Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pediatr Neurol ; 157: 5-13, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38833907

RESUMEN

BACKGROUND: Congenital myasthenic syndromes (CMS) are a group of inherited neuromuscular junction (NMJ) disorders arising from gene variants encoding diverse NMJ proteins. Recently, the VAMP1 gene, responsible for encoding the vesicle-associated membrane protein 1 (VAMP1), has been associated with CMS. METHODS: This study presents a characterization of five new individuals with VAMP1-related CMS, providing insights into the phenotype. RESULTS: The individuals with VAMP1-related CMS exhibited early disease onset, presenting symptoms prenatally or during the neonatal period, alongside severe respiratory involvement and feeding difficulties. Generalized weakness at birth was a common feature, and none of the individuals achieved independent walking ability. Notably, all cases exhibited scoliosis. The clinical course remained stable, without typical exacerbations seen in other CMS types. The response to anticholinesterase inhibitors and salbutamol was only partial, but the addition of 3,4-diaminopyridine (3,4-DAP) led to significant and substantial improvements, suggesting therapeutic benefits of 3,4-DAP for managing VAMP1-related CMS symptoms. Noteworthy is the identification of the VAMP1 (NM_014231.5): c.340delA; p.Ile114SerfsTer72 as a founder variant in the Iberian Peninsula and Latin America. CONCLUSIONS: This study contributes valuable insights into VAMP1-related CMS, emphasizing their early onset, arthrogryposis, facial and generalized weakness, respiratory involvement, and feeding difficulties. Furthermore, the potential efficacy of 3,4-DAP as a useful therapeutic option warrants further exploration. The findings have implications for clinical management and genetic counseling in affected individuals. Additional research is necessary to elucidate the long-term outcomes of VAMP1-related CMS.


Asunto(s)
Amifampridina , Síndromes Miasténicos Congénitos , Fenotipo , Proteína 1 de Membrana Asociada a Vesículas , Humanos , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/fisiopatología , Femenino , Masculino , Amifampridina/farmacología , Proteína 1 de Membrana Asociada a Vesículas/genética , Niño , Adolescente , 4-Aminopiridina/análogos & derivados , 4-Aminopiridina/farmacología , 4-Aminopiridina/uso terapéutico , Preescolar , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Lactante
2.
J Biol Chem ; 296: 100302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33465376

RESUMEN

3,4-Diaminopyridine (3,4-DAP) increases transmitter release from neuromuscular junctions (NMJs), and low doses of 3,4-DAP (estimated to reach ∼1 µM in serum) are the Food and Drug Administration (FDA)-approved treatment for neuromuscular weakness caused by Lambert-Eaton myasthenic syndrome. Canonically, 3,4-DAP is thought to block voltage-gated potassium (Kv) channels, resulting in prolongation of the presynaptic action potential (AP). However, recent reports have shown that low millimolar concentrations of 3,4-DAP have an off-target agonist effect on the Cav1 subtype ("L-type") of voltage-gated calcium (Cav) channels and have speculated that this agonist effect might contribute to 3,4-DAP effects on transmitter release at the NMJ. To address 3,4-DAP's mechanism(s) of action, we first used the patch-clamp electrophysiology to characterize the concentration-dependent block of 3,4-DAP on the predominant presynaptic Kv channel subtypes found at the mammalian NMJ (Kv3.3 and Kv3.4). We identified a previously unreported high-affinity (1-10 µM) partial antagonist effect of 3,4-DAP in addition to the well-known low-affinity (0.1-1 mM) antagonist activity. We also showed that 1.5-µM DAP had no effects on Cav1.2 or Cav2.1 current. Next, we used voltage imaging to show that 1.5- or 100-µM 3,4-DAP broadened the AP waveform in a dose-dependent manner, independent of Cav1 calcium channels. Finally, we demonstrated that 1.5- or 100-µM 3,4-DAP augmented transmitter release in a dose-dependent manner and this effect was also independent of Cav1 channels. From these results, we conclude that low micromolar concentrations of 3,4-DAP act solely on Kv channels to mediate AP broadening and enhance transmitter release at the NMJ.


Asunto(s)
Amifampridina/farmacología , Fármacos Neuromusculares/farmacología , Unión Neuromuscular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Terminales Presinápticos/efectos de los fármacos , Canales de Potasio Shaw/metabolismo , Acetilcolina/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica , Masculino , Ratones , Microelectrodos , Unión Neuromuscular/metabolismo , Terminales Presinápticos/metabolismo , Rana pipiens , Canales de Potasio Shaw/antagonistas & inhibidores , Canales de Potasio Shaw/genética , Técnicas de Cultivo de Tejidos
3.
Drugs Today (Barc) ; 56(10): 623-641, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33185628

RESUMEN

Lambert-Eaton myasthenic syndrome (LEMS) is a presynaptic autoimmune disabling neuromuscular disease caused by antibodies against presynaptic voltage-gated calcium channels. It reduces the quantal release of acetylcholine (Ach), causing muscle weakness, reduced or absent reflex and dysautonomia. About half of LEMS patients have associated small cell lung cancer. For symptomatic treatment, amifampridine (3,4-diaminopyridine [3,4-DAP]) is ideal because it increases the release of Ach at the presynaptic membrane. Since the first use of 3,4-DAP in LEMS patients in the 1980s, 136 LEMS patients were treated with amifampridines in the open-label studies and 208 patients in the eight randomized studies. These studies showed that amifampridine is the most effective drug for symptomatic treatment in LEMS. Now, 3,4-DAPP (3,4-DAP phosphate) is approved for adult LEMS patients and 3,4-DAP for pediatric patients. The recommended dose is 80 mg a day, divided 3 or 4 times a day. Side effects are usually mild, and the most frequently reported are paresthesia.


Asunto(s)
Amifampridina/farmacología , Síndrome Miasténico de Lambert-Eaton , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Adulto , Niño , Humanos , Síndrome Miasténico de Lambert-Eaton/diagnóstico , Síndrome Miasténico de Lambert-Eaton/tratamiento farmacológico
4.
JCI Insight ; 5(2)2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-31996484

RESUMEN

Botulinum neurotoxins (BoNTs) are potent neuroparalytic toxins that cause mortality through respiratory paralysis. The approved medical countermeasure for BoNT poisoning is infusion of antitoxin immunoglobulins. However, antitoxins have poor therapeutic efficacy in symptomatic patients; thus, there is an urgent need for treatments that reduce the need for artificial ventilation. We report that the US Food and Drug Administration-approved potassium channel blocker 3,4-diaminopyridine (3,4-DAP) reverses respiratory depression and neuromuscular weakness in murine models of acute and chronic botulism. In ex vivo studies, 3,4-DAP restored end-plate potentials and twitch contractions of diaphragms isolated from mice at terminal stages of BoNT serotype A (BoNT/A) botulism. In vivo, human-equivalent doses of 3,4-DAP reversed signs of severe respiratory depression and restored mobility in BoNT/A-intoxicated mice at terminal stages of respiratory collapse. Multiple-dosing administration of 3,4-DAP improved respiration and extended survival at up to 5 LD50 BoNT/A. Finally, 3,4-DAP reduced gastrocnemius muscle paralysis and reversed respiratory depression in sublethal models of serotype A-, B-, and E-induced botulism. These findings make a compelling argument for repurposing 3,4-DAP to symptomatically treat symptoms of muscle paralysis caused by botulism, independent of serotype. Furthermore, they suggest that 3,4-DAP is effective for a range of botulism symptoms at clinically relevant time points.


Asunto(s)
Amifampridina/farmacología , Amifampridina/uso terapéutico , Antitoxinas/farmacología , Antitoxinas/uso terapéutico , Botulismo/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Amifampridina/química , Animales , Antitoxinas/química , Toxinas Botulínicas , Toxinas Botulínicas Tipo A/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Dosificación Letal Mediana , Ratones , Músculo Esquelético , Parálisis/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/química , Serogrupo , Estados Unidos , United States Food and Drug Administration
5.
Expert Rev Clin Pharmacol ; 12(11): 1013-1018, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31639317

RESUMEN

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.


Asunto(s)
Amifampridina/administración & dosificación , Síndrome Miasténico de Lambert-Eaton/tratamiento farmacológico , Fármacos Neuromusculares/administración & dosificación , Amifampridina/efectos adversos , Amifampridina/farmacología , Humanos , Síndrome Miasténico de Lambert-Eaton/fisiopatología , Fármacos Neuromusculares/efectos adversos , Fármacos Neuromusculares/farmacología , Bloqueadores de los Canales de Potasio/administración & dosificación , Bloqueadores de los Canales de Potasio/efectos adversos , Bloqueadores de los Canales de Potasio/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Comprimidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...