Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 559
Filtrar
1.
Molecules ; 29(12)2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38931004

RESUMEN

Potassium channels have recently emerged as suitable target for the treatment of epileptic diseases. Among potassium channels, KCNT1 channels are the most widely characterized as responsible for several epileptic and developmental encephalopathies. Nevertheless, the medicinal chemistry of KCNT1 blockers is underdeveloped so far. In the present review, we describe and analyse the papers addressing the issue of KCNT1 blockers' development and identification, also evidencing the pros and the cons of the scientific approaches therein described. After a short introduction describing the epileptic diseases and the structure-function of potassium channels, we provide an extensive overview of the chemotypes described so far as KCNT1 blockers, and the scientific approaches used for their identification.


Asunto(s)
Química Farmacéutica , Epilepsia , Bloqueadores de los Canales de Potasio , Humanos , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Química Farmacéutica/métodos , Epilepsia/tratamiento farmacológico , Epilepsia/metabolismo , Relación Estructura-Actividad , Animales , Anticonvulsivantes/química , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Canales de Potasio de Dominio Poro en Tándem/química , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de potasio activados por Sodio
2.
J Med Chem ; 67(11): 9731-9744, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38807539

RESUMEN

Recent literature reports highlight the importance of the renal outer medullary potassium (ROMK) channel in renal sodium and potassium homeostasis and emphasize the potential impact that ROMK inhibitors could have as a novel mechanism diuretic in heart failure patients. A series of piperazine-based ROMK inhibitors were designed and optimized to achieve excellent ROMK potency, hERG selectivity, and ADME properties, which led to the identification of compound 28 (BMS-986308). BMS-986308 demonstrated efficacy in the volume-loaded rat diuresis model as well as promising in vitro and in vivo profiles and was therefore advanced to clinical development.


Asunto(s)
Insuficiencia Cardíaca , Bloqueadores de los Canales de Potasio , Animales , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Ratas , Bloqueadores de los Canales de Potasio/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/farmacocinética , Bloqueadores de los Canales de Potasio/síntesis química , Relación Estructura-Actividad , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/metabolismo , Descubrimiento de Drogas , Diuresis/efectos de los fármacos , Piperazinas/farmacología , Piperazinas/química , Piperazinas/uso terapéutico , Piperazinas/síntesis química , Piperazinas/farmacocinética , Masculino , Ratas Sprague-Dawley
3.
Biomed Pharmacother ; 175: 116651, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692062

RESUMEN

Voltage-gated potassium channel 1.3 (Kv1.3) has emerged as a pivotal player in numerous biological processes and pathological conditions, sparking considerable interest as a potential therapeutic target across various diseases. In this review, we present a comprehensive examination of Kv1.3 channels, highlighting their fundamental characteristics and recent advancements in utilizing Kv1.3 inhibitors for treating autoimmune disorders, neuroinflammation, and cancers. Notably, Kv1.3 is prominently expressed in immune cells and implicated in immune responses and inflammation associated with autoimmune diseases and chronic inflammatory conditions. Moreover, its aberrant expression in certain tumors underscores its role in cancer progression. While preclinical studies have demonstrated the efficacy of Kv1.3 inhibitors, their clinical translation remains pending. Molecular imaging techniques offer promising avenues for tracking Kv1.3 inhibitors and assessing their therapeutic efficacy, thereby facilitating their development and clinical application. Challenges and future directions in Kv1.3 inhibitor research are also discussed, emphasizing the significant potential of targeting Kv1.3 as a promising therapeutic strategy across a spectrum of diseases.


Asunto(s)
Canal de Potasio Kv1.3 , Neoplasias , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/metabolismo , Animales , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Bloqueadores de los Canales de Potasio/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/metabolismo , Terapia Molecular Dirigida , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo
4.
Int J Mol Sci ; 25(3)2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-38338871

RESUMEN

Peripheral cytokine levels may serve as biomarkers for treatment response and disease monitoring in patients with multiple sclerosis (pwMS). The objectives were to assess changes in plasma biomarkers in PwMS after 14 days of fampridine treatment and to explore correlations between changes in performance measures and plasma biomarkers. We included 27 PwMS, 14 women and 13 men, aged 52.0 ± 11.6 years, with a disease duration of 17 ± 8.5 years, and an Expanded Disability Status Scale of 6 [IQR 5.0/6.5]. Gait and hand function were assessed using performance tests completed prior to fampridine and after 14 days of treatment. Venous blood was obtained, and chemiluminescence analysis conducted to assess plasma cytokines and neurodegenerative markers. All performance measures demonstrated improvements. Biomarkers showed decreased tumor necrosis factor (TNF) receptor-2 levels. Associations were found between change scores in (i) Six Spot Step Test and Interleukin (IL)-2, IL-8, and IL-17 levels; (ii) timed 25-foot walk and interferon-γ, IL-2, IL-8, TNF-α, and neurofilament light levels, and (iii) 12-Item Multiple Sclerosis Walking Scale and IL-17 levels. The associations may reflect increased MS-related inflammatory activity rather than a fampridine-induced response or that a higher level of inflammation induces a better response to fampridine.


Asunto(s)
Esclerosis Múltiple , Masculino , Humanos , Femenino , Esclerosis Múltiple/tratamiento farmacológico , Interleucina-17 , Bloqueadores de los Canales de Potasio/uso terapéutico , Interleucina-8 , Resultado del Tratamiento , 4-Aminopiridina/uso terapéutico
5.
Int J Mol Sci ; 24(22)2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-38003453

RESUMEN

Modulation of the human Ether-à-go-go-Related Gene (hERG) channel, a crucial voltage-gated potassium channel in the repolarization of action potentials in ventricular myocytes of the heart, has significant implications on cardiac electrophysiology and can be either antiarrhythmic or proarrhythmic. For example, hERG channel blockade is a leading cause of long QT syndrome and potentially life-threatening arrhythmias, such as torsades de pointes. Conversely, hERG channel blockade is the mechanism of action of Class III antiarrhythmic agents in terminating ventricular tachycardia and fibrillation. In recent years, it has been recognized that less proarrhythmic hERG blockers with clinical potential or Class III antiarrhythmic agents exhibit, in addition to their hERG-blocking activity, a second action that facilitates the voltage-dependent activation of the hERG channel. This facilitation is believed to reduce the proarrhythmic potential by supporting the final repolarizing of action potentials. This review covers the pharmacological characteristics of hERG blockers/facilitators, the molecular mechanisms underlying facilitation, and their clinical significance, as well as unresolved issues and requirements for research in the fields of ion channel pharmacology and drug-induced arrhythmias.


Asunto(s)
Canales de Potasio Éter-A-Go-Go , Bloqueadores de los Canales de Potasio , Humanos , Canal de Potasio ERG1 , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Antiarrítmicos/efectos adversos , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/tratamiento farmacológico , Miocitos Cardíacos , Potenciales de Acción
7.
Neurol Sci ; 44(9): 3059-3069, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37055710

RESUMEN

BACKGROUND: Gait imbalance is one of the frequent complications in subjects with multiple sclerosis (MS). Fampridine (4-aminopyridine) is a potassium-channel blocker that is administered for gait imbalance in MS. Different studies showed the effects of fampridine on gait status based on various tests in subjects with MS. Some showed significant improvement after treatment, and others did not. So, we designed this systematic review, and meta-analysis to estimate the pooled effects of fampridine on gait status in patients with MS. METHODS: The main goal is the evaluation of times of different gait test pre and post fampridine treatment. Two independent expert researchers conducted a systematic and comprehensive search in PubMed, Scopus, EMBASE, Web of Science, and Google Scholar and also gray literature, including references of the references and conference abstracts. The search was done on September 16, 2022. Before-after studies trials reporting scores of the walking tests. We extracted data regarding the total number of participants, first author, publication year, country of origin, mean age, Expanded Disability Status Scale (EDSS), and the results of walking tests. RESULTS: The literature search revealed 1963 studies; after deleting duplicates, 1098 studies remained. Seventy-seven full texts were evaluated. Finally, 18 studies were included for meta-analysis, while most of them were not placebo-controlled trials. The most frequent country of origin was Germany, and the mean age and EDSS ranged between 44 and 56 years and 4 and 6, respectively. The studies were published between 2013 and 2019. The pooled standardized mean difference (SMD) (after-before) of the MS Walking Scale (MSWS-12) was - 1.97 (95%CI: - 1.7, - 1.03) (I2 = 93.1%, P < 0.001). The pooled SMD (after-before) of the six-minute walk test (6MWT) was 0.49 (95%CI: 0.22, - 0.76) (I2 = 0%, P = 0.7). The pooled SMD (after-before) of T Timed 25-Foot Walk (T25FW) was - 0.99(95%CI: - 1.52, - 0.47) (I2 = 97.5%, P < 0.001). CONCLUSION: This systematic review and meta-analysis show that fampridine improves gait imbalance in patients with MS.


Asunto(s)
Esclerosis Múltiple , Humanos , Adulto , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/tratamiento farmacológico , Resultado del Tratamiento , 4-Aminopiridina/uso terapéutico , 4-Aminopiridina/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Marcha/fisiología , Caminata/fisiología
8.
Proc Natl Acad Sci U S A ; 120(2): e2211977120, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36595694

RESUMEN

Engineered microbes for the delivery of biologics are a promising avenue for the treatment of various conditions such as chronic inflammatory disorders and metabolic disease. In this study, we developed a genetically engineered probiotic delivery system that delivers a peptide to the intestinal tract with high efficacy. We constructed an inducible system in the probiotic Lactobacillus reuteri to secrete the Kv1.3 potassium blocker ShK-235 (LrS235). We show that LrS235 culture supernatants block Kv1.3 currents and preferentially inhibit human T effector memory (TEM) lymphocyte proliferation in vitro. A single oral gavage of healthy rats with LrS235 resulted in sufficient functional ShK-235 in the circulation to reduce inflammation in a delayed-type hypersensitivity model of atopic dermatitis mediated by TEM cells. Furthermore, the daily oral gavage of LrS235 dramatically reduced clinical signs of disease and joint inflammation in rats with a model of rheumatoid arthritis without eliciting immunogenicity against ShK-235. This work demonstrates the efficacy of using the probiotic L. reuteri as a novel oral delivery platform for the peptide ShK-235 and provides an efficacious strategy to deliver other biologics with great translational potential.


Asunto(s)
Artritis Reumatoide , Probióticos , Ratas , Humanos , Animales , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Péptidos/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Probióticos/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico
9.
Neurol Sci ; 44(1): 393-396, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36094772

RESUMEN

BACKGROUND: Fampridine is a potassium channel blocker drug used to improve walking ability in patients with multiple sclerosis (MS). We evaluated the effect of fampridine in patients with MS in the acute phase of transverse myelitis. METHODS: In a randomized, placebo-controlled trial, 30 patients who had their first episode of cervical myelitis with quadriparesis presentation, with the final diagnosis of MS, were randomly divided into two equal groups. The intervention group received intravenous methylprednisolone (IVMP) for 7 days plus fampridine. The placebo group received IVMP for 7 days plus placebo. To compare the treatment results, we compared the Barthel index (BI) scores of the groups at the start of the trial and the 21st day after the start of treatment. RESULTS: There was no significant difference in baseline characteristics between the intervention and placebo groups in terms of mean age, sex, and mean admission BI (p > 0.05). Mean (SD) admission BI in placebo and intervention groups was 27.20 (7.341) and 27.87(5.78), respectively (p = 0.784). The measured mean (SD) BI after treatment was 48.73 (15.54) in the placebo and 64.93 (11.81) in the intervention group (p = 0.003) after 3 weeks. CONCLUSION: Using fampridine plus IVMP in the acute phase of transverse myelitis in MS patients improved the disease's symptoms and increased the daily activity ability of patients.


Asunto(s)
Esclerosis Múltiple , Mielitis Transversa , Humanos , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/tratamiento farmacológico , Mielitis Transversa/complicaciones , Mielitis Transversa/tratamiento farmacológico , Mielitis Transversa/inducido químicamente , 4-Aminopiridina/uso terapéutico , Bloqueadores de los Canales de Potasio/uso terapéutico , Resultado del Tratamiento , Metilprednisolona/uso terapéutico , Método Doble Ciego
10.
Anal Chim Acta ; 1231: 340397, 2022 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-36220288

RESUMEN

Potassium ion channels are expressed on the cell membranes, implicated in wide variety of cell functions and intimately linked to cancer cell behaviors. This work reports the first bioplatform described to date allowing simple and rapid detection of ion channel activity and the effect of their inhibitors in cancer cells. The methodology involves interrogation of the channel of interest from cells specifically captured on magnetic immunoconjugates using specific detection antibodies that are labeled with horseradish peroxidase enzyme. The channel activity is reflected by an amperometric signal transduction of the resulting magnetic bioconjugates onto screen-printed carbon electrodes. The bioplatform feasibility was proven for the detection of the Kv channels in U87 human glioblastoma cells and their blocking by scorpion venom KAaH1 and KAaH2 peptides. The obtained results confirm the high sensitivity (detection of 5 U87 cells⋅mL-1 and 0.06 µg mL-1 of KAaH2) of the proposed bioplatform and their versatility to detect both potassium channel activity and their potential inhibitors, in a given cancer cell line, with high sensitivity in a simple and fast way. This bioplatform presents potential applications in cancer and theranostic of channelopathies.


Asunto(s)
Inmunoconjugados , Neoplasias , Venenos de Escorpión , Carbono , Peroxidasa de Rábano Silvestre , Humanos , Canales Iónicos , Neoplasias/tratamiento farmacológico , Péptidos , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio , Venenos de Escorpión/farmacología
11.
Pharm Pat Anal ; 11(2): 45-56, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35369761

RESUMEN

Slack channels are sodium-activated potassium channels that are encoded by the KCNT1 gene. Several KCNT1 gain of function mutations have been linked to malignant migrating partial seizures of infancy. Quinidine is an anti-arrhythmic drug that functions as a moderately potent inhibitor of Slack channels; however, quinidine use is limited by its poor selectivity, safety and pharmacokinetic profile. Slack channels represent an interesting target for developing novel therapeutics for the treatment of malignant migrating partial seizures of infancy and other childhood epilepsies; thus, ongoing efforts are directed toward the discovery of small-molecules that inhibit Slack currents. This review summarizes patent applications published in 2020-2021 that describe the discovery of novel small-molecule Slack inhibitors.


Asunto(s)
Epilepsia , Proteínas del Tejido Nervioso , Bloqueadores de los Canales de Potasio , Canales de potasio activados por Sodio , Niño , Epilepsia/tratamiento farmacológico , Humanos , Mutación , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de potasio activados por Sodio/antagonistas & inhibidores , Quinidina/uso terapéutico , Convulsiones/tratamiento farmacológico
13.
Clin Biomech (Bristol, Avon) ; 86: 105382, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34000628

RESUMEN

BACKGROUND: People with multiple sclerosis have reduced walking speed and impaired gait pattern. Prolonged release-fampridine is a potassium channel blocker that improves nerve conduction in patients with multiple sclerosis, leading to walking benefits. Whether fampridine alters gait pattern is unknown. METHODS: In this crossover, randomized controlled trial, patients with multiple sclerosis were tested for responder status during a 4-week run-in period. Patients were considered responders if they improved their 25-ft walk test by 10% and improved their perceived walking capacity. Responders were randomized to prolonged release-fampridine (10 mg b.i.d.) or placebo for a 6-week period. After a 2-week wash-out period, they were allocated to the other treatment for 6 weeks. Participants were assessed before and after both conditions. Three-dimensional gait analysis assessed kinematic, kinetic, mechanic and energetic variables while walking on a treadmill at comfortable speed. Six-minute walk test and 25-ft walk test were used to assess walking speed on middle and short-distances, respectively. Patient-reported outcome measures were also used. Repeated measures ANCOVAs were applied to assess the treatment effects. FINDINGS: Out of 39 included patients, 24 responders (12 women; Expanded Disability Status Scale:4.25[4-5]; age:46 ± 10 years; maximal speed:0.93 ± 0.38 m·s-1) were identified. Among them, prolonged release-fampridine reduced the external mechanical work (-0.039 J·kg-1·m-1;p = 0.02), and improved knee flexion during swing phase (+5.3°; p = 0.02). No differences were found in other walking tests and patient-reported outcomes, at group-level. INTERPRETATION: Prolonged release-fampridine increases knee flexion during swing phase and lowers mechanical external work. Whether these changes are related to clinically meaningful improvements in walking capacity and other functional variables should be further investigated.


Asunto(s)
Esclerosis Múltiple , 4-Aminopiridina/farmacología , 4-Aminopiridina/uso terapéutico , Adulto , Femenino , Marcha , Humanos , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Resultado del Tratamiento , Caminata
14.
Cell Physiol Biochem ; 55(S3): 131-144, 2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34043300

RESUMEN

The Kca3.1 channels, previously designated as IK1 or SK4 channels and encoded by the KCNN4 gene, are activated by a rise of the intracellular Ca2+ concentration. These K+ channels are widely expressed in many organs and involved in many pathologies. In particular, Kca3.1 channels have been studied intensively in the context of cancer. They are not only a marker and a valid prognostic tool for cancer patients, but have an important share in driving cancer progression. Their function is required for many characteristic features of the aggressive cancer cell behavior such as migration, invasion and metastasis as well as proliferation and therapy resistance. In the context of cancer, another property of Kca3.1 is now emerging. These channels can be a target for novel small molecule-based imaging probes, as it has been validated in case of fluorescently labeled senicapoc-derivatives. The aim of this review is (i) to give an overview on the role of Kca3.1 channels in cancer progression and in shaping the cancer microenvironment, (ii) discuss the potential of using Kca3.1 targeting drugs for cancer imaging, (iii) and highlight the possibility of combining molecular dynamics simulations to image inhibitor binding to Kca3.1 channels in order to provide a deeper understanding of Kca3.1 channel pharmacology. Alltogether, Kca3.1 is an attractive therapeutic target so that senicapoc, originally developed for the treatment of sickle cell anemia, should be repurposed for the treatment of cancer patients.


Asunto(s)
Acetamidas/uso terapéutico , Antineoplásicos/uso terapéutico , Calcio/metabolismo , Neoplasias/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Compuestos de Tritilo/uso terapéutico , Antineoplásicos/química , Antidrepanocíticos/química , Antidrepanocíticos/uso terapéutico , Sitios de Unión , Señalización del Calcio , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Simulación de Dinámica Molecular , Terapia Molecular Dirigida/métodos , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Bloqueadores de los Canales de Potasio/química , Estructura Secundaria de Proteína , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/química , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
15.
J Pharmacol Exp Ther ; 377(2): 265-272, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674391

RESUMEN

Drug-induced long QT syndrome (LQTS) is an established cardiac side effect of a wide range of medications and represents a significant concern for drug safety. The rapidly and slowly activating delayed rectifier K+ currents, mediated by channels encoded by the human ether-a-go-go-related gene (hERG) and KCNQ1 + KCNE1, respectively, are two main currents responsible for ventricular repolarization. The common cause for drugs to induce LQTS is through impairing the hERG channel. For the recent emergence of COVID-19, caused by severe acute respiratory syndrome coronavirus 2, several drugs have been investigated as potential therapies; however, there are concerns about their QT prolongation risk. Here, we studied the effects of chloroquine, hydroxychloroquine, azithromycin, and remdesivir on hERG channels. Our results showed that although chloroquine acutely blocked hERG current (IhERG), with an IC50 of 3.0 µM, hydroxychloroquine acutely blocked IhERG 8-fold less potently, with an IC50 of 23.4 µM. Azithromycin and remdesivir did not acutely affect IhERG When these drugs were added at 10 µM to the cell culture medium for 24 hours, remdesivir increased IhERG by 2-fold, which was associated with an increased mature hERG channel expression. In addition, these four drugs did not acutely or chronically affect KCNQ1 + KCNE1 channels. Our data provide insight into COVID-19 drug-associated LQTS and cardiac safety concerns. SIGNIFICANCE STATEMENT: This work demonstrates that, among off-label potential COVID-19 treatment drugs chloroquine, hydroxychloroquine, azithromycin, and remdesivir, the former two drugs block hERG potassium channels, whereas the latter two drugs do not. All four drugs do not affect KCNQ1 + KCNE1. As hERG and KCNQ1 + KCNE1 are two main K+ channels responsible for ventricular repolarization, and most drugs that induce long QT syndrome (LQTS) do so by impairing hERG channels, these data provide insight into COVID-19 drug-associated LQTS and cardiac safety concerns.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Azitromicina/farmacología , Tratamiento Farmacológico de COVID-19 , Cloroquina/farmacología , Canal de Potasio ERG1/antagonistas & inhibidores , Hidroxicloroquina/farmacología , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/farmacología , Alanina/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Azitromicina/uso terapéutico , COVID-19/metabolismo , Cloroquina/uso terapéutico , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/metabolismo , Células HEK293 , Humanos , Hidroxicloroquina/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico
16.
Int J Mol Sci ; 22(3)2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-33513859

RESUMEN

Substance use disorders (SUDs) are ubiquitous throughout the world. However, much remains to be done to develop pharmacotherapies that are very efficacious because the focus has been mostly on using dopaminergic agents or opioid agonists. Herein we discuss the potential of using potassium channel activators in SUD treatment because evidence has accumulated to support a role of these channels in the effects of rewarding drugs. Potassium channels regulate neuronal action potential via effects on threshold, burst firing, and firing frequency. They are located in brain regions identified as important for the behavioral responses to rewarding drugs. In addition, their expression profiles are influenced by administration of rewarding substances. Genetic studies have also implicated variants in genes that encode potassium channels. Importantly, administration of potassium agonists have been shown to reduce alcohol intake and to augment the behavioral effects of opioid drugs. Potassium channel expression is also increased in animals with reduced intake of methamphetamine. Together, these results support the idea of further investing in studies that focus on elucidating the role of potassium channels as targets for therapeutic interventions against SUDs.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio/genética , Trastornos Relacionados con Sustancias/genética , Potenciales de Acción/genética , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Humanos , Metanfetamina , Canales de Potasio/metabolismo , Trastornos Relacionados con Sustancias/patología
17.
J Neurosci ; 41(2): 376-389, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33219005

RESUMEN

Dysfunction of neuronal circuits is an important determinant of neurodegenerative diseases. Synaptic dysfunction, death, and intrinsic activity of neurons are thought to contribute to the demise of normal behavior in the disease state. However, the interplay between these major pathogenic events during disease progression is poorly understood. Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by a deficiency in the ubiquitously expressed protein SMN and is characterized by motor neuron death, skeletal muscle atrophy, as well as dysfunction and loss of both central and peripheral excitatory synapses. These disease hallmarks result in an overall reduction of neuronal activity in the spinal sensory-motor circuit. Here, we show that increasing neuronal activity by chronic treatment with the FDA-approved potassium channel blocker 4-aminopyridine (4-AP) improves motor behavior in both sexes of a severe mouse model of SMA. 4-AP restores neurotransmission and number of proprioceptive synapses and neuromuscular junctions (NMJs), while having no effects on motor neuron death. In addition, 4-AP treatment with pharmacological inhibition of p53-dependent motor neuron death results in additive effects, leading to full correction of sensory-motor circuit pathology and enhanced phenotypic benefit in SMA mice. Our in vivo study reveals that 4-AP-induced increase of neuronal activity restores synaptic connectivity and function in the sensory-motor circuit to improve the SMA motor phenotype.SIGNIFICANCE STATEMENT Spinal muscular atrophy (SMA) is a neurodegenerative disease, characterized by synaptic loss, motor neuron death, and reduced neuronal activity in spinal sensory-motor circuits. However, whether these are parallel or dependent events is unclear. We show here that long-term increase of neuronal activity by the FDA-approved drug 4-aminopyridine (4-AP) rescues the number and function of central and peripheral synapses in a SMA mouse model, resulting in an improvement of the sensory-motor circuit and motor behavior. Combinatorial treatment of pharmacological inhibition of p53, which is responsible for motor neuron death and 4-AP, results in additive beneficial effects on the sensory-motor circuit in SMA. Thus, neuronal activity restores synaptic connections and improves significantly the severe SMA phenotype.


Asunto(s)
Trastornos del Movimiento/tratamiento farmacológico , Atrofia Muscular Espinal/tratamiento farmacológico , Desempeño Psicomotor/efectos de los fármacos , Trastornos de la Sensación/tratamiento farmacológico , 4-Aminopiridina/uso terapéutico , Animales , Muerte Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Neuronas Motoras/efectos de los fármacos , Trastornos del Movimiento/etiología , Trastornos del Movimiento/psicología , Atrofia Muscular Espinal/complicaciones , Atrofia Muscular Espinal/psicología , Unión Neuromuscular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/uso terapéutico , Propiocepción/efectos de los fármacos , Trastornos de la Sensación/etiología , Trastornos de la Sensación/psicología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
18.
Artículo en Inglés | MEDLINE | ID: mdl-33178487

RESUMEN

The diagnosis of a paroxysmal dyskinesia is difficult and status dystonicus is a rare life threatening movement disorder characterised by severe, frequent or continuous episodes of dystonic spasms. A 25 year old woman with chronic ataxia and paroxysmal dyskinesia presented with facial twitching, writhing of arms, oculogyric crisis and visual and auditory hallucinations. She developed respiratory failure and was ventilated. No cause was found so whole exome sequencing was performed and this revealed a novel, non-synonymous heterozygous variant in exon 11 of the KCNMA1 gene, K457E (c 1369A>G) in the patient but not her parents. This variant has not been previously reported in gnomAD or ClinVar. The finding of a de novo variant in a potassium channel gene guided a trial of the potassium channel antagonist 3,4 diaminopyridine resulting in significant improvement, discharge from the intensive care unit and ultimately home.


Asunto(s)
Ataxia/genética , Corea/genética , Distonía/genética , Alucinaciones/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Trastornos de la Motilidad Ocular/genética , Adulto , Amifampridina/uso terapéutico , Ataxia/tratamiento farmacológico , Ataxia/fisiopatología , Corea/tratamiento farmacológico , Corea/fisiopatología , Distonía/tratamiento farmacológico , Distonía/fisiopatología , Electroencefalografía , Femenino , Alucinaciones/tratamiento farmacológico , Alucinaciones/fisiopatología , Humanos , Trastornos de la Motilidad Ocular/tratamiento farmacológico , Trastornos de la Motilidad Ocular/fisiopatología , Bloqueadores de los Canales de Potasio/uso terapéutico
19.
Biochem Biophys Res Commun ; 533(4): 1255-1261, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33066958

RESUMEN

Oxidative stress is one of the most important risk factors for cataractogenesis. Previous studies have indicated that BDS-II, a Kv3 channel blocker, plays pivotal roles in oxidative stress-related diseases. This study demonstrates that BDS-II exerts a protective effect on cataractogenesis. Specifically, BDS-II was observed to inhibit lens opacity induced by H2O2. BDS-II was also determined to inhibit cataract progression in a sodium selenite-induced in vivo cataract model by inhibiting reduction of the total GSH. In addition, BDS-II was demonstrated to protect human lens epithelial cells against H2O2-induced cell death. Our results suggest that BDS-II is a potential pharmacological candidate in cataract therapy.


Asunto(s)
Catarata/prevención & control , Estrés Oxidativo/efectos de los fármacos , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio Shaw/antagonistas & inhibidores , Animales , Muerte Celular , Línea Celular , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Femenino , Humanos , Cristalino/citología , Masculino , Bloqueadores de los Canales de Potasio/farmacología , Ratas Sprague-Dawley , Canales de Potasio Shaw/metabolismo
20.
Neurodegener Dis Manag ; 10(6): 409-423, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33054615

RESUMEN

Symptomatic treatment has a great relevance for the management of patients with neurologic diseases, since it reduces disease burden and improves quality of life. Aminopyridines (APs) are a group of potassium (K+) channel blocking agents that exert their activity both at central nervous system level and on neuromuscular junction. This review describes the use of APs for the symptomatic treatment of neurological conditions. We will describe trials leading to the approval of the extended-release 4-aminopyridine for MS and evidence in support of the use in other neurological diseases.


Asunto(s)
Aminopiridinas/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/uso terapéutico , 4-Aminopiridina/uso terapéutico , Humanos , Síndrome Miasténico de Lambert-Eaton/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Calidad de Vida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...