Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Pharmacol ; 181(12): 1734-1756, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38157867

RESUMEN

BACKGROUND AND PURPOSE: Neuropathic pain affects up to 10% of the global population and is caused by an injury or a disease affecting the somatosensory, peripheral, or central nervous system. NP is characterized by chronic, severe and opioid-resistant properties. Therefore, its clinical management remains very challenging. The N-type voltage-gated calcium channel, Cav2.2, is a validated target for therapeutic intervention in chronic and neuropathic pain. The conotoxin ziconotide (Prialt®) is an FDA-approved drug that blocks Cav2.2 channel but needs to be administered intrathecally. Thus, although being principally efficient, the required application route is very much in disfavour. EXPERIMENTAL APPROACH AND KEY RESULTS: Here, we describe an orally available drug candidate, RD2, which competes with ziconotide binding to Cav2.2 at nanomolar concentrations and inhibits Cav2.2 almost completely reversible. Other voltage-gated calcium channel subtypes, like Cav1.2 and Cav3.2, were affected by RD2 only at concentrations higher than 10 µM. Data from sciatic inflammatory neuritis rat model demonstrated the in vivo proof of concept, as low-dose RD2 (5 mg·kg-1) administered orally alleviated neuropathic pain compared with vehicle controls. High-dose RD2 (50 mg·kg-1) was necessary to reduce pain sensation in acute thermal response assessed by the tail flick test. CONCLUSIONS AND IMPLICATIONS: Taken together, these results demonstrate that RD2 has antiallodynic properties. RD2 is orally available, which is the most convenient application form for patients and caregivers. The surprising and novel result from standard receptor screens opens the room for further optimization into new promising drug candidates, which address an unmet medical need.


Asunto(s)
Bloqueadores de los Canales de Calcio , Canales de Calcio Tipo N , Neuralgia , Animales , Humanos , Masculino , Ratones , Ratas , Administración Oral , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo N/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ratones Endogámicos C57BL , Neuralgia/tratamiento farmacológico , omega-Conotoxinas/administración & dosificación , omega-Conotoxinas/farmacología , omega-Conotoxinas/uso terapéutico , Ratas Endogámicas Lew
2.
J Pharm Pharmacol ; 73(7): 874-880, 2021 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-33822115

RESUMEN

OBJECTIVES: Neuropathic pain (NP) is a chronic inflammation of the sciatic nerve, associated with complex pathophysiological events like neuronal ectopic discharge with changes in neurotransmitters, growth factors, receptors/ion channels including N-methyl-d-aspartate receptors, Transient receptor cation channels, Voltage-gated calcium channels. All these events eventually lead to inflammation and apoptosis of the sciatic nerve in NP. Icariin (ICA), a natural flavonoid is well known for its anti-inflammatory potential. Hence, the present study is designed to evaluate its anti-inflammatory potential against neuropathic pain using in silico and in vivo studies. METHODS: In silico studies were conducted using targets of N-methyl-D-aspartate receptor subtype-2B (NR2B), The capsaicin receptor transient receptor cation channel subfamily-V member-1 (TRPV1), N-type voltage-gated calcium (CaV2.2) channels. In in vivo studies, after partial sciatic nerve ligation surgery to animals, received their respective treatment for 21 days, further TNF-α, IL-6, Bax (proapoptotic) and Bcl-2 (antiapoptotic) expressions were estimated. KEY FINDINGS: ICA decreased the expressions of TNF-α, IL-6, Bax and increased expression of Bcl-2. In silico studies revealed a good energy binding score towards NR2B, TRPV1 receptors and CaV2.2 ion Channel. CONCLUSIONS: ICA could be a promising agent in alleviating neuropathic pain by inhibiting NR2B, TRPV1 receptors and Cav2.2 channels, which induces anti-apoptotic potential and inhibits inflammation.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Flavonoides/farmacología , Neuralgia , Receptores de N-Metil-D-Aspartato/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Antiinflamatorios/farmacología , Proteínas Reguladoras de la Apoptosis/análisis , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Ratas , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología
3.
Toxicon ; 188: 80-88, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33038354

RESUMEN

Opioids are the "gold standard" treatment for postoperative pain, but these drugs also have limiting adverse effects. Thus, adjuvant drugs might be useful in opioid therapy for postoperative pain. The aim of the present study was to evaluate the effect of Phα1ß, a dual blocker of Cav2 and TRPA1 channels, on antinociceptive and adverse actions of morphine in a model of postoperative pain. Phα1ß (100-300 pmol/site) or morphine (3-10 mg/kg), alone, largely reduced postoperative nociception. However, Phα1ß (100 pmol/site) or morphine (10 mg/kg) also produced motor impairment. Lower doses of Phα1ß (30 pmol/site) or morphine (1 mg/kg), that did not have an effect alone, showed antinociceptive effect when concomitantly administrated. Moreover, co-administration of Phα1ß (30 pmol/site) with morphine (1 or 10 mg/kg) was unable to cause motor impairment. Preoperative repeated treatment with morphine increased the expression of Cav2 and TRPA1 channels in spinal cord, and caused tolerance and withdrawal syndrome, which were reversed with a single injection of Phα1ß (30 pmol/site). When injected postoperatively, escalating doses of morphine worsened postoperative hyperalgesia, induced tolerance, and withdrawal syndrome. Similarly, Phα1ß (30 pmol/site) reversed these adverse effects. Single or repeated morphine caused constipation, which was not altered by Phα1ß. Thus, a low dose of Phα1ß potentiated the analgesia, and reversed some adverse effects of morphine on operated mice, indicating the potential use of this agent as an adjuvant drug in opioid therapy for postoperative pain.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Quimioterapia Adyuvante/métodos , Dolor Postoperatorio/tratamiento farmacológico , Venenos de Araña/uso terapéutico , Analgésicos , Animales , Canales de Calcio Tipo N/metabolismo , Hiperalgesia/inducido químicamente , Ratones , Morfina , Venenos de Araña/farmacología , Canal Catiónico TRPA1/metabolismo
4.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1715-1728, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32388600

RESUMEN

The present research work was designed to examine the neuroprotective effect of ethanolic extract of Solanum virginianum Linn. (SV) in chronic construction injury (CCI) of sciatic nerve-induced neuropathic pain in rats. The extract was initially standardized by high-performance thin-layer chromatography using solasodine as a biomarker and was then subjected to assess the degree of mechanical allodynia, thermal allodynia, mechanical hyperalgesia, thermal hyperalgesia and biochemical evaluations. Administration of SV (100 and 200 mg/kg; p.o.) and pregabalin (10 mg/kg; p.o.) as a reference standard significantly debilitated hyperalgesia and allodynia and notably restored the altered antioxidant level and pro-inflammatory cytokine (IL-1ß and TNF-α) expression in a dose-dependent manner. Further, to appraise the mechanistic approach of solasodine, docking simulation studies were done on the 3D structure of the voltage-gated N-type calcium channel (Cav 2.2), R-type calcium channel (Cav 2.3) and sodium channel (Nav 1.7), and the results revealed that solasodine properly positioned into Phe 19, Leu 32, Met 51 and Met 71 (FLMM pocket) of Cav 2.2 and Cav 2.3 and being a competitor of Ca2+/N-lobe it may inactivate these calcium channels but did not bind into the desired binding pocket of Nav 1.7. Thus, the study confirmed the role of solasodine as a major biomarker for the observed neuroprotective nature of Solanum virginianum.


Asunto(s)
Analgésicos/farmacología , Hiperalgesia/prevención & control , Simulación del Acoplamiento Molecular , Neuralgia/prevención & control , Umbral del Dolor/efectos de los fármacos , Extractos Vegetales/farmacología , Neuropatía Ciática/tratamiento farmacológico , Alcaloides Solanáceos/farmacología , Solanum , Analgésicos/aislamiento & purificación , Analgésicos/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Sitios de Unión , Unión Competitiva , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo N/metabolismo , Modelos Animales de Enfermedad , Etanol/química , Femenino , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Neuralgia/metabolismo , Neuralgia/fisiopatología , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/metabolismo , Unión Proteica , Ratas Wistar , Neuropatía Ciática/metabolismo , Neuropatía Ciática/fisiopatología , Alcaloides Solanáceos/aislamiento & purificación , Alcaloides Solanáceos/metabolismo , Solanum/química , Solventes/química
5.
Pharmacol Rep ; 72(3): 600-611, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32399819

RESUMEN

BACKGROUND: Orofacial pain is clinically challenging, having therapeutic failures and side effects. This study evaluated the antinociceptive activities of the CTK 01512-2 toxin, the TRPA1 channel antagonist, and the selective inhibitor of the N-type voltage-gated calcium channels (N-type VGCC), in different pain models. MATERIALS AND METHODS: The trigeminal ganglia were stimulated in vitro with capsaicin. The in vivo models received subcutaneous (sc) injections of formalin into the upper lip of the rats, Freund's Complete Adjuvant (FCA) into the temporomandibular joint (TMJ), and infraorbital nerve constrictions (IONC). CTK 01512-2 at concentrations of 30, 100, and 300 pmol/site, intrathecally (ith), and MVIIA at 10, 30, and 100 pmol/site in the formalin test, guided the doses for the models. The glutamate levels in the CSF of the rats that were submitted to IONC were analyzed. RESULTS: CTK 01512-2 decreased the nociceptive behavior in the inflammatory phase of the formalin test (65.94 ± 7.35%) and MVIIA in the neurogenic phase (81.23 ± 3.36%). CTK 01512-2 reduced facial grooming with FCA in the TMJ (96.7 ± 1.6%), and in the IONC neuropathy model, it decreased heat hyperalgesia (100%) and cold hyperalgesia (81.61 ± 9.02%). The levels of glutamate in the trigeminal ganglia in vitro (81.40 ± 8.59%) and in the CSF in vivo (70.0 ± 9.2%) were reduced. CONCLUSIONS: The roles of TRPA1 in pain transduction and the performance of CTK 01512-2 in the inhibition of the N-type VGCCs were reinforced. This dual activity may represent an advantage in clinical treatments.


Asunto(s)
Analgésicos/farmacología , Dolor Facial/tratamiento farmacológico , Canal Catiónico TRPA1/antagonistas & inhibidores , omega-Conotoxinas/farmacología , Animales , Canales de Calcio Tipo N/metabolismo , Capsaicina/farmacología , Modelos Animales de Enfermedad , Adyuvante de Freund , Ácido Glutámico/metabolismo , Hiperalgesia/tratamiento farmacológico , Masculino , Neuralgia/tratamiento farmacológico , Dimensión del Dolor , Ratas , Ratas Wistar
6.
Pain ; 160(1): 117-135, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30169422

RESUMEN

The Federal Pain Research Strategy recommended development of nonopioid analgesics as a top priority in its strategic plan to address the significant public health crisis and individual burden of chronic pain faced by >100 million Americans. Motivated by this challenge, a natural product extracts library was screened and identified a plant extract that targets activity of voltage-gated calcium channels. This profile is of interest as a potential treatment for neuropathic pain. The active extract derived from the desert lavender plant native to southwestern United States, when subjected to bioassay-guided fractionation, afforded 3 compounds identified as pentacyclic triterpenoids, betulinic acid (BA), oleanolic acid, and ursolic acid. Betulinic acid inhibited depolarization-evoked calcium influx in dorsal root ganglion (DRG) neurons predominantly through targeting low-voltage-gated (Cav3 or T-type) and CaV2.2 (N-type) calcium channels. Voltage-clamp electrophysiology experiments revealed a reduction of Ca, but not Na, currents in sensory neurons after BA exposure. Betulinic acid inhibited spontaneous excitatory postsynaptic currents and depolarization-evoked release of calcitonin gene-related peptide from lumbar spinal cord slices. Notably, BA did not engage human mu, delta, or kappa opioid receptors. Intrathecal administration of BA reversed mechanical allodynia in rat models of chemotherapy-induced peripheral neuropathy and HIV-associated peripheral sensory neuropathy as well as a mouse model of partial sciatic nerve ligation without effects on locomotion. The broad-spectrum biological and medicinal properties reported, including anti-HIV and anticancer activities of BA and its derivatives, position this plant-derived small molecule natural product as a potential nonopioid therapy for management of chronic pain.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo T/metabolismo , Infecciones por VIH/complicaciones , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Paclitaxel/toxicidad , Triterpenos/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/toxicidad , Células CHO , Cricetulus , Diprenorfina/farmacocinética , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/citología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Triterpenos Pentacíclicos , Traumatismos de los Nervios Periféricos/inducido químicamente , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/etiología , Traumatismos de los Nervios Periféricos/virología , Ratas , Ratas Sprague-Dawley , Tritio/farmacocinética , Ácido Betulínico
7.
Pain ; 158(2): 347-360, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28092651

RESUMEN

Treatments for chronic pain are inadequate, and new options are needed. Nonpharmaceutical approaches are especially attractive with many potential advantages including safety. Light therapy has been suggested to be beneficial in certain medical conditions such as depression, but this approach remains to be explored for modulation of pain. We investigated the effects of light-emitting diodes (LEDs), in the visible spectrum, on acute sensory thresholds in naive rats as well as in experimental neuropathic pain. Rats receiving green LED light (wavelength 525 nm, 8 h/d) showed significantly increased paw withdrawal latency to a noxious thermal stimulus; this antinociceptive effect persisted for 4 days after termination of last exposure without development of tolerance. No apparent side effects were noted and motor performance was not impaired. Despite LED exposure, opaque contact lenses prevented antinociception. Rats fitted with green contact lenses exposed to room light exhibited antinociception arguing for a role of the visual system. Antinociception was not due to stress/anxiety but likely due to increased enkephalins expression in the spinal cord. Naloxone reversed the antinociception, suggesting involvement of central opioid circuits. Rostral ventromedial medulla inactivation prevented expression of light-induced antinociception suggesting engagement of descending inhibition. Green LED exposure also reversed thermal and mechanical hyperalgesia in rats with spinal nerve ligation. Pharmacological and proteomic profiling of dorsal root ganglion neurons from green LED-exposed rats identified changes in calcium channel activity, including a decrease in the N-type (CaV2.2) channel, a primary analgesic target. Thus, green LED therapy may represent a novel, nonpharmacological approach for managing pain.


Asunto(s)
Analgésicos/uso terapéutico , Dolor Crónico/terapia , Fototerapia/métodos , Animales , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Color , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Femenino , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Masculino , Bulbo Raquídeo , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Umbral del Dolor/efectos de la radiación , Estimulación Física/efectos adversos , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/efectos de la radiación , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de la radiación
8.
Am J Chin Med ; 44(5): 943-62, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27430911

RESUMEN

Interest in the health benefits of flavonoids, particularly their effects on neurodegenerative disease, is increasing. This study evaluated the role of baicalein, a flavonoid compound isolated from the traditional Chinese medicine Scutellaria baicalensis, in glutamate release and glutamate neurotoxicity in the rat hippocampus. In the rat hippocampal nerve terminals (synaptosomes), baicalein inhibits depolarization-induced glutamate release, and this phenomenon is prevented by chelating the extracellular Ca[Formula: see text] ions and blocking presynaptic Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel activity. In slice preparations, whole cell patch-clamp experiments revealed that baicalein reduced the frequency of miniature excitatory postsynaptic currents, without affecting their amplitude. In a kainic acid rat model, intraperitoneally administering baicalein to rats before the kainic acid intraperitoneal injection substantially attenuated kainic acid-induced neuronal cell death, c-Fos expression, and the activation of the mammalian target of rapamycin in the hippocampus. This study is the first to demonstrate that the natural compound baicalein inhibits glutamate release from hippocampal nerve terminals, and executes a protective action against kainic acid-induced excitotoxicity in vivo. The findings enhance the understanding of baicalein's action in the brain, and suggest that this natural compound is valuable for treating brain disorders related to glutamate excitotoxicity.


Asunto(s)
Encefalopatías/tratamiento farmacológico , Encefalopatías/fisiopatología , Flavanonas/administración & dosificación , Ácido Glutámico/toxicidad , Neuronas/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Sustancias Protectoras/administración & dosificación , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Encefalopatías/genética , Encefalopatías/metabolismo , Calcio/metabolismo , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Ácido Glutámico/metabolismo , Humanos , Ácido Kaínico/efectos adversos , Masculino , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Scutellaria baicalensis/metabolismo
9.
J Gen Physiol ; 146(3): 205-19, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26283199

RESUMEN

The growth hormone secretagogue receptor type 1a (GHSR1a) has the highest known constitutive activity of any G protein-coupled receptor (GPCR). GHSR1a mediates the action of the hormone ghrelin, and its activation increases transcriptional and electrical activity in hypothalamic neurons. Although GHSR1a is present at GABAergic presynaptic terminals, its effect on neurotransmitter release remains unclear. The activities of the voltage-gated calcium channels, CaV2.1 and CaV2.2, which mediate neurotransmitter release at presynaptic terminals, are modulated by many GPCRs. Here, we show that both constitutive and agonist-dependent GHSR1a activity elicit a strong impairment of CaV2.1 and CaV2.2 currents in rat and mouse hypothalamic neurons and in a heterologous expression system. Constitutive GHSR1a activity reduces CaV2 currents by a Gi/o-dependent mechanism that involves persistent reduction in channel density at the plasma membrane, whereas ghrelin-dependent GHSR1a inhibition is reversible and involves altered CaV2 gating via a Gq-dependent pathway. Thus, GHSR1a differentially inhibits CaV2 channels by Gi/o or Gq protein pathways depending on its mode of activation. Moreover, we present evidence suggesting that GHSR1a-mediated inhibition of CaV2 attenuates GABA release in hypothalamic neurons, a mechanism that could contribute to neuronal activation through the disinhibition of postsynaptic neurons.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Ghrelina/metabolismo , Hipotálamo/fisiología , Neuronas/fisiología , Receptores de Ghrelina/metabolismo , Animales , Secuencia de Bases , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cultivadas , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Ratones , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/genética
11.
Biochem Biophys Res Commun ; 465(2): 188-93, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26239659

RESUMEN

Growing evidence suggests that mammalian peripheral somatosensory neurons express functional receptors for gamma-aminobutyric acid, GABAA and GABAB. Moreover, local release of GABA by pain-sensing (nociceptive) nerve fibres has also been suggested. Yet, the functional significance of GABA receptor triggering in nociceptive neurons is not fully understood. Here we used patch-clamp recordings from small-diameter cultured DRG neurons to investigate effects of GABAB receptor agonist baclofen on voltage-gated Ca(2+) currents. We found that baclofen inhibited both low-voltage activated (LVA, T-type) and high-voltage activated (HVA) Ca(2+) currents in a proportion of DRG neurons by 22% and 32% respectively; both effects were sensitive to Gi/o inhibitor pertussis toxin. Inhibitory effect of baclofen on both current types was about twice less efficacious as compared to that of the µ-opioid receptor agonist DAMGO. Surprisingly, only HVA but not LVA current modulation by baclofen was partially prevented by G protein inhibitor GDP-ß-S. In contrast, only LVA but not HVA current modulation was reversed by the application of a reducing agent dithiothreitol (DTT). Inhibition of T-type Ca(2+) current by baclofen and the recovery of such inhibition by DTT were successfully reconstituted in the expression system. Our data suggest that inhibition of LVA current in DRG neurons by baclofen is partially mediated by an unconventional signaling pathway that involves a redox mechanism. These findings reinforce the idea of targeting peripheral GABA receptors for pain relief.


Asunto(s)
Baclofeno/farmacología , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo T/metabolismo , Agonistas de Receptores GABA-B/farmacología , Receptores de GABA-B/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Ditiotreitol/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Ganglios Espinales , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Células HEK293 , Humanos , Nocicepción/fisiología , Dolor/metabolismo , Dolor/fisiopatología , Técnicas de Placa-Clamp , Toxina del Pertussis/farmacología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Transducción de Señal , Tionucleótidos/farmacología , Ácido gamma-Aminobutírico/metabolismo
12.
Neuroscience ; 304: 198-208, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26208839

RESUMEN

Ataxia is the predominant clinical manifestation of cerebellar dysfunction. Mutations in the human CACNA1A gene, encoding the pore-forming α1 subunit of CaV2.1 (P/Q-type) calcium channels, underlie several neurological disorders, including Episodic Ataxia type 2 and Familial Hemiplegic Migraine type 1 (FHM1). Several mouse mutants exist that harbor mutations in the orthologous Cacna1a gene. The spontaneous Cacna1a mutants Rolling Nagoya (tg(rol)), Tottering (tg) and Leaner (tg(ln)) mice exhibit behavioral motor phenotypes, including ataxia. Transgenic knock-in (KI) mouse strains with the human FHM1 R192Q and S218L missense mutations have been generated. R192Q KI mice are non-ataxic, whereas S218L KI mice display a complex behavioral phenotype that includes cerebellar ataxia. Given the dependence of γ-aminobutyric acid type A (GABAA) receptor subunit functioning on localized calcium currents, and the functional link between GABAergic inhibition and ataxia, we hypothesized that cerebellar GABAA receptor expression is differentially affected in Cacna1a mutants and contributes to the ataxic phenotype. Herein we quantified functional GABAA receptors and pharmacologically dissociated cerebellar GABAA receptors in several Cacna1a mutants. We did not identify differences in the expression of GABAA receptor subunits or in the number of functional GABAA receptors in the non-ataxic R192Q KI strain. In contrast, tg(rol) mice had a ∼15% decrease in the number of functional GABAA receptors, whereas S218L KI mice showed a ∼29% increase. Our data suggest that differential changes in cerebellar GABAA receptor expression profile may contribute to the neurological phenotype of cerebellar ataxia and that targeting GABAA receptors might represent a feasible complementary strategy to treat cerebellar ataxia.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Cerebelo/metabolismo , Cerebelo/patología , Neuronas/metabolismo , Neuronas/patología , Animales , Ataxia/metabolismo , Ataxia/patología , Canales de Calcio Tipo N/genética , Técnicas de Sustitución del Gen , Humanos , Ratones Transgénicos , Mutación , Fenotipo , Receptores de GABA-A/metabolismo
13.
Arthritis Rheumatol ; 67(6): 1657-67, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25733371

RESUMEN

OBJECTIVE: A hallmark of rheumatoid arthritis (RA) is the chronic pain that accompanies inflammation and joint deformation. Patients with RA rate pain relief as the highest priority; however, few studies have addressed the efficacy and safety of therapies directed specifically toward pain pathways. The ω-conotoxin MVIIA (ziconotide) is used in humans to alleviate persistent pain syndromes, because it specifically blocks the voltage-gated calcium 2.2 (CaV 2.2) channel, which mediates the release of neurotransmitters and proinflammatory mediators from peripheral nociceptor nerve terminals. The aims of this study were to investigate whether blockade of CaV 2.2 can suppress arthritis pain, and to examine the progression of induced arthritis during persistent CaV 2.2 blockade. METHODS: Transgenic mice expressing a membrane-tethered form of MVIIA under the control of a nociceptor-specific gene (MVIIA-transgenic mice) were used in the experiments. The mice were subjected to unilateral induction of joint inflammation using a combination of antigen and collagen. RESULTS: CaV 2.2 blockade mediated by tethered MVIIA effectively suppressed arthritis-induced pain; however, in contrast to their wild-type littermates, which ultimately regained use of their injured joint as inflammation subsided, MVIIA-transgenic mice showed continued inflammation, with up-regulation of the osteoclast activator RANKL and concomitant joint and bone destruction. CONCLUSION: Taken together, our results indicate that alleviation of peripheral pain by blockade of CaV 2.2- mediated calcium influx and signaling in nociceptor sensory neurons impairs recovery from induced arthritis and point to the potentially devastating effects of using CaV 2.2 channel blockers as analgesics during inflammation.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Canales de Calcio Tipo N/metabolismo , Dolor Nociceptivo/metabolismo , Nociceptores/metabolismo , Ligando RANK/metabolismo , Rodilla de Cuadrúpedos/metabolismo , omega-Conotoxinas/genética , Animales , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo N/inmunología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Dolor Nociceptivo/tratamiento farmacológico , Dolor Nociceptivo/inmunología , Rodilla de Cuadrúpedos/patología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , omega-Conotoxinas/uso terapéutico
15.
J Med Food ; 18(5): 516-23, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25340625

RESUMEN

The excessive release of glutamate is a critical element in the neuropathology of acute and chronic brain disorders. The purpose of the present study was to investigate the effect and possible mechanism of myricetin, a naturally occurring flavonoid with a neuroprotective profile, on endogenous glutamate release in the nerve terminals (synaptosomes) of the rat cerebral cortex. The release of glutamate was evoked by the K(+) channel blocker 4-aminopyridine (4-AP) and measured by one-line enzyme-coupled fluorometric assay. We also used a membrane potential-sensitive dye to assay the synaptosomal plasma membrane potential, and a Ca(2+) indicator Fura-2 to monitor cytosolic Ca(2+) concentrations ([Ca(2+)]C). Results show that myricetin inhibited 4-AP-evoked glutamate release, and this effect was prevented by chelating extracellular Ca(2+) ions and the vesicular transporter inhibitor bafilomycin A1. However, the glutamate transporter inhibitor dl-threo-beta-benzyl-oxyaspartate had no effect on myricetin action. Myricetin did not alter the synaptosomal membrane potential, but decreased 4-AP-induced increases in the cytosolic free Ca(2+) concentration. Furthermore, the myricetin effect on 4-AP-evoked glutamate release was prevented by blocking the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channels, but not by blocking intracellular Ca(2+) release. These results suggest that myricetin inhibits glutamate release from cerebrocortical synaptosomes by attenuating voltage-dependent Ca(2+) entry. This implies that the inhibition of glutamate release is an important pharmacological activity of myricetin that may play a critical role in the apparent clinical efficacy of this compound.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Flavonoides/farmacología , Ácido Glutámico/metabolismo , Terminaciones Nerviosas/efectos de los fármacos , Animales , Calcio/metabolismo , Canales de Calcio Tipo N/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Masculino , Terminaciones Nerviosas/metabolismo , Ratas , Ratas Sprague-Dawley , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
16.
Bioorg Med Chem ; 22(24): 6899-907, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25456079

RESUMEN

N-type calcium channels represent a promising target for the treatment of neuropathic pain. The selective N-type calcium channel blocker ziconotide ameliorates severe chronic pain but has a narrow therapeutic window and requires intrathecal administration. We identified tetrahydroisoquinoline derivative 1a as a novel potent N-type calcium channel blocker. However, this compound also exhibited potent inhibitory activity against hERG channels. Structural optimizations led to identification of (1S)-(1-cyclohexyl-3,4-dihydroisoquinolin-2(1H)-yl)-2-{[(1-hydroxycyclohexyl)methyl]amino}ethanone ((S)-1h), which exhibited high selectivity for hERG channels while retaining potency for N-type calcium channel inhibition. (S)-1h went on to demonstrate in vivo efficacy as an orally available N-type calcium channel blocker in a rat spinal nerve ligation model of neuropathic pain.


Asunto(s)
Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo N/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Tetrahidroisoquinolinas/química , Animales , Bloqueadores de los Canales de Calcio/metabolismo , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo N/química , Línea Celular Tumoral , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Evaluación Preclínica de Medicamentos , Canales de Potasio Éter-A-Go-Go/química , Humanos , Masculino , Neuralgia/tratamiento farmacológico , Unión Proteica , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Tetrahidroisoquinolinas/metabolismo , Tetrahidroisoquinolinas/uso terapéutico
17.
Biochemistry ; 53(38): 6052-62, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25188201

RESUMEN

In neurons, entry of extracellular calcium (Ca(2+)) into synaptic terminals through Cav2.1 (P/Q-type) Ca(2+) channels is the driving force for exocytosis of neurotransmitter-containing synaptic vesicles. This class of Ca(2+) channel is, therefore, pivotal during normal neurotransmission in higher organisms. In response to channel opening and Ca(2+) influx, specific Ca(2+)-binding proteins associate with cytoplasmic regulatory domains of the P/Q channel to modulate subsequent channel opening. Channel modulation in this way influences synaptic plasticity with consequences for higher-level processes such as learning and memory acquisition. The ubiquitous Ca(2+)-sensing protein calmodulin (CaM) regulates the activity of all types of mammalian voltage-gated Ca(2+) channels, including the P/Q class, by direct binding to specific regulatory motifs. More recently, experimental evidence has highlighted a role for additional Ca(2+)-binding proteins, particularly of the CaBP and NCS families in the regulation of P/Q channels. NCS-1 is a protein found from yeast to humans and that regulates a diverse number of cellular functions. Physiological and genetic evidence indicates that NCS-1 regulates P/Q channel activity, including calcium-dependent facilitation, although a direct physical association between the proteins has yet to be demonstrated. In this study, we aimed to determine if there is a direct interaction between NCS-1 and the C-terminal cytoplasmic tail of the Cav2.1 α-subunit. Using distinct but complementary approaches, including in vitro binding of bacterially expressed recombinant proteins, fluorescence spectrophotometry, isothermal titration calorimetry, nuclear magnetic resonance, and expression of fluorescently tagged proteins in mammalian cells, we show direct binding and demonstrate that CaM can compete for it. We speculate about how NCS-1/Cav2.1 association might add to the complexity of calcium channel regulation mediated by other known calcium-sensing proteins and how this might help to fine-tune neurotransmission in the mammalian central nervous system.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Proteínas Sensoras del Calcio Neuronal/metabolismo , Neuropéptidos/metabolismo , Calcio/metabolismo , Canales de Calcio Tipo N/química , Clonación Molecular , Humanos , Proteínas Sensoras del Calcio Neuronal/química , Neuropéptidos/química , Unión Proteica
18.
Mol Pain ; 9: 51, 2013 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-24139484

RESUMEN

BACKGROUND: Antagonists of N-type voltage-gated calcium channels (VGCC), Ca(v)2.2, can manage severe chronic pain with intrathecal use and may be effective systemically. A series of novel ω-conotoxins that selectively inhibit N-type VGCCs was isolated from Conus catus. In the present study, the potency and reversibility of ω-conotoxins CVID, CVIE and CVIF to inhibit N-type calcium currents were investigated in mouse isolated dorsal root ganglion (DRG) neurons. The systemic potency of each ω-conotoxin to reverse signs of mouse chronic inflammatory pain was also compared. RESULTS: In DRG neurons, the rank order of potency to inhibit N-type calcium currents was CVIE > CVIF > CVID. After subcutaneous administration, CVID and CVIE, but not CVIF, partially reversed impaired weight bearing in mice injected with Freund's complete adjuvant (CFA) three days prior to testing. No side-effects associated with systemic administration of ω-conotoxins were observed. CONCLUSIONS: The present study indicates a potential for CVID and CVIE to be developed as systemically active analgesics with no accompanying neurological side-effects.


Asunto(s)
Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo N/metabolismo , Dolor/tratamiento farmacológico , omega-Conotoxinas/uso terapéutico , Analgésicos/administración & dosificación , Analgésicos/uso terapéutico , Animales , Bloqueadores de los Canales de Calcio/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/metabolismo , Técnicas de Placa-Clamp , omega-Conotoxinas/administración & dosificación
19.
Bioorg Med Chem Lett ; 22(11): 3639-42, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22560585

RESUMEN

A structure-activity relationship study of 6-unsubstituted-1,4-dihydropyridine and 2,6-unsubstituted-1,4-dihydropyridine derivatives was conducted in an attempt to discover N-type calcium channel blockers that were highly selective over L-type calcium channel blockers. Among the tested compounds, (+)-4-(3,5-dichloro-4-methoxy-phenyl)-1,4-dihydro-pyridine-3,5-dicarboxylic acid 3-cinnamyl ester was found to be an effective and selective N-type calcium channel blocker with oral analgesic potential.


Asunto(s)
Analgésicos/química , Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo N/química , Ácidos Carboxílicos/química , Dihidropiridinas/química , Administración Oral , Analgésicos/síntesis química , Analgésicos/farmacología , Animales , Bloqueadores de los Canales de Calcio/síntesis química , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/metabolismo , Ácidos Carboxílicos/síntesis química , Ácidos Carboxílicos/farmacología , Evaluación Preclínica de Medicamentos , Formaldehído/toxicidad , Dimensión del Dolor/efectos de los fármacos , Ratas , Relación Estructura-Actividad
20.
J Neurophysiol ; 108(1): 148-59, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22490555

RESUMEN

The molecular and physiological basis of the touch-unresponsive zebrafish mutant fakir has remained elusive. Here we report that the fakir phenotype is caused by a missense mutation in the gene encoding voltage-gated calcium channel 2.1b (CACNA1Ab). Injection of RNA encoding wild-type CaV2.1 restores touch responsiveness in fakir mutants, whereas knockdown of CACNA1Ab via morpholino oligonucleotides recapitulates the fakir mutant phenotype. Fakir mutants display normal current-evoked synaptic communication at the neuromuscular junction but have attenuated touch-evoked activation of motor neurons. NMDA-evoked fictive swimming is not affected by the loss of CaV2.1b, suggesting that this channel is not required for motor pattern generation. These results, coupled with the expression of CACNA1Ab by sensory neurons, suggest that CaV2.1b channel activity is necessary for touch-evoked activation of the locomotor network in zebrafish.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Activación del Canal Iónico/genética , Tacto/genética , Acetilcolina/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Vías Aferentes/fisiología , Animales , Animales Modificados Genéticamente , Bungarotoxinas/metabolismo , Canales de Calcio Tipo N/genética , Curare/farmacología , Relación Dosis-Respuesta a Droga , Embrión no Mamífero , Reacción de Fuga/efectos de los fármacos , Reacción de Fuga/fisiología , Potenciales Evocados/genética , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Leucina/genética , Locomoción/efectos de los fármacos , Locomoción/genética , Modelos Moleculares , Morfolinas/farmacología , Actividad Motora/genética , Neuronas Motoras/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Mutagénesis Sitio-Dirigida/métodos , Mutación/genética , Mutación Missense/genética , Red Nerviosa/fisiología , Antagonistas Nicotínicos/farmacología , Médula Espinal/citología , Médula Espinal/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Tacto/fisiología , Valina/genética , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA