did not affect ARDS development, it reduced mortality from 66% with NS to 14% with NS-L (p=0.03, hazard ratio 0.14; 95% confidence interval of the hazard ratio: 0.03-0.65). Median survival time was 240 min with NS and >360 min with NS-L. As compared with NS treated animals that survived the observation period (n=3), however, plasma lactate and creatinine concentrations at t=360 min were higher with NS-L (n=6; p<0.05). Our findings extend therapeutic potential of NS-L from hypovolemic/hemorrhagic shock to hemodynamic instability under normovolemic conditions during organ ischemia-reperfusion injury. Possible adverse effects of NS-L, such as impairment of renal function and/or organ hypoperfusion, require further evaluation in long-term pre-clinical models.


Subject(s)
Fluid Therapy , Indoles/administration & dosage , Potassium Channel Blockers/administration & dosage , Pyridines/administration & dosage , Reperfusion Injury/complications , Respiratory Distress Syndrome/therapy , Resuscitation , Saline Solution/administration & dosage , Animals , Blood Pressure/drug effects , Disease Models, Animal , Infusions, Intravenous , Male , Rats, Sprague-Dawley , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/physiopathology , Time Factors
7.
Int J Cardiol ; 324: 255-260, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33075384

ABSTRACT

The antiretroviral drug lopinavir/ritonavir has been recently repurposed for the treatment of COVID-19. Its empirical use has been associated with multiple cardiac adverse reactions pertaining to its ancillary multi-channel blocking properties, vaguely characterized until now. We aimed to characterize qualitatively the cardiotoxicity associated with lopinavir/ritonavir in the setting of COVID-19. Spontaneous notifications of cardiac adverse drug reactions reported to the national Pharmacovigilance Network were collected for 8 weeks since March 1st 2020. The Nice Regional Center of Pharmacovigilance, whose scope of expertise is drug-induced long QT syndrome, analyzed the cases, including the reassessment of all available ECGs. QTc ≥ 500 ms and delta QTc > 60 ms from baseline were deemed serious. Twenty-two cases presented with 28 cardiac adverse reactions associated with the empirical use of lopinavir/ritonavir in a hospital setting. Most adverse reactions reflected lopinavir/ritonavir potency to block voltage-gated potassium channels with 5 ventricular arrhythmias and 17 QTc prolongations. An average QTc augmentation of 97 ± 69 ms was reported. Twelve QTc prolongations were deemed serious. Other cases were likely related to lopinavir/ritonavir potency to block sodium channels: 1 case of bundle branch block and 5 recurrent bradycardias. The incidence of cardiac adverse reactions of lopinavir/ritonavir was estimated between 0.3% and 0.4%. These cardiac adverse drug reactions offer a new insight in its ancillary multi-channel blocking functions. Lopinavir/ritonavir cardiotoxicity may be of concern for its empirical use during the COVID-19 pandemic. Caution should be exerted relative to this risk where lopinavir/ritonavir summary of product characteristics should be implemented accordingly.


Subject(s)
COVID-19 Drug Treatment , COVID-19/epidemiology , Cardiotoxicity/epidemiology , Lopinavir/administration & dosage , Lopinavir/adverse effects , Pharmacovigilance , Ritonavir/administration & dosage , Ritonavir/adverse effects , Aged , Aged, 80 and over , COVID-19/diagnosis , Cardiotoxicity/diagnosis , Drug Combinations , Electrocardiography/drug effects , Electrocardiography/trends , Female , France/epidemiology , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/adverse effects , Humans , Long QT Syndrome/chemically induced , Long QT Syndrome/diagnosis , Long QT Syndrome/epidemiology , Male , Middle Aged , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/adverse effects
8.
Curr Pain Headache Rep ; 24(12): 77, 2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33270149

ABSTRACT

PURPOSE OF REVIEW: Migraine remains a challenging condition to treat, thus highlighting the need for a better understanding of its molecular mechanisms. This review intends to unravel a new emerging target in migraine pathophysiology, the adenosine 5'-triphosphate-sensitive K+ (KATP) channel. RECENT FINDINGS: KATP channel is a common denominator in the cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) mediated intracellular cascades, both of which are involved in migraine. Intravenous infusion of KATP channel opener, levcromakalim, provoked migraine attack associated with dilation of extracerebral arteries in all persons with migraine. Preclinical and clinical studies implicate KATP channels in migraine initiation. KATP channel is a novel therapeutic target for the acute and preventive treatment of migraine. Future studies are warranted to provide a better understanding of the role of KATP channel subgroups in migraine.


Subject(s)
KATP Channels/agonists , KATP Channels/antagonists & inhibitors , Migraine Disorders/drug therapy , Migraine Disorders/physiopathology , Potassium Channel Blockers/administration & dosage , Animals , Bronchodilator Agents/adverse effects , Cromakalim/adverse effects , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Drug Delivery Systems/methods , Humans , KATP Channels/metabolism , Migraine Disorders/chemically induced , Migraine Disorders/metabolism
9.
Can J Physiol Pharmacol ; 98(11): 753-762, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33095677

ABSTRACT

The aim of this study was to examine if the peripheral antinociceptive effects of the opioid agonist/antagonist nalbuphine and buprenorphine involve the sequential participation of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) synthesis followed by K+ channel opening in the formalin test. Wistar rats (180-220 g) were injected in the dorsal surface of the right hind paw with formalin (1%). Rats received a subcutaneous (s.c.) injection into the dorsal surface of the paw of vehicles or increasing doses of nalbuphine (50-200 µg/paw) or buprenorphine (1-5 µg/paw) 20 min before formalin injection into the paw. Nalbuphine antinociception was reversed by the s.c. injection into the paw of the inhibitor of NO synthesis (NG-nitro-l-arginine methyl ester (L-NAME)), by the inhibitor of guanylyl cyclase (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ)), by the Kir6.1-2, ATP-sensitive K+ channel inhibitors (glibenclamide and glipizide), by the KCa2.1-3, small conductance Ca2+-activated K+ channel blocker (apamin), by the KCa1.1, large conductance Ca2+-activated K+ channel blocker (charybdotoxin), and by the KV, voltage-dependent K+ channel inhibitors (4-aminopyridine (4-AP) and tetraethylammonium chloride (TEA)). The antinociceptive effect produced by buprenorphine was blocked by the s.c. injection of 4-AP and TEA but not by L-NAME, ODQ, glibenclamide, glipizide, apamin, or charybdotoxin. The present results provide evidence for differences in peripheral mechanisms of action between these opioid drugs.


Subject(s)
Analgesics, Opioid/pharmacology , Narcotic Antagonists/pharmacology , Nociception/drug effects , Pain/drug therapy , Signal Transduction/drug effects , Animals , Buprenorphine/pharmacology , Cyclic GMP/metabolism , Disease Models, Animal , Glyburide/administration & dosage , Humans , Injections, Subcutaneous , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Male , NG-Nitroarginine Methyl Ester/administration & dosage , Nalbuphine/pharmacology , Nitric Oxide/metabolism , Nociception/physiology , Pain/chemically induced , Pain/diagnosis , Pain Measurement , Potassium Channel Blockers/administration & dosage , Rats , Receptors, Opioid/metabolism , Signal Transduction/physiology
10.
Biochem Biophys Res Commun ; 532(2): 265-270, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32863001

ABSTRACT

Methotrexate (MTX) has been widely used for the treatment of many types of autoimmune diseases, such as rheumatoid arthritis, psoriasis and dermatomyositis. However, its pharmacological mechanism is still unclear completely. In this study, we found that MTX is a potent and selective inhibitor of the Kv1.3 channel, a class of potassium channels highly associated with autoimmune diseases. Electrophysiological experiments showed that MTX inhibited human Kv1.3 channel with an IC50 of 41.5 ± 24.9 nM, and 1 µM MTX inhibited 32.6 ± 1.3% and 25.6 ± 2.2% of human Kv1.1 and Kv1.2 channel currents, respectively. These data implied the unique selectivity of MTX towards the Kv1.3 channel. Excitingly, using channel activation and chimeric experiments, we found that MTX bound to the outer pore region of Kv1.3 channel. Mutagenesis experiments in the Kv.3 channel extracellular pore region further showed that the Dsp371, Thr373 and His399 residues of outer pore region of Kv1.3 channel played important roles in MTX inhibiting activities. In conclusion, MTX inhibited Kv1.3 channel by targeting extracellular pore region, which is different form all the report small molecules, such as PAP-1 and 4-AP, but similar with many natural animal toxin peptides, such as ChTX, ShK and BmKTX. To the best of our knowledge, MTX is the first small molecular scaffold targeting the Kv1.3 channel extracellular pore region, suggesting its potential applications for designing novel Kv1.3 lead drugs and treating Kv1.3 channel-associated autoimmune diseases.


Subject(s)
Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Methotrexate/chemistry , Methotrexate/pharmacology , Potassium Channel Blockers/pharmacology , Binding Sites , Dose-Response Relationship, Drug , ERG1 Potassium Channel/antagonists & inhibitors , ERG1 Potassium Channel/metabolism , HEK293 Cells , Humans , Kv1.3 Potassium Channel/genetics , Methotrexate/administration & dosage , Mutagenesis , Patch-Clamp Techniques , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/chemistry
11.
Drug Des Devel Ther ; 14: 2549-2560, 2020.
Article in English | MEDLINE | ID: mdl-32669836

ABSTRACT

PURPOSE: The mechanism of cardioprotection by Kv7.1-5 (KCNQ1-5) channels inhibition by XE991 is unclear. We examined the impact of administration time on the cardioprotective efficacy of XE991, the involvement of key pro-survival kinases, and the importance of the Kv7 subchannels. METHODS: Isolated perfused rat hearts were divided into five groups: 1) vehicle, 2) pre-, 3) post- or 4) pre- and post-ischemic administration of XE991 or 5) chromanol 293B (Kv7.1 inhibitor) followed by infarct size quantification. HL-1 cells undergoing simulated ischemia/reperfusion were exposed to either a) vehicle, b) pre-, c) per-, d) post-ischemic administration of XE991 or pre-, per- and post-ischemic administration of e) XE991, f) Chromanol 293B or g) HMR1556 (Kv7.1 inhibitor). HL-1 cell injury was evaluated by propidium iodide/Hoechst staining. Pro-survival kinase activation of Akt, Erk and STAT3 in XE991-mediated HL-1 cell protection was evaluated using phosphokinase inhibitors. Kv7 subtype expression was examined by RT-PCR and qPCR. RESULTS: XE991, but not Chromanol 293B, reduced infarct size and improved hemodynamic recovery in all isolated heart groups. XE991 protected HL-1 cells when administered during simulated ischemia. Minor activation of the survival kinases was observed in cells exposed to XE991 but pharmacological inhibition of kinase activation did not reduce XE991-mediated protection. Kv7 subchannels 1-5 were all present in rat hearts but predominately Kv7.1 and Kv7.4 were present in HL-1 cells and selective Kv7.1 did not reduce ischemia/reperfusion injury. CONCLUSION: The cardioprotective efficacy of XE991 seems to depend on its presence during ischemia and early reperfusion and do not rely on RISK (p-Akt and p-Erk) and SAFE (p-STAT3) pathway activation. The protective effect of XE991 seems mainly mediated through the Kv7.4 subchannel.


Subject(s)
Chromans/pharmacology , KCNQ1 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Protective Agents/pharmacology , Sulfonamides/pharmacology , Animals , Chromans/administration & dosage , Male , Potassium Channel Blockers/administration & dosage , Protective Agents/administration & dosage , Rats , Rats, Wistar , Sulfonamides/administration & dosage , Time Factors
12.
Glia ; 68(10): 2119-2135, 2020 10.
Article in English | MEDLINE | ID: mdl-32220118

ABSTRACT

Spinal microglia change their phenotype and proliferate after nerve injury, contributing to neuropathic pain. For the first time, we have characterized the electrophysiological properties of microglia and the potential role of microglial potassium channels in the spared nerve injury (SNI) model of neuropathic pain. We observed a strong increase of inward currents restricted at 2 days after injury associated with hyperpolarization of the resting membrane potential (RMP) in microglial cells compared to later time-points and naive animals. We identified pharmacologically and genetically the current as being mediated by Kir2.1 ion channels whose expression at the cell membrane is increased 2 days after SNI. The inhibition of Kir2.1 with ML133 and siRNA reversed the RMP hyperpolarization and strongly reduced the currents of microglial cells 2 days after SNI. These electrophysiological changes occurred coincidentally to the peak of microglial proliferation following nerve injury. In vitro, ML133 drastically reduced the proliferation of BV2 microglial cell line after both 2 and 4 days in culture. In vivo, the intrathecal injection of ML133 significantly attenuated the proliferation of microglia and neuropathic pain behaviors after nerve injury. In summary, our data implicate Kir2.1-mediated microglial proliferation as an important therapeutic target in neuropathic pain.


Subject(s)
Cell Proliferation/physiology , Microglia/metabolism , Neuralgia/metabolism , Potassium Channel Blockers/administration & dosage , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Spinal Cord/metabolism , Animals , Cell Line, Transformed , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Injections, Spinal , Male , Mice , Mice, Transgenic , Microglia/drug effects , Neuralgia/prevention & control , Phenanthrolines/administration & dosage , Potassium Channels, Inwardly Rectifying/biosynthesis , Spinal Cord/cytology , Spinal Cord/drug effects
13.
Mult Scler Relat Disord ; 40: 101971, 2020 May.
Article in English | MEDLINE | ID: mdl-32062444

ABSTRACT

BACKGROUND: Modified-release 4-aminopyridine (fampridine-MR) is used in the symptomatic treatment of walking disability in patients with multiple sclerosis (MS).  Its potential for use in other MS symptoms remains unproven and its mode of action in this context is uncertain. Interest is growing in the use of upper limb outcome measures in clinical trials in patients with Multiple Sclerosis, particularly in advanced or progressive disease.  This study tests the following hypotheses: (1) Fampridine-MR improves upper limb function in patients with MS and upper limb impairment.  (2) Treatment with fampridine-MR is associated with measurable alterations in objective electrophysiological parameters (evoked potentials and transcranial magnetic stimulation (TMS)) which may predict response to drug treatment. METHODS: Study population: patients with MS of any disease subtype, duration and severity who have symptomatic impairment of one or both upper limbs.  A healthy control group was included for validation of clinical and electrophysiological measures.  Study design: randomised double blind placebo-controlled trial.  Treatment details: participants allocated to either fampridine-MR 10 mg bd or placebo of identical appearance for 8 weeks.  Primary outcome: performance on 9-hole peg test (9HPT) after 4 weeks.  Secondary outcomes: persistence of effect on 9HPT; grip strength; visual acuity and contrast sensitivity; modified fatigue impact scale score; sensory discrimination capacity; visual, somatosensory and motor evoked potentials; resting motor threshold; paired-pulse TMS; peripheral nerve conduction studies. RESULTS: 40 patients with MS (60% female, median age 52, median disease duration 13.5 years, median EDSS 6.0) were enrolled.  Treatment with fampridine-MR was not associated with any change in upper limb function as measured by the clinical primary or secondary outcomes.  Treatment with fampridine-MR was also not associated with any difference in electrophysiological measures of upper limb function.  This held true after adjustment for hand dominance, disease duration and severity.  Four patients withdrew from the trial because of lack of efficacy or side-effects; all were in the placebo arm.  Three patients were admitted to hospital during the study period; one with MS exacerbation (placebo group), one with syncope (drug group) and one with UTI (drug group); otherwise there were no serious adverse events. CONCLUSION: Treatment with fampridine-MR was well-tolerated but did not produce clinical benefit in terms of upper limb function, vision or fatigue, nor was there any measurable effect on objective electrophysiological parameters.


Subject(s)
4-Aminopyridine/pharmacology , Electrophysiological Phenomena/drug effects , Fatigue/drug therapy , Multiple Sclerosis/drug therapy , Potassium Channel Blockers/pharmacology , Upper Extremity/physiopathology , Vision Disorders/drug therapy , 4-Aminopyridine/administration & dosage , Adult , Double-Blind Method , Fatigue/etiology , Female , Humans , Male , Middle Aged , Multiple Sclerosis/complications , Outcome Assessment, Health Care , Potassium Channel Blockers/administration & dosage , Vision Disorders/etiology
14.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R390-R398, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31913684

ABSTRACT

Local skin heating to 42°C causes cutaneous thermal hyperemia largely via nitric oxide (NO) synthase (NOS)-related mechanisms. We assessed the hypothesis that ATP-sensitive K+ (KATP) channels interact with NOS to mediate cutaneous thermal hyperemia. In 13 young adults (6 women, 7 men), cutaneous vascular conductance (CVC) was measured at four intradermal microdialysis sites that were continuously perfused with 1) lactated Ringer solution (control), 2) 5 mM glibenclamide (KATP channel blocker), 3) 20 mM NG-nitro-l-arginine methyl ester (NOS inhibitor), or 4) a combination of KATP channel blocker and NOS inhibitor. Local skin heating to 42°C was administered at all four treatment sites to elicit cutaneous thermal hyperemia. Thirty minutes after the local heating, 1.25 mM pinacidil (KATP channel opener) and subsequently 25 mM sodium nitroprusside (NO donor) were administered to three of the four sites (each 25-30 min). The local heating-induced prolonged elevation in CVC was attenuated by glibenclamide (19%), but the transient initial peak was not. However, glibenclamide had no effect on the prolonged elevation in CVC in the presence of NOS inhibition. Pinacidil caused an elevation in CVC, but this response was abolished at the glibenclamide-treated skin site, demonstrating its effectiveness as a KATP channel blocker. The pinacidil-induced increase in CVC was unaffected by NOS inhibition, whereas the increase in CVC elicited by sodium nitroprusside was partly (15%) inhibited by glibenclamide. In summary, we showed an interactive effect of KATP channels and NOS for the plateau of cutaneous thermal hyperemia. This interplay may reflect a vascular smooth muscle cell KATP channel activation by NO.


Subject(s)
Hyperemia/enzymology , KATP Channels/metabolism , Microcirculation , Microvessels/enzymology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Skin/blood supply , Vasodilation , Adult , Blood Flow Velocity , Enzyme Inhibitors/administration & dosage , Female , Humans , Hyperemia/etiology , Hyperemia/physiopathology , Hypothermia, Induced , Ion Channel Gating , KATP Channels/antagonists & inhibitors , Male , Microcirculation/drug effects , Microvessels/drug effects , Microvessels/physiopathology , Nitric Oxide Donors/administration & dosage , Nitric Oxide Synthase/antagonists & inhibitors , Potassium Channel Blockers/administration & dosage , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/administration & dosage , Young Adult
15.
Behav Brain Res ; 378: 112315, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31654662

ABSTRACT

Schizophrenia is a major psychiatric disorder associated with positive and negative symptoms and cognitive impairments. In this study, we used animal models of behavior to evaluate the antipsychotic activity of ASP2905, a potent and selective inhibitor of the potassium channel Kv12.2 encoded by the Kcnh3/BEC1 gene. ASP2905 inhibited hyperlocomotion induced by methamphetamine and by phencyclidine. In contrast, ASP2905 did not affect spontaneous locomotion, suggesting that ASP2905 selectively inhibits abnormal behaviors induced by stimulants. Chronic infusion of ASP2905 significantly ameliorated phencyclidine-induced prolongation of immobility time in mice subjected to the forced swimming test. These findings suggest that ASP2905 potentially mitigates symptoms of schizophrenia, such as apathy. The antipsychotic clozapine also reversed phencyclidine-induced prolonged immobility, while risperidone and haloperidol had no effect. Assessment of the effects of ASP2905 on latent learning deficits in mice treated with phencyclidine as neonates subjected to the water-finding task showed that ASP2905 significantly ameliorated phencyclidine-induced prolongation of finding latency, which reflects latent learning performance. These findings suggest that ASP2905 potentially mitigates cognitive impairments caused by schizophrenia, such as attention deficits. In contrast, administration of clozapine did not ameliorate phencyclidine-induced prolongation of finding latency. Therefore, ASP2905 may alleviate the broad spectrum of symptoms of schizophrenia, including positive and negative symptoms and cognitive impairments, which is in contrast to currently available antipsychotics, which are generally only partially effective for ameliorating these symptoms.


Subject(s)
Antipsychotic Agents/pharmacology , Cognitive Dysfunction/drug therapy , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Hyperkinesis/drug therapy , Learning/drug effects , Locomotion/drug effects , Potassium Channel Blockers/pharmacology , Pyrimidines/pharmacology , Schizophrenia/drug therapy , Triazines/pharmacology , Animals , Antipsychotic Agents/administration & dosage , Behavior, Animal/drug effects , Cognitive Dysfunction/etiology , Disease Models, Animal , Hyperkinesis/chemically induced , Male , Memory, Short-Term , Mice , Potassium Channel Blockers/administration & dosage , Pyrimidines/administration & dosage , Schizophrenia/chemically induced , Schizophrenia/complications , Triazines/administration & dosage
16.
Mult Scler Relat Disord ; 38: 101489, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31731213

ABSTRACT

BACKGROUND: Fampridine is an effective treatment to improve ambulation for some multiple sclerosis (MS) patients. Remarkable discrepancies exist between responder rates in clinical trials and the proportion of patients continuing treatment in clinical practice. This may be related to clinical phenotypes of MS patients, and the influence of patient reported outcome (PRO) on treatment decision making. OBJECTIVE: To analyse responder rates to fampridine on ambulation and upper extremity function (UEF) and the influence on treatment decision making in different clinical subgroups in a real-world setting. METHODS: MS patients with ambulatory impairment treated with fampridine were included. Patients were subdivided based on disease duration, clinical phenotype, Expanded Disability Status Scale (EDSS), baseline walking speed, and presence of UEF impairment. Ambulatory response was assessed with the Timed 25-Foot Walk (T25FW, responder defined as ≥20% improvement) and with the MS Walking Scale (MSWS, responder defined as ≥8 points improvement) as a PRO. For patients also reporting impaired UEF, the Arm Function in MS Questionnaire (AMSQ, responder defined as ≥15 improvement) was the PRO of choice. Decision on treatment continuation was based on improvement of T25FW, MSWS and the clinicians' overall impression for improvement. RESULTS: In total 344 patients were included of which 75.3% continued treatment. More patients with a relapsing clinical phenotype continued treatment vs patients with a progressive phenotype (83.6 vs 68.6%, p < 0.01). A positive linear trend was found between severity of walking disability, as determined by baseline walking speed, and T25FW response (p < 0.01), while there was an inverse linear association between walking disability and MSWS response (p = 0.03). However, the proportion of patients continuing treatment was similar between subgroups of baseline walking speed. Impaired UEF was reported by 183 (66.5%) patients, of which 64 (39.3%) were AMSQ responders. Patients responding on AMSQ compared to non-responders, were also more frequently MSWS responders (82.8 vs 65.3%, p = 0.02), while response on T25FW was similar, and continued treatment more often (85.9 vs 70.7%, p = 0.04). This suggests an influence of PRO on treatment decision making. CONCLUSION: Responder rates and treatment continuation of fampridine differed between clinical subgroups of MS. PROs influenced treatment decision making of fampridine in clinical practice, particularly in patients with mild ambulatory impairment or those reporting UEF impairment. To some extent, these findings explain discrepancies found between clinical trials and clinical practice, and support the importance of subgroup analyses and incorporation of PROs in clinical trials. For clinical practice, using PROs to assess patients experience in conjunction with performance measures helps in treatment decision making.


Subject(s)
4-Aminopyridine/pharmacology , Clinical Decision-Making , Multiple Sclerosis, Chronic Progressive/drug therapy , Multiple Sclerosis, Chronic Progressive/physiopathology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Patient Reported Outcome Measures , Potassium Channel Blockers/pharmacology , Severity of Illness Index , 4-Aminopyridine/administration & dosage , Adult , Female , Follow-Up Studies , Humans , Male , Middle Aged , Mobility Limitation , Phenotype , Potassium Channel Blockers/administration & dosage , Upper Extremity/physiopathology , Walking Speed/physiology
17.
Expert Rev Clin Pharmacol ; 12(11): 1013-1018, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31639317

ABSTRACT

Introduction: Lambert-Eaton myasthenic syndrome is an autoimmune disease of the neuromuscular junction characterized by a presynaptic defect of neuromuscular transmission resulting in muscle weakness and fatigability. Diagnostic features are specific neurophysiological alterations and autoantibody detection. The present review is focused on the use of Amifampridine Phosphate to treat LEMS patients.Areas covered: Medline search from 1990 to 2019 was examined using the free subject terms: Lambert-Eaton myasthenic syndrome, LEMS, Amifampridine, 3,4-diaminopyridine, which were then combined with Treatment, Therapy, Clinical Trial, Controlled Clinical Trial, Randomized Clinical Trial and Cochrane Review. The author has done a supervised analysis of the retrieved articles and focused on those subjectively evaluated as most relevant.Expert commentary: Data from randomized clinical trials and case series have demonstrated that Lambert-Eaton myasthenic syndrome symptoms were successfully treated by Amifampridine Phosphate. Hence, the drug represents a substantial step forward in the symptomatic treatment of the disease due to its efficacy, safety and reliable GMP formulation. As Amifampridine Phosphate works by enhancing the release of acetylcholine at the neuromuscular junction by blocking K+ efflux at the pre-synaptic membrane, it is also conceivable to use it for other diseases of the neuromuscular junction in which such an effect is searched for.


Subject(s)
Amifampridine/administration & dosage , Lambert-Eaton Myasthenic Syndrome/drug therapy , Neuromuscular Agents/administration & dosage , Amifampridine/adverse effects , Amifampridine/pharmacology , Humans , Lambert-Eaton Myasthenic Syndrome/physiopathology , Neuromuscular Agents/adverse effects , Neuromuscular Agents/pharmacology , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/adverse effects , Potassium Channel Blockers/pharmacology , Randomized Controlled Trials as Topic , Tablets
18.
J Neurol ; 266(Suppl 1): 93-100, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31270663

ABSTRACT

Vestibulo-ocular reflexes (VOR) are mediated by frequency-tuned pathways that separately transform the different dynamic and static aspects of head motion/position-related sensory signals into extraocular motor commands. Voltage-dependent potassium conductances such as those formed by Kv1.1 are important for the ability of VOR circuit elements to encode highly transient motion components. Here we describe the impact of the Kv1.1 channel blocker 4-aminopyridine (4-AP) on spontaneous and motion-evoked discharge of superior oblique motoneurons. Spike activity was recorded from the motor nerve in isolated preparations of Xenopus laevis tadpoles. Under static conditions, bath application of 1-10 µM 4-AP increased the spontaneous firing rate and provoked repetitive bursts of spikes. During motion stimulation 4-AP also augmented and delayed the peak firing rate suggesting that this drug affects the magnitude and timing of vestibular-evoked eye movements. The exclusive Kv1.1 expression in thick vestibular afferent fibers in larval Xenopus at this developmental stage suggests that the altered extraocular motor output in the presence of 4-AP mainly derives from a firing rate increase of irregular firing vestibular afferents that propagates along the VOR circuitry. Clinically and pharmacologically, the observed 4-AP-mediated increase of peripheral vestibular input under resting and dynamic conditions can contribute to the observed therapeutic effects of 4-AP in downbeat and upbeat nystagmus as well as episodic ataxia type 2, by an indirect increase of cerebellar Purkinje cell discharge.


Subject(s)
4-Aminopyridine/administration & dosage , Eye Movements/drug effects , Potassium Channel Blockers/administration & dosage , Reflex, Vestibulo-Ocular/drug effects , Vestibule, Labyrinth/drug effects , Animals , Eye Movements/physiology , Female , Male , Reflex, Vestibulo-Ocular/physiology , Vestibule, Labyrinth/physiology , Xenopus laevis
19.
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
20.
Article in English | MEDLINE | ID: mdl-31207562

ABSTRACT

Talatisamine, as the efficacy ingredient of Aconitum, was known as a novel specific blocker for the delayed rectifier K+ channels in rat hippocampal neurons. In this study, a rapid, selective and reproducible UPLC-MS/MS separation method was established and fully validated for the quantitative determination of talatisamine levels in ICR (Institute of Cancer Research) mouse blood. A total of 24 healthy male ICR mice were divided into four groups that was administered talatisamine via intravenous at a dose of 1 mg/kg and oral administration of three doses (2, 4, 8 mg/kg). All blood samples were protein precipitate by using acetonitrile with an internal standard (IS) deltaline. The effective chromatographic separation was carried out through an UPLC BEH C18 analytical column (2.1 mm × 50 mm, 1.7 µm) with an initial mobile phase that consisted of acetonitrile and 10 mmol/L ammonium acetate aqueous solution (containing 0.1% formic acid) with a gradient elution pumped at a flow rate of 0.4 mL/min. Also, an electrospray ionization (ESI) was applied to quantify the talatisamine in the positive ions mode. The method validation demonstrated good linearity over the range of 1-1000 ng/mL (r2 ≥ 0.9993) for talatisamine in mouse blood with a lower limit of quantification (LLOQ) at 1 ng/mL. The accuracy values of the method were within 89.4% to 113.3%, and the matrix effects were between 103.2% and 106.3%. The mean extraction recoveries for talatisamine obtained from four concentrations of QC blood samples were exceeded 71.7%, and the relative standard deviation (RSD) both of intra- and inter-day precision values for replicate quality control samples did not exceed 15% respectively for all analytes during the assay validation. This method was successfully applied to the evaluation of the pharmacokinetic of talatisamine, regardless of intragastric or intravenous administration in mice. Based on the pharmacokinetics data, the bioavailability of talatisamine in mice was >65.0% after oral administration, exhibiting an excellent oral absorption.


Subject(s)
Aconitine/analogs & derivatives , Chromatography, Liquid/methods , Potassium Channel Blockers/pharmacokinetics , Tandem Mass Spectrometry/methods , Aconitine/administration & dosage , Aconitine/blood , Aconitine/pharmacokinetics , Animals , Male , Mice , Mice, Inbred ICR , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/blood , Potassium Channels
SELECTION OF CITATIONS
SEARCH DETAIL