Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Ethnopharmacol ; 326: 117996, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38431110

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Schisandra chinensis, the dried and ripe fruit of the magnolia family plant Schisandra chinensis (Turcz.) Baill, was commonly used in traditional analgesic prescription. Studies have shown that the extract of Schisandra chinensis (SC) displayed analgesic activity. However, the analgesic active component and the exact mechanisms have yet to be revealed. AIM OF THE STUDY: The present study was to investigate the anti-nociceptive constituent of Schisandra chinensis, assess its analgesic effect, and explore the potential molecular mechanisms. MATERIALS AND METHODS: The effects of a series of well-recognized compounds from SC on glycine receptors were investigated. The analgesic effect of the identified compound was evaluated in three pain models. Mechanistic studies were performed using patch clamp technique on various targets expressed in recombinant cells. These targets included glycine receptors, Nav1.7 sodium channels, Cav2.2 calcium channels et al. Meanwhile, primary cultured spinal dorsal horn (SDH) neurons and dorsal root ganglion (DRG) neurons were also utilized. RESULTS: Schisandrin B (SchB) was a positive allosteric modulator of glycine receptors in spinal dorsal horn neurons. The EC50 of SchB on glycine receptors in spinal dorsal horn neurons was 2.94 ± 0.28 µM. In three pain models, the analgesic effect of SchB was comparable to that of indomethacin at the same dose. Besides, SchB rescued PGE2-induced suppression of α3 GlyR activity and alleviated persistent pain. Notably, SchB could also potently decrease the frequency of action potentials and inhibit sodium and calcium channels in DRG neurons. Consistent with the data from DRG neurons, SchB was also found to significantly block Nav1.7 sodium channels and Cav2.2 channels in recombinant cells. CONCLUSION: Our results demonstrated that, Schisandrin B, the primary lignan component of Schisandra chinensis, may exert its analgesic effect by acting on multiple ion channels, including glycine receptors, Nav1.7 channels, and Cav2.2 channels.


Asunto(s)
Lignanos , Compuestos Policíclicos , Schisandra , Receptores de Glicina , Lignanos/farmacología , Dolor , Canales de Calcio Tipo N , Analgésicos/farmacología , Analgésicos/uso terapéutico , Canales de Sodio , Ciclooctanos
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.
Epilepsy Behav ; 111: 107251, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32593873

RESUMEN

OBJECTIVE: Ziconotide (ω-conotoxin MVIIA peptide) is a novel analgesic agent acting on voltage-gated calcium channels and is administered intrathecally for neuropathic pain. While antiepileptic activities of other types of calcium channel blockers (T- or L-type) are well established, there is no information regarding the effect of ziconotide as an N-type calcium channel antagonist in pentylenetetrazol-induced seizures or its anxiolytic and sedative activities. The present study is the first to report on these effects. METHODS: To evaluate the anticonvulsant activity of ziconotide in the pentylenetetrazol (60 mg/kg) seizure model, ziconotide was administered intracerebroventricular (i.c.v.) as a single dose (1 µg/rat) or repeatedly (chronic administration: 0.1, 0.3, or 1 µg/rat once a day for seven days). The anxiolytic and sedative actions of ziconotide were evaluated with the elevated plus maze, light/dark (LD) box, and pentobarbital-induced sleep tests. Immediately after behavioral testing, the amygdala was completely removed bilaterally to determine corticosterone levels by immunoassay. RESULTS: In all dosing regimens, ziconotide significantly decreased the seizure frequency and also delayed the latency period compared with control. Chronic administration affected the percentage of mortality protection, while a single dose of ziconotide did not. In behavioral tests, ziconotide significantly increased both the number of entries and the percentage of time spent in the open arms of the elevated plus maze. Furthermore, ziconotide significantly increased the latency period and the number of entries into the light compartment during the LD box examination. Chronic administration of ziconotide significantly reduced the latency to sleep and increased sleeping time, whereas these parameters were not affected by a single dose. Additionally, amygdala corticosterone levels were significantly decreased in rats treated with ziconotide compared with control. CONCLUSION: Ziconotide displays beneficial neurobehavioral effects in a model of epilepsy with anxiety as its comorbid event. It seems that at least one of the mechanisms involved in these effects is associated with a decrease in brain corticosterone levels. The main advantage of ziconotide over benzodiazepines (routine anxiolytic and sedative drugs) is that it does not cause tolerance, dependency, and addiction. Therefore, more than ever, it is necessary to improve the convenience of drug delivery protocols and attenuate the adverse effects associated with ziconotide-based therapies.


Asunto(s)
Ansiolíticos/administración & dosificación , Anticonvulsivantes/administración & dosificación , Bloqueadores de los Canales de Calcio/administración & dosificación , Hipnóticos y Sedantes/administración & dosificación , Convulsiones/tratamiento farmacológico , omega-Conotoxinas/administración & dosificación , Animales , Canales de Calcio Tipo N/fisiología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Masculino , Pentilenotetrazol/toxicidad , Proyectos Piloto , Ratas , Ratas Wistar , Convulsiones/inducido químicamente , Convulsiones/fisiopatología
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.
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
7.
Eur J Neurosci ; 51(7): 1583-1604, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31603587

RESUMEN

Voltage-gated Ca2+ channels (VGCCs) are considered to play a key role in auditory perception and information processing within the murine inner ear and brainstem. In the past, Cav 1.3 L-type VGCCs gathered most attention as their ablation causes congenital deafness. However, isolated patch-clamp investigation and localization studies repetitively suggested that Cav 2.3 R-type VGCCs are also expressed in the cochlea and further components of the ascending auditory tract, pointing to a potential functional role of Cav 2.3 in hearing physiology. Thus, we performed auditory profiling of Cav 2.3+/+ controls, heterozygous Cav 2.3+/- mice and Cav 2.3 null mutants (Cav 2.3-/- ) using brainstem-evoked response audiometry. Interestingly, click-evoked auditory brainstem responses (ABRs) revealed increased hearing thresholds in Cav 2.3+/- mice from both genders, whereas no alterations were observed in Cav 2.3-/- mice. Similar observations were made for tone burst-related ABRs in both genders. However, Cav 2.3 ablation seemed to prevent mutant mice from total hearing loss particularly in the higher frequency range (36-42 kHz). Amplitude growth function analysis revealed, i.a., significant reduction in ABR wave WI and WIII amplitude in mutant animals. In addition, alterations in WI -WIV interwave interval were observed in female Cav 2.3+/- mice whereas absolute latencies remained unchanged. In summary, our results demonstrate that Cav 2.3 VGCCs are mandatory for physiological auditory information processing in the ascending auditory tract.


Asunto(s)
Audiometría de Respuesta Evocada , Umbral Auditivo , Canales de Calcio Tipo N , Potenciales Evocados Auditivos del Tronco Encefálico , Estimulación Acústica , Animales , Tronco Encefálico , Canales de Calcio , Femenino , Masculino , Ratones
8.
Brain ; 143(1): 161-174, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31800012

RESUMEN

Inborn errors of CACNA1A-encoded P/Q-type calcium channels impair synaptic transmission, producing early and lifelong neurological deficits, including childhood absence epilepsy, ataxia and dystonia. Whether these impairments owe their pathologies to defective channel function during the critical period for thalamic network stabilization in immature brain remains unclear. Here we show that mice with tamoxifen-induced adult-onset ablation of P/Q channel alpha subunit (iKOp/q) display identical patterns of dysfunction, replicating the inborn loss-of-function phenotypes and, therefore demonstrate that these neurological defects do not rely upon developmental abnormality. Unexpectedly, unlike the inborn model, the adult-onset pattern of excitability changes believed to be pathogenic within the thalamic network is non-canonical. Specifically, adult ablation of P/Q channels does not promote Cacna1g-mediated burst firing or T-type calcium current (IT) in the thalamocortical relay neurons; however, burst firing in thalamocortical relay neurons remains essential as iKOp/q mice generated on a Cacna1g deleted background show substantially diminished seizure generation. Moreover, in thalamic reticular nucleus neurons, burst firing is impaired accompanied by attenuated IT. Interestingly, inborn deletion of thalamic reticular nucleus-enriched, human childhood absence epilepsy-linked gene Cacna1h in iKOp/q mice reduces thalamic reticular nucleus burst firing and promotes rather than reduces seizure, indicating an epileptogenic role for loss-of-function Cacna1h gene variants reported in human childhood absence epilepsy cases. Together, our results demonstrate that P/Q channels remain critical for maintaining normal thalamocortical oscillations and motor control in the adult brain, and suggest that the developmental plasticity of membrane currents regulating pathological rhythmicity is both degenerate and age-dependent.


Asunto(s)
Ataxia/genética , Canales de Calcio Tipo N/genética , Corteza Cerebral/metabolismo , Epilepsia Tipo Ausencia/genética , Neuronas/metabolismo , Tálamo/metabolismo , Potenciales de Acción , Factores de Edad , Animales , Ataxia/metabolismo , Ataxia/fisiopatología , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Epilepsia Tipo Ausencia/metabolismo , Epilepsia Tipo Ausencia/fisiopatología , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Inhibidores/genética , Potenciales de la Membrana/genética , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Núcleos Talámicos/citología , Tálamo/fisiopatología
9.
J Neurosci ; 39(49): 9841-9851, 2019 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31645463

RESUMEN

Migraine is a complex brain disorder, characterized by attacks of unilateral headache and global dysfunction in multisensory information processing, whose underlying cellular and circuit mechanisms remain unknown. The finding of enhanced excitatory, but unaltered inhibitory, neurotransmission at intracortical synapses in mouse models of familial hemiplegic migraine (FHM) suggested the hypothesis that dysregulation of the excitatory-inhibitory balance in specific circuits is a key pathogenic mechanism. Here, we investigated the thalamocortical (TC) feedforward inhibitory microcircuit in FHM1 mice of both sexes carrying a gain-of-function mutation in CaV2.1. We show that TC synaptic transmission in somatosensory cortex is enhanced in FHM1 mice. Due to similar gain of function of TC excitation of layer 4 excitatory and fast-spiking inhibitory neurons elicited by single thalamic stimulations, neither the excitatory-inhibitory balance nor the integration time window set by the TC feedforward inhibitory microcircuit was altered in FHM1 mice. However, during repetitive thalamic stimulation, the typical shift of the excitatory-inhibitory balance toward excitation and the widening of the integration time window were both smaller in FHM1 compared with WT mice, revealing a dysregulation of the excitatory-inhibitory balance, whereby the balance is relatively skewed toward inhibition. This is due to an unexpected differential effect of the FHM1 mutation on short-term synaptic plasticity at TC synapses on cortical excitatory and fast-spiking inhibitory neurons. Our findings point to enhanced transmission of sensory, including trigeminovascular nociceptive, signals from thalamic nuclei to cortex and TC excitatory-inhibitory imbalance as mechanisms that may contribute to headache, increased sensory gain, and sensory processing dysfunctions in migraine.SIGNIFICANCE STATEMENT Migraine is a complex brain disorder, characterized by attacks of unilateral headache and by global dysfunction in multisensory information processing, whose underlying cellular and circuit mechanisms remain unknown. Here we provide insights into these mechanisms by investigating thalamocortical (TC) synaptic transmission and the function of the TC feedforward inhibitory microcircuit in a mouse model of a rare monogenic migraine. This microcircuit is critical for gating information flow to cortex and for sensory processing. We reveal increased TC transmission and dysregulation of the cortical excitatory-inhibitory balance set by the TC feedforward inhibitory microcircuit, whereby the balance is relatively skewed toward inhibition during repetitive thalamic activity. These alterations may contribute to headache, increased sensory gain, and sensory processing dysfunctions in migraine.


Asunto(s)
Corteza Cerebral/fisiopatología , Retroalimentación Fisiológica , Migraña con Aura/fisiopatología , Vías Nerviosas/fisiopatología , Transmisión Sináptica , Tálamo/fisiopatología , Animales , Canales de Calcio Tipo N/genética , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Migraña con Aura/genética , Mutación , Inhibición Neural , Nocicepción , Técnicas de Placa-Clamp , Transducción de Señal , Nervio Trigémino/fisiopatología
10.
Sci Rep ; 8(1): 11328, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30054511

RESUMEN

Sophorae Flavescentis Radix (SFR) is a medicinal herb with many functions that are involved in anti-inflammation, antinociception, and anticancer. SFR is also used to treat a variety of itching diseases. Matrine (MT) is one of the main constituents in SFR and also has the effect of relieving itching, but the antipruritic mechanism is still unclear. Here, we investigated the effect of MT on anti-pruritus. In acute and chronic itch models, MT significantly inhibited the scratching behavior not only in acute itching induced by histamine (His), chloroquine (CQ) and compound 48/80 with a dose-depended manner, but also in the chronic pruritus models of atopic dermatitis (AD) and acetone-ether-water (AEW) in mice. Furthermore, MT could be detected in the blood after intraperitoneal injection (i.p.) and subcutaneous injection (s.c.). Finally, electrophysiological and calcium imaging results showed that MT inhibited the excitatory synaptic transmission from dorsal root ganglion (DRG) to the dorsal horn of the spinal cord by suppressing the presynaptic N-type calcium channel. Taken together, we believe that MT is a novel drug candidate in treating pruritus diseases, especially for histamine-independent and chronic pruritus, which might be attributed to inhibition of the presynaptic N-type calcium channel.


Asunto(s)
Alcaloides/administración & dosificación , Antipruriginosos/administración & dosificación , Bloqueadores de los Canales de Calcio/administración & dosificación , Prurito/tratamiento farmacológico , Quinolizinas/administración & dosificación , Alcaloides/química , Animales , Antipruriginosos/química , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo N/efectos de los fármacos , Canales de Calcio Tipo N/genética , Modelos Animales de Enfermedad , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Masculino , Ratones , Prurito/genética , Prurito/patología , Quinolizinas/química , Sophora/química , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Matrinas
11.
Nano Lett ; 17(2): 886-892, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28094958

RESUMEN

Techniques offering remote control of neural activity with high spatiotemporal resolution and specificity are invaluable for deciphering the physiological roles of different classes of neurons in brain development and disease. Here, we first confirm that microfabricated substrates with enhanced magnetic field gradients allow for wireless stimulation of neural circuits dosed with magnetic nanoparticles using calcium indicator dyes. We also investigate the mechanism of mechano-transduction in this system and identify that N-type mechano-sensitive calcium ion channels play a key role in signal generation in response to magnetic force. We next applied this method for chronic stimulation of a fragile X syndrome (FXS) neural network model and found that magnetic force-based stimulation modulated the expression of mechano-sensitive ion channels which are out of equilibrium in a number of neurological diseases including FXS. This technique can serve as a tool for acute and chronic modulation of endogenous ion channel expression in neural circuits in a spatially localized manner to investigate a number of disease processes in the future.


Asunto(s)
Canales de Calcio Tipo N/fisiología , Nanopartículas de Magnetita/química , Red Nerviosa/fisiopatología , Animales , Fenómenos Biomecánicos , Encéfalo/patología , Encéfalo/fisiopatología , Calcio/metabolismo , Señalización del Calcio , Campos Electromagnéticos , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Síndrome del Cromosoma X Frágil/fisiopatología , Síndrome del Cromosoma X Frágil/terapia , Humanos , Magnetoterapia , Mecanotransducción Celular , Red Nerviosa/patología , Neuronas/fisiología
12.
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
13.
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
14.
J Neurosci ; 36(2): 405-18, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26758833

RESUMEN

Generalized spike-wave seizures involving abnormal synchronization of cortical and underlying thalamic circuitry represent a major category of childhood epilepsy. Inborn errors of Cacna1a, the P/Q-type voltage-gated calcium channel α subunit gene, expressed throughout the brain destabilize corticothalamic rhythmicity and produce this phenotype. To determine the minimal cellular lesion required for this network disturbance, we used neurotensin receptor 1 (Ntsr1) cre-driver mice to ablate floxed Cacna1a in layer VI pyramidal neurons, which supply the sole descending cortical synaptic input to thalamocortical relay cells and reticular interneurons and activate intrathalamic circuits. Targeted Cacna1a ablation in layer VI cells resulted in mice that display a robust spontaneous spike-wave absence seizure phenotype accompanied by behavioral arrest and inhibited by ethosuximide. To verify the selectivity of the molecular lesion, we determined that P/Q subunit proteins were reduced in corticothalamic relay neuron terminal zones, and confirmed that P/Q-mediated glutamate release was reduced at these synapses. Spike-triggered exocytosis was preserved by N-type calcium channel rescue, demonstrating that evoked release at layer VI terminals relies on both P/Q and N-type channels. Whereas intrinsic excitability of the P/Q channel depleted layer VI neurons was unaltered, T-type calcium currents in the postsynaptic thalamic relay and reticular cells were dramatically elevated, favoring rebound bursting and seizure generation. We find that an early P/Q-type release defect, limited to synapses of a single cell-type within the thalamocortical circuit, is sufficient to remodel synchronized firing behavior and produce a stable generalized epilepsy phenotype. SIGNIFICANCE STATEMENT: This study dissects a critical component of the corticothalamic circuit in spike-wave epilepsy and identifies the developmental importance of P/Q-type calcium channel-mediated presynaptic glutamate release at layer VI pyramidal neuron terminals. Genetic ablation of Cacna1a in layer VI neurons produced synchronous spike-wave discharges in the cortex and thalamus that were inhibited by ethosuximide. These mice also displayed N-type calcium channel compensation at descending thalamic synapses, and consistent with other spike-wave models increased low-threshold T-type calcium currents within postsynaptic thalamic relay and reticular neurons. These results demonstrate, for the first time, that preventing the developmental homeostatic switch from loose to tightly coupled synaptic release at a single class of deep layer cortical excitatory output neurons results in generalized spike-wave epilepsy.


Asunto(s)
Canales de Calcio Tipo N/deficiencia , Epilepsia Tipo Ausencia/patología , Neuronas/patología , Tálamo/patología , Corteza Visual/patología , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Canales de Calcio Tipo N/genética , Modelos Animales de Enfermedad , Epilepsia Tipo Ausencia/tratamiento farmacológico , Epilepsia Tipo Ausencia/genética , Etosuximida/uso terapéutico , Potenciales Postsinápticos Excitadores/genética , Femenino , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Motores/etiología , Trastornos Motores/genética , Mutación/genética , Tiempo de Reacción/genética , Receptores de Neurotensina/metabolismo
15.
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
16.
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
18.
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
19.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 35(4): 461-5, 2015 Apr.
Artículo en Chino | MEDLINE | ID: mdl-26043571

RESUMEN

OBJECTIVE: To study whether the analgesis of oxymatrine (OMT) affects N-type voltage-gated calcium channels (VGCCs). METHODS: Totally 45 mice were randomly divided into the sham-operation group, the model group [established by partial sciatic nerve ligation (PSNL)] , and the OMT treatment group according to random digit table, 15 in each group. The dorsal root ganglions (DRG) were separated in PSNL pain model mice. Intracellular calcium concentration ([Ca2+]i) was determined with Fluo-3 AM immunofluorescent probe in cultured DRG neurons. Different protein expression levels of N-type (Cav2. 2) and L-type ( Cav1. 3) among VGCCs from brain and DRG tissues were detected with Western blot. RESULTS: Compared with the sham-operation group, [Ca2+]i, increased in cultured DRG neurons (P <0. 05) , protein expression levels of Cav2. 2 in the brain tissue increased (P <0. 05), protein expression levels of Cav2. 2 in DRG tissues decreased in the model group (P <0. 01). Compared with the model group, [Ca2+]i, decreased in cultured DRG neurons (P < 0. 05), protein expression levels of Cav2. 2 in the brain tissue decreased (P <0. 01), protein expression levels of Cav2. 2 in DRG tissues increased in the OMT treatment group (P <0. 01). There was no statistical difference in Cav1. 3 expressions in cultured DRG neurons and the brain (P >0. 05). CONCLUSION: Analgesic effect of OMT might be related to Cav2. 2 channel mediated calcium ion flux.


Asunto(s)
Alcaloides/farmacología , Analgésicos/farmacología , Canales de Calcio Tipo N/fisiología , Quinolizinas/farmacología , Analgesia/métodos , Compuestos de Anilina , Animales , Calcio , Ganglios Espinales , Ratones , Neuronas , Dolor , Xantenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA