Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Zhen Ci Yan Jiu ; 48(4): 372-7, 2023 Apr 25.
Artículo en Chino | MEDLINE | ID: mdl-37186202

RESUMEN

OBJECTIVE: To observe the effects of electroacupuncture (EA) combined with acellular nerve allograft (ANA) on the morphological structure of spinal ganglion cells and the protein expressions of nerve growth factor (NGF) and phosphorylated protein kinase B (p-Akt) in rats with sciatic nerve injury (SNI), so as to explore the protective mechanism of EA combined with ANA on spinal ganglia. METHODS: SPF male SD rats were randomly divided into normal, model, single ANA bridging (bridging) and EA + ANA (combination) groups, with 10 rats in each group. The SNI rat model was established by right sciatic nerve transection. Rats in the bridging group were bridged with ANA to the two broken ends of injured sciatic nerves. Rats in the combination group were treated with EA at "Yanglingquan" (GB34) and "Huantiao" (GB30) 2 d after ANA bridging, with dilatational wave, frequency of 1 Hz/20 Hz, intensity of 1 mA, 15 min/d, 7 d as a course of treatment for 4 consecutive courses. Sciatic function index (SFI) was observed by footprint test. Wet weight ratio of tibialis anterior muscle was calculated after weighing. Morphology of rat spinal ganglion cells was observed after Nissl staining. The protein expressions of NGF and p-Akt were detected by immunofluorescence and Western blot. RESULTS: Compared with the normal group, the SFI and wet weight ratio of tibialis anterior muscle were significantly decreased (P<0.05), the number of Nissl bodies in spinal ganglion cells was significantly reduced (P<0.05) with dissolution and incomplete structure, the protein expressions of NGF and p-Akt in ganglion cells were significantly decreased (P<0.05) in the model group. Following the interventions and in comparison with the model group, the SFI and the wet weight ratio of tibialis anterior muscle were significantly increased (P<0.05), the damage of Nissl bodies in ganglion cells was reduced and the number was obviously increased (P<0.05), and the protein expressions of NGF and p-Akt in ganglion cells were significantly increased (P<0.05) in the bridging and combination groups. Compared with the bridging group, the SFI and the wet weight ratio of tibialis anterior muscle were increased (P<0.05), the morphology of Nissl bodies in ganglion cells was more regular and the number was increased (P<0.05), the protein expressions of NGF and p-Akt in spinal ganglion cells were significantly increased (P<0.05) in the combination group. CONCLUSION: EA combined with ANA can improve the SFI and the wet weight ratio of tibialis anterior muscle in SNI rats, improve the morphology and structure of Nissl bodies in spinal ganglion cells, and increase the protein expressions of NGF and p-Akt in spinal ganglion, so as to play a protective role on spinal ganglia.


Asunto(s)
Aloinjertos , Electroacupuntura , Ganglios Espinales , Traumatismos de los Nervios Periféricos , Nervio Ciático , Animales , Masculino , Ratas , Aloinjertos/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Traumatismos de los Nervios Periféricos/terapia , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Nervio Ciático/lesiones
2.
J Ethnopharmacol ; 285: 114896, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896207

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: 'Cold feeling' is a subjective feeling of unusual coldness that aggravates fatigue, stiffness, and other symptoms, thereby reducing quality of life. Tokishakuyakusan (TSS) is a Kampo medicine reported to improve cold feeling and is used to treat symptoms aggravated by cold feeling. However, the mechanism of action of TSS is unclear. Cold feeling may involve reduced blood flow and subsequent inhibition of heat transport. Therefore, elucidating the effects of TSS on blood flow is one of the most important research topics for clarifying the mechanism of action of TSS. AIM OF THE STUDY: We aimed to evaluate the effect of TSS on recovery from lowered body temperature by the immersion of rats in cold water and to clarify the involvement of blood flow in the action of TSS. MATERIALS AND METHODS: After female Wistar rats underwent 9 days of low room temperature stress loading (i.e. room temperature of 18 °C), they were subjected to immersion in cold water (15 °C) for 15 min. Body surface temperature, rectal temperature, and plantar temperature were measured before and after immersion in cold water. Blood flow was measured before and after immersion in cold water without low room temperature stress loading. TSS (0.5 g/kg or 1 g/kg) or the vehicle (i.e. distilled water) was orally administered once daily for 10 days for the measurement of body temperature or once 30 min before immersion in cold water for the measurement of blood flow. In addition, we examined the effect of TSS on calcitonin gene-related peptide (CGRP) release from dorsal root ganglion (DRG) cells, the effect of TSS ingredients on transient receptor potential (TRP) channels, and the effect of TSS ingredients on the membrane potential of vascular smooth muscle cells and evaluated the mechanism of the effects of TSS on blood flow. RESULTS: Body temperature and blood flow decreased after immersion in cold water and then recovered over time. A comparison of body temperature at each timepoint or area under the curve showed that TSS (1 g/kg) accelerated the recovery of body surface temperature, rectal temperature, and blood flow. TSS significantly increased CGRP release from DRG cells, which disappeared after pretreatment with HC-030031 (a transient receptor potential ankyrin 1 [TRPA1] antagonist). The effects of seven TSS ingredients on TRP channels were examined. The agonistic effect on TRPA1 was observed for atractylodin, atractylodin carboxylic acid and levistolide A. Among the TSS ingredients, atractylodin carboxylic acid had significant hyperpolarising effects. CONCLUSIONS: The mechanism by which TSS accelerates the recovery of lowered body temperature in rats after immersion in cold water may involve the acceleration of the recovery of lowered blood flow. Increased CGRP release from DRG cells by TSS, TRPA1 activation by TSS ingredients, and membrane potential changes in vascular smooth muscle cells caused by TSS ingredients are part of the mechanism of action of TSS. These findings may partly contribute to the interpretation of the beneficial effects of TSS on cold feeling.


Asunto(s)
Circulación Sanguínea/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Frío , Medicamentos Herbarios Chinos/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/química , Femenino , Ganglios Espinales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Medicina Kampo , Miocitos del Músculo Liso/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Wistar , Arterias Umbilicales/citología
3.
Int Immunopharmacol ; 101(Pt B): 108364, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34844873

RESUMEN

BACKGROUND: Treatment of chronic inflammatory pain remains a major goal in the clinic. It is thus of prime importance to characterize inherent pathophysiological pathways to design new therapeutic strategies and analgesics for pain management. Paeoniflorin (PF), a monoterpenoid glycoside from Paeonia lactiflora Pallas plants, possesses promising anti-nociceptive property. However, therapeutic effect and underlying mechanism of action of PF on inflammatory pain have not yet been fully elucidated. In this study, we aim to investigate the analgesic effect further and clarify its mechanism of action of PF on complete freund's adjuvant (CFA)-evoked inflammatory pain. METHODS: Twenty-four male mice were divided into 3 groups: sham, CFA, and CFA + PF groups (n = 8/group). Mice were treated with normal saline or PF (30 mg/kg) for 11 days. Footpad swelling (n = 8/group), mechanical (n = 8/group) and thermal hypersensitivity (n = 8/group) were measured to evaluate the analgesic effect of PF on CFA-injected mice. At the end of the animal experiment, blood and L4-L6 dorsal root ganglion neurons were collected to assess the therapeutic effect of PF on CFA-induced inflammatory pain. Next, hematoxylin and eosin, quantitative realtime PCR, ELISA, capsaicin and dimethyl succinate induced pain test (n = 8/group), motor coordination test (n = 8/group), tail flicking test (n = 8/group), pyruvate and succinate dehydrogenase assay (n = 6/group), immunohistochemical staining, were performed to clarify the action mechanism of PF on CFA-evoked inflammatory pain. Besides, the effect of PF on TRPV1 was evaluated by whole-cell patch clamp recording on primary neurons (n = 7). Finally, molecular docking further performed to evaluate the binding ability of PF to TRPV1. RESULTS: PF significantly relieved inflammatory pain (P < 0.001) and paw edema (P < 0.001) on a complete Freund adjuvant (CFA)-induced peripheral inflammatory pain model. Furthermore, PF inhibited neutrophil infiltration (P < 0.01), IL-1ß increase (P < 0.01), and pain-related peptide substance P release (P < 0.001). Intriguingly, CFA-induced succinate aggregation was notably reversed by PF via modulating pyruvate and SDH activity (P < 0.01). In addition, PF dampened the high expression of subsequent succinate receptor SUCNR1 (P < 0.01), HIF-1α (P < 0.05), as well as the activation of NLPR3 inflammasome (P < 0.05) and TRPV1 (P < 0.05). More importantly, both capsaicin and dimethyl succinate supplementation obviously counteracted the pain-relieving effect of PF and TRPV1 (P < 0.01 or P < 0.001). CONCLUSION: Our findings suggest that PF can significantly relieve CFA-induced paw swelling, as well as mechanical and thermal hyperalgesia. PF alleviated inflammatory pain partly through inhibiting the activation of TRPV1 and succinate/SUCNR1-HIF-1α/NLPR3 pathway. Furthermore, we found that PF exerted its analgesic effect without affecting motor coordination and pain-related cold ion-channels. In summary, this study may provide valuable evidence for the potential application of PF as therapeutic strategy for inflammatory pain treatment.


Asunto(s)
Antiinflamatorios no Esteroideos , Glucósidos , Monoterpenos , Neuronas , Receptores Acoplados a Proteínas G , Ácido Succínico , Animales , Masculino , Ratones , Analgésicos , Antiinflamatorios no Esteroideos/farmacología , Capsaicina , Adyuvante de Freund/toxicidad , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Glucósidos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación , Monoterpenos/farmacología , Neuronas/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Ácido Succínico/metabolismo , Canales Catiónicos TRPV
4.
Physiol Rep ; 9(16): e14975, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34405579

RESUMEN

Voltage-gated ion channels play a key role in the action potential (AP) initiation and its propagation in sensory neurons. Modulation of their activity during chronic inflammation creates a persistent pain state. In this study, we sought to determine how peripheral inflammation caused by complete Freund's adjuvant (CFA) alters the fast sodium (INa ), L-type calcium (ICaL ), and potassium (IK ) currents in primary afferent fibers to increase nociception. In our model, intraplantar administration of CFA induced mechanical allodynia and thermal hyperalgesia at day 14 post-injection. Using whole-cell patch-clamp recording in dissociated small (C), medium (Aδ), and large-sized (Aß) rat dorsal root ganglion (DRG) neurons, we found that CFA prolonged the AP duration and increased the amplitude of the tetrodotoxin-resistant (TTX-r) INa in Aß fibers. In addition, CFA accelerated the recovery of INa from inactivation in C and Aδ nociceptive fibers but enhanced the late sodium current (INaL ) only in Aδ and Aß neurons. Inflammation similarly reduced the amplitude of ICaL in each neuronal cell type. Fourteen days after injection, CFA reduced both components of IK (IKdr and IA ) in Aδ fibers. We also found that IA was significantly larger in C and Aδ neurons in normal conditions and during chronic inflammation. Our data, therefore, suggest that targeting the transient potassium current IA represents an efficient way to shift the balance toward antinociception during inflammation, since its activation will selectively decrease the AP duration in nociceptive fibers. Altogether, our data indicate that complex interactions between IK , INa , and ICaL reduce pain threshold by concomitantly enhancing the activity of nociceptive neurons and reducing the inhibitory action of Aß fibers during chronic inflammation.


Asunto(s)
Potenciales de Acción , Neuronas Aferentes/metabolismo , Dolor Nociceptivo/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiología , Masculino , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Nocicepción , Dolor Nociceptivo/fisiopatología , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología
5.
Pharm Res ; 38(7): 1179-1186, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34244893

RESUMEN

PURPOSE: Pharmaceutical buffer systems, especially for injectable biologics such as monoclonal antibodies, are an important component of successful FDA-approved medications. Clinical studies indicate that buffer components may be contributing factors for increased injection site pain. METHODS: To determine the potential nociceptive effects of clinically relevant buffer systems, we developed an in vitro multi-electrode array (MEA) based recording system of rodent dorsal root ganglia (DRG) sensory neuron cell culture. This system monitors sensory neuron activity/firing as a surrogate of nociception when challenged with buffer components used in formulating monoclonal antibodies and other injectable biologics. RESULTS: We show that citrate salt and citrate mannitol buffer systems cause an increase in mean firing rate, burst frequency, and burst duration in DRG sensory neurons, unlike histidine or saline buffer systems at the same pH value. Lowering the concentration of citrate leads to a lower firing intensity of DRG sensory neurons. CONCLUSION: Increased activity/firing of DRG sensory neurons has been suggested as a key feature underlying nociception. Our results support the utility of an in vitro MEA assay with cultured DRG sensory neurons to probe the nociceptive potential of clinically relevant buffer components used in injectable biologics.


Asunto(s)
Productos Biológicos/administración & dosificación , Reacción en el Punto de Inyección/prevención & control , Inyecciones/efectos adversos , Nocicepción/efectos de los fármacos , Dolor/prevención & control , Animales , Productos Biológicos/química , Tampones (Química) , Células Cultivadas , Evaluación Preclínica de Medicamentos/instrumentación , Electrodos , Ganglios Espinales/citología , Dolor/etiología , Cultivo Primario de Células , Ratas , Células Receptoras Sensoriales/efectos de los fármacos
6.
J Mol Neurosci ; 71(6): 1290-1300, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33417168

RESUMEN

To study the effect of photobiomodulation (PBM) on axon regeneration and secretion change of dorsal root ganglion (DRG) under oxidative stress after spinal cord injury (SCI), and further explore the effect of changes in DRG secretion caused by PBM on the polarization of macrophages. The PBM-DRG model was constructed to perform PBM on neurons under oxidative stress simulated in vitro. And the irradiation conditions were as follows: wavelength, 810 nm; power density, 2 mW/cm2; irradiation area, 4.5 cm2; and irradiation time, 440 s. Then resulted in an energy of 4 J (2 mW/cm2 × 4.5 cm2 × 440 s). About 100 µM H202 was added to the culture medium to simulate oxidative stress after SCI. An ROS (reactive oxygen species) assay kit was used to measure ROS contend in the DRG. The survival level of the neurons was measured using the CCK-8 method, and the axon regeneration of neurons was observed by using immunofluorescence. The secretion level of CCL2 from DRG was determined by RT-qPCR and ELISA. Further culturing macrophages of DRG-conditioned medium culture, the expression level of iNOS and Arg-1 in macrophages was assessed using Western blot analysis. The expression level of TNF-α and IL-1ß was determined by ELISA. After adding the neutralizing antibody of CCL2 to the DRG neuron-conditioned medium following PBM irradiation to culture macrophages to observe the effects on macrophage polarization and secretion. PBM could reduce ROS levels in neurons, increase neuronal survival under oxidative stress, and promote neuronal axon regeneration. In addition, PBM could also promote CCL2 secretion by DRG under oxidative stress. By constructing a DRG supernatant-M1 macrophage adoptive culture model, we found that the supernatant of DRG after PBM intervention could reduce the expression level of iNOS and the secretion of TNF-α and IL-1ß in M1 macrophages; at the same time, it could also up-regulate the expression of Arg-1, one of the markers of M2 macrophages. Furthermore, these effects could be prevented by the addition of neutralizing antibodies of CCL2. PBM could promote survival and axonal regeneration of DRG under SCI oxidative stress, increase the secretion level of CCL2 by DRG, and this change can reduce the polarization of macrophages to M1, further indicating that PBM could promote spinal cord injury repair.


Asunto(s)
Axones/metabolismo , Quimiocina CCL2/metabolismo , Macrófagos/citología , Estrés Oxidativo , Fototerapia/métodos , Traumatismos de la Médula Espinal/terapia , Regeneración de la Medula Espinal , Animales , Axones/efectos de la radiación , Diferenciación Celular , Células Cultivadas , Quimiocina CCL2/genética , Femenino , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiología , Interleucina-1beta/metabolismo , Luz , Macrófagos/inmunología , Macrófagos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa/metabolismo
7.
Biochem J ; 478(1): 1-20, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33305808

RESUMEN

Friedreich ataxia (FA) is a neurodegenerative disease caused by the deficiency of frataxin, a mitochondrial protein. In primary cultures of dorsal root ganglia neurons, we showed that frataxin depletion resulted in decreased levels of the mitochondrial calcium exchanger NCLX, neurite degeneration and apoptotic cell death. Here, we describe that frataxin-deficient dorsal root ganglia neurons display low levels of ferredoxin 1 (FDX1), a mitochondrial Fe/S cluster-containing protein that interacts with frataxin and, interestingly, is essential for the synthesis of calcitriol, the active form of vitamin D. We provide data that calcitriol supplementation, used at nanomolar concentrations, is able to reverse the molecular and cellular markers altered in DRG neurons. Calcitriol is able to recover both FDX1 and NCLX levels and restores mitochondrial membrane potential indicating an overall mitochondrial function improvement. Accordingly, reduction in apoptotic markers and neurite degeneration was observed and, as a result, cell survival was also recovered. All these beneficial effects would be explained by the finding that calcitriol is able to increase the mature frataxin levels in both, frataxin-deficient DRG neurons and cardiomyocytes; remarkably, this increase also occurs in lymphoblastoid cell lines derived from FA patients. In conclusion, these results provide molecular bases to consider calcitriol for an easy and affordable therapeutic approach for FA patients.


Asunto(s)
Calcitriol/farmacología , Ferredoxinas/metabolismo , Ataxia de Friedreich/metabolismo , Proteínas de Unión a Hierro/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Neuronas/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Apoptosis/efectos de los fármacos , Calcitriol/biosíntesis , Calcitriol/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Vitamina D/metabolismo , Frataxina
8.
Mol Pain ; 16: 1744806920970368, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33307981

RESUMEN

The embryonic rat dorsal root ganglion (DRG) neuron-derived 50B11 cell line is a promising sensory neuron model expressing markers characteristic of NGF and GDNF-dependent C-fibre nociceptors. Whether these cells have the capacity to develop into distinct nociceptive subtypes based on NGF- or GDNF-dependence has not been investigated. Here we show that by augmenting forskolin (FSK) and growth factor supplementation with NGF or GDNF, 50B11 cultures can be driven to acquire differential functional responses to common nociceptive agonists capsaicin and ATP respectively. In addition, to previous studies, we also demonstrate that a differentiated neuronal phenotype can be maintained for up to 7 days. Western blot analysis of nociceptive marker proteins further demonstrates that the 50B11 cells partially recapitulate the functional phenotypes of classical NGF-dependent (peptidergic) and GDNF-dependent (non-peptidergic) neuronal subtypes described in DRGs. Further, 50B11 cells differentiated with NGF/FSK, but not GDNF/FSK, show sensitization to acute prostaglandin E2 treatment. Finally, RNA-Seq analysis confirms that differentiation with NGF/FSK or GDNF/FSK produces two 50B11 cell subtypes with distinct transcriptome expression profiles. Gene ontology comparison of the two subtypes of differentiated 50B11 cells to rodent DRG neurons studies shows significant overlap in matching or partially matching categories. This transcriptomic analysis will aid future suitability assessment of the 50B11 cells as a high-throughput nociceptor model for a broad range of experimental applications. In conclusion, this study shows that the 50B11 cell line is capable of partially recapitulating features of two distinct types of embryonic NGF and GDNF-dependent nociceptor-like cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ganglios Espinales/citología , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Factor de Crecimiento Nervioso/farmacología , Nociceptores/citología , Potenciales de Acción/efectos de los fármacos , Adenosina Trifosfato/farmacología , Animales , Biomarcadores/metabolismo , Capsaicina/farmacología , Diferenciación Celular/genética , Línea Celular , Forma de la Célula/efectos de los fármacos , Colforsina/farmacología , Dinoprostona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Variación Genética , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nociceptores/efectos de los fármacos , Fenotipo , Análisis de Componente Principal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Canales de Sodio/metabolismo
9.
Theranostics ; 10(26): 12111-12126, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33204332

RESUMEN

Rationale: Psoriasis is a chronic inflammatory disease caused by a complex interplay between the immune and nervous systems with recurrent scaly skin plaques, thickened stratum corneum, infiltration and activation of inflammatory cells, and itch. Despite an increasing availability of immune therapies, they often have adverse effects, high costs, and dissociated effects on inflammation and itch. Activation of sensory neurons innervating the skin and TRPV1 (transient receptor potential vanilloid 1) are emerging as critical components in the pathogenesis of psoriasis, but little is known about their endogenous inhibitors. Recent studies have demonstrated that resolvins, endogenous lipid mediators derived from omega-3 fatty acids, are potent inhibitors of TRP channels and may offer new therapies for psoriasis without known adverse effects. Methods: We used behavioral, electrophysiological and biochemical approaches to investigate the therapeutic effects of resolvin D3 (RvD3), a novel family member of resolvins, in a preclinical model of psoriasis consisting of repeated topical applications of imiquimod (IMQ) to murine skin, which provokes inflammatory lesions that resemble human psoriasis. Results: We report that RvD3 specifically reduced TRPV1-dependent acute pain and itch in mice. Mechanistically, RvD3 inhibited capsaicin-induced TRPV1 currents in dissociated dorsal root ganglion (DRG) neurons via the N-formyl peptide receptor 2 (i.e. ALX/FPR2), a G-protein coupled receptor. Single systemic administration of RvD3 (2.8 mg/kg) reversed itch after IMQ, and repeated administration largely prevented the development of both psoriasiform itch and skin inflammation with concomitant decreased in calcitonin gene-related peptide (CGRP) expression in DRG neurons. Accordingly, specific knockdown of CGRP in DRG was sufficient to prevent both psoriasiform itch and skin inflammation similar to the effects following RvD3 administration. Finally, we elevated the translational potential of this study by showing that RvD3 significantly inhibited capsaicin-induced TRPV1 activity and CGRP release in human DRG neurons. Conclusions: Our findings demonstrate a novel role for RvD3 in regulating TRPV1/CGRP in mouse and human DRG neurons and identify RvD3 and its neuronal pathways as novel therapeutic targets to treat psoriasis.


Asunto(s)
Ácidos Grasos Insaturados/farmacología , Dolor/tratamiento farmacológico , Prurito/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Canales Catiónicos TRPV/antagonistas & inhibidores , Animales , Biopsia , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Capsaicina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Ácidos Grasos Insaturados/uso terapéutico , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/inmunología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/inmunología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/inducido químicamente , Dolor/inmunología , Dolor/patología , Técnicas de Placa-Clamp , Cultivo Primario de Células , Prurito/inducido químicamente , Prurito/inmunología , Prurito/patología , Psoriasis/complicaciones , Psoriasis/inmunología , Psoriasis/patología , Piel/efectos de los fármacos , Piel/inmunología , Piel/inervación , Canales Catiónicos TRPV/metabolismo
10.
Mol Brain ; 13(1): 73, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393368

RESUMEN

Chronic pain can be the result of an underlying disease or condition, medical treatment, inflammation, or injury. The number of persons experiencing this type of pain is substantial, affecting upwards of 50 million adults in the United States. Pharmacotherapy of most of the severe chronic pain patients includes drugs such as gabapentinoids, re-uptake blockers and opioids. Unfortunately, gabapentinoids are not effective in up to two-thirds of this population and although opioids can be initially effective, their long-term use is associated with multiple side effects. Therefore, there is a great need to develop novel non-opioid alternative therapies to relieve chronic pain. For this purpose, we screened a small library of natural products and their derivatives in the search for pharmacological inhibitors of voltage-gated calcium and sodium channels, which are outstanding molecular targets due to their important roles in nociceptive pathways. We discovered that the acetylated derivative of the ent-kaurane diterpenoid, geopyxin A, 1-O-acetylgeopyxin A, blocks voltage-gated calcium and tetrodotoxin-sensitive voltage-gated sodium channels but not tetrodotoxin-resistant sodium channels in dorsal root ganglion (DRG) neurons. Consistent with inhibition of voltage-gated sodium and calcium channels, 1-O-acetylgeopyxin A reduced reduce action potential firing frequency and increased firing threshold (rheobase) in DRG neurons. Finally, we identified the potential of 1-O-acetylgeopyxin A to reverse mechanical allodynia in a preclinical rat model of HIV-induced sensory neuropathy. Dual targeting of both sodium and calcium channels may permit block of nociceptor excitability and of release of pro-nociceptive transmitters. Future studies will harness the core structure of geopyxins for the generation of antinociceptive drugs.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Ganglios Espinales/efectos de los fármacos , Limoninas/farmacología , Neuralgia/tratamiento farmacológico , Preparaciones Farmacéuticas/administración & dosificación , Bloqueadores de los Canales de Sodio/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio/fisiología , Femenino , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/fisiopatología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/virología , Limoninas/administración & dosificación , Limoninas/química , Neuralgia/metabolismo , Neuralgia/virología , Nociceptores/efectos de los fármacos , Preparaciones Farmacéuticas/metabolismo , Ratas , Ratas Sprague-Dawley , Canales de Sodio/efectos de los fármacos , Canales de Sodio/fisiología , Tetrodotoxina/farmacología
11.
ACS Chem Neurosci ; 10(12): 4834-4846, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31697467

RESUMEN

Naringenin (2S)-5,7-dihydroxy-2-(4-hydroxyphenyl)-3,4-dihydro-2H-1-benzopyran-4-one is a natural flavonoid found in fruits from the citrus family. Because (2S)-naringenin is known to racemize, its bioactivity might be related to one or both enantiomers. Computational studies predicted that (2R)-naringenin may act on voltage-gated ion channels, particularly the N-type calcium channel (CaV2.2) and the NaV1.7 sodium channel-both of which are key for pain signaling. Here we set out to identify the possible mechanism of action of naringenin. Naringenin inhibited depolarization-evoked Ca2+ influx in acetylcholine-, ATP-, and capsaicin-responding rat dorsal root ganglion (DRG) neurons. This was corroborated in electrophysiological recordings from DRG neurons. Pharmacological dissection of each of the voltage-gated Ca2+ channels subtypes could not pinpoint any selectivity of naringenin. Instead, naringenin inhibited NaV1.8-dependent and tetrodotoxin (TTX)-resistant while sparing tetrodotoxin sensitive (TTX-S) voltage-gated Na+ channels as evidenced by the lack of further inhibition by the NaV1.8 blocker A-803467. The effects of the natural flavonoid were validated ex vivo in spinal cord slices where naringenin decreased both the frequency and amplitude of sEPSC recorded in neurons within the substantia gelatinosa. The antinociceptive potential of naringenin was evaluated in male and female mice. Naringenin had no effect on the nociceptive thresholds evoked by heat. Naringenin's reversed allodynia was in mouse models of postsurgical and neuropathic pain. Here, driven by a call by the National Center for Complementary and Integrative Health's strategic plan to advance fundamental research into basic biological mechanisms of the action of natural products, we advance the antinociceptive potential of the flavonoid naringenin.


Asunto(s)
Analgésicos/farmacología , Flavanonas/farmacología , Ganglios Espinales/citología , Canal de Sodio Activado por Voltaje NAV1.8/efectos de los fármacos , Nocicepción/efectos de los fármacos , Células Receptoras Sensoriales/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Sodio/metabolismo , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Canales de Calcio/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Flavanonas/química , Flavanonas/metabolismo , Flavanonas/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/tratamiento farmacológico , Dolor Postoperatorio/tratamiento farmacológico , Conformación Proteica , Mapeo de Interacción de Proteínas , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/clasificación , Células Receptoras Sensoriales/metabolismo , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/uso terapéutico , Organismos Libres de Patógenos Específicos , Relación Estructura-Actividad
12.
Adv Exp Med Biol ; 1155: 923-934, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31468457

RESUMEN

Diabetic neuropathy (DN) is the most common chronic complication of DM and its major pathological changes show axonal dysfunction, atrophy and loss. However, there are few reports that taurine promotes neurite growth of dorsal root ganglion (DRG) cells. In current study, DRG neurons were exposed to high glucose (HG) with or without taurine. The neurite outgrowth of DRG neurons was observed by fluorescent immunohistochemistry method. Expression of Gap-43, Akt, phosphorylated Akt, mTOR and phosphorylated mTOR was determined by Western blot assay. Our results showed that HG significantly decreased the neurite outgrowth and expression of Gap-43 in DRG neurons. Moreover, phosphorylated levels of Akt and mTOR were downregulated in DRG neurons exposed to HG. On the contrary, taurine supplementation significantly reversed the decreased neurite outgrowth and Gap-43 expression, and the downregulated phosphorylated levels of Akt and mTOR. However, the protective effects of taurine were blocked in the presence of PI3K antagonists LY294002 or Akt antagonists Perifosine. These results indicate that taurine promotes neurite outgrowth of DRG neurons exposed to HG via activating Akt/mTOR signal pathway.


Asunto(s)
Ganglios Espinales/citología , Neuronas/efectos de los fármacos , Taurina/farmacología , Células Cultivadas , Proteína GAP-43/metabolismo , Glucosa , Humanos , Neuritas/efectos de los fármacos , Neuronas/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
13.
Biotechniques ; 67(1): 11-15, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31124698

RESUMEN

A large number of animal experiments and clinical trials have confirmed that electrical stimulation can accelerate the growth of axons and recovery of motor function, all of which are inseparable from the formation of myelin. Therefore, establishment of a suitable electrical stimulation platform to study the effects of electrical stimulation on the myelin process of dorsal root ganglia and Schwann cells is of great significance for understanding the recovery of electrical stimulation. We designed a simple conductive glass cell culture system to overcome the shortcomings of direct contact of the electrode with the culture solution, and the number of culture chambers can be selected based on the purpose of the experiment in order to reduce experimental time and cost.


Asunto(s)
Técnicas de Cocultivo/instrumentación , Estimulación Eléctrica/instrumentación , Ganglios Espinales/citología , Vaina de Mielina/metabolismo , Células de Schwann/citología , Animales , Células Cultivadas , Técnicas de Cocultivo/métodos , Estimulación Eléctrica/métodos , Terapia por Estimulación Eléctrica , Diseño de Equipo , Ganglios Espinales/metabolismo , Ratas Sprague-Dawley , Células de Schwann/metabolismo
14.
Pain ; 160(1): 136-150, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30157131

RESUMEN

Clinical studies indicate that cannabidiol (CBD), the primary nonaddictive component of cannabis that interacts with the serotonin (5-HT)1A receptor, may possess analgesic and anxiolytic effects. However, its effects on 5-HT neuronal activity, as well as its impact on models of neuropathic pain are unknown. First, using in vivo single-unit extracellular recordings in rats, we demonstrated that acute intravenous (i.v.) increasing doses of CBD (0.1-1.0 mg/kg) decreased the firing rate of 5-HT neurons in the dorsal raphe nucleus, which was prevented by administration of the 5-HT1A antagonist WAY 100635 (0.3 mg/kg, i.v.) and the TRPV1 antagonist capsazepine (1 mg/kg, i.v.) but not by the CB1 receptor antagonist AM 251 (1 mg/kg, i.v.). Repeated treatment with CBD (5 mg/kg/day, subcutaneously [s.c.], for 7 days) increased 5-HT firing through desensitization of 5-HT1A receptors. Rats subjected to the spared nerve injury model for 24 days showed decreased 5-HT firing activity, mechanical allodynia, and increased anxiety-like behavior in the elevated plus maze test, open-field test, and novelty-suppressed feeding test. Seven days of treatment with CBD reduced mechanical allodynia, decreased anxiety-like behavior, and normalized 5-HT activity. Antiallodynic effects of CBD were fully prevented by capsazepine (10 mg/kg/day, s.c., for 7 days) and partially prevented by WAY 100635 (2 mg/kg/day, s.c., for 7 days), whereas the anxiolytic effect was blocked only by WAY. Overall, repeated treatment with low-dose CBD induces analgesia predominantly through TRPV1 activation, reduces anxiety through 5-HT1A receptor activation, and rescues impaired 5-HT neurotransmission under neuropathic pain conditions.


Asunto(s)
Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Cannabidiol/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Neuralgia/complicaciones , Serotonina/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Capsaicina/análogos & derivados , Capsaicina/farmacología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Ganglios Espinales/citología , Hiperalgesia/terapia , Dietilamida del Ácido Lisérgico/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Neuralgia/patología , Piperazinas/uso terapéutico , Piperidinas/farmacología , Pirazoles/farmacología , Piridinas/uso terapéutico , Ratas , Ratas Wistar , Antagonistas de la Serotonina/farmacología , Natación
15.
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
16.
Pain ; 160(2): 508-527, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30335684

RESUMEN

Sensitization of the transient receptor potential ion channel vanilloid 1 (TRPV1) is critically involved in inflammatory pain. To date, manifold signaling cascades have been shown to converge onto TRPV1 and enhance its sensitization. However, many of them also play a role for nociceptive pain, which limits their utility as targets for therapeutic intervention. Here, we show that the vesicle transport through interaction with t-SNAREs homolog 1B (Vti1b) protein promotes TRPV1 sensitization upon inflammation in cell culture but leaves normal functioning of TRPV1 intact. Importantly, the effect of Vti1b can be recapitulated in vivo: Virus-mediated knockdown of Vti1b in sensory neurons attenuated thermal hypersensitivity during inflammatory pain without affecting mechanical hypersensitivity or capsaicin-induced nociceptive pain. Interestingly, TRPV1 and Vti1b are localized in close vicinity as indicated by proximity ligation assays and are likely to bind to each other, either directly or indirectly, as suggested by coimmunoprecipitations. Moreover, using a mass spectrometry-based quantitative interactomics approach, we show that Vti1b is less abundant in TRPV1 protein complexes during inflammatory conditions compared with controls. Alongside, we identify numerous novel and pain state-dependent binding partners of native TRPV1 in dorsal root ganglia. These data represent a unique resource on the dynamics of the TRPV1 interactome and facilitate mechanistic insights into TRPV1 regulation. We propose that inflammation-related differences in the TRPV1 interactome identified here could be exploited to specifically target inflammatory pain in the future.


Asunto(s)
Regulación de la Expresión Génica/genética , Hiperalgesia/genética , Dolor/metabolismo , Proteínas Qb-SNARE/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Capsaicina/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Ganglios Espinales/citología , Humanos , Hiperalgesia/fisiopatología , Inflamación/inducido químicamente , Inflamación/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dolor/etiología , Proteínas Qb-SNARE/genética , Interferencia de ARN/fisiología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Transducción de Señal , Canales Catiónicos TRPV/genética
17.
ACS Appl Mater Interfaces ; 11(1): 356-372, 2019 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-30516370

RESUMEN

Magnetic electrospun fibers are of interest for minimally invasive biomaterial applications that also strive to provide cell guidance. Magnetic electrospun fibers can be injected and then magnetically positioned in situ, and the aligned fiber scaffolds provide consistent topographical guidance to cells. In this study, magnetically responsive aligned poly-l-lactic acid electrospun fiber scaffolds were developed and tested for neural applications. Incorporating oleic acid-coated iron oxide nanoparticles significantly increased neurite outgrowth, reduced the fiber alignment, and increased the surface nanotopography of the electrospun fibers. After verifying neuron viability on two-dimensional scaffolds, the system was tested as an injectable three-dimensional scaffold. Small conduits of aligned magnetic fibers were easily injected in a collagen or fibrinogen hydrogel solution and repositioned using an external magnetic field. The aligned magnetic fibers provided internal directional guidance to neurites within a three-dimensional collagen or fibrin model hydrogel, supplemented with Matrigel. Neurites growing from dorsal root ganglion explants extended 1.4-3× farther on the aligned fibers compared with neurites extending in the hydrogel alone. Overall, these results show that magnetic electrospun fiber scaffolds can be injected and manipulated with a magnetic field in situ to provide directional guidance to neurons inside an injectable hydrogel. Most importantly, this injectable guidance system increased both neurite alignment and neurite length within the hydrogel scaffold.


Asunto(s)
Ganglios Espinales/fisiología , Hidrogeles/química , Regeneración Nerviosa , Neuritas/metabolismo , Andamios del Tejido/química , Animales , Ganglios Espinales/citología , Ratas , Ratas Sprague-Dawley
18.
Anat Rec (Hoboken) ; 301(10): 1628-1637, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30334365

RESUMEN

Peripheral nerve injuries (PNI) have a high prevalence and can be debilitating, resulting in life-long loss or disturbance in end-organ function, which compromises quality of life for patients. Current therapies use microsurgical approaches but there is the potential for enhancing recovery through other therapeutic modalities such as; cell-based conduits, gene therapy and small molecules. A number of molecular targets and drugs which have the potential to improve nerve regeneration have been identified, however, there are challenges associated with moving therapies toward clinical translation. Due to the lack of detailed knowledge about the pro-regenerative effect of potential drug treatments, there is a need for effective in vitro models to screen compounds to inform future pre-clinical and clinical studies. The interaction between regenerating neurites and supporting Schwann cells is a key feature of the nerve environment, therefore, in vitro models that mimic this cellular association are useful tools. In this study, we have investigated various cell culture models, including simple monolayer systems and more complex 3D-engineered co-cultures, as models for use in PNI drug development. Anat Rec, 301:1628-1637, 2018. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Asunto(s)
Técnicas de Cocultivo/métodos , Evaluación Preclínica de Medicamentos/métodos , Regeneración Nerviosa/efectos de los fármacos , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Descubrimiento de Drogas , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Ibuprofeno/farmacología , Ibuprofeno/uso terapéutico , Células PC12 , Ratas , Ratas Sprague-Dawley , Ratas Wistar
19.
Acta Biochim Biophys Sin (Shanghai) ; 50(12): 1219-1226, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30339176

RESUMEN

CC chemokine ligand 2 (CCL2) has been implicated in pathological pain, but the mechanism underlying the pronociceptive effect of CCL2 is not fully understood. Voltage-gated sodium (Nav) channels are important determinants of the excitability of sensory neurons. Hence we tested the hypothesis that CCL2 contributes to inflammatory pain via modulating Nav channel activity of primary afferent neurons. Chronic inflammatory pain was induced in rats by intraplantar injection of the complete Freud adjuvant (CFA) to one of the hind paws. Control rats received intraplantar injection of equal volume of saline. A significant increase of CCL2 mRNA and CCL2 receptor (CCR2) protein expression was detected in the ipsilateral dorsal root ganglion (DRG) in CFA-treated rats. Intraplantar injection of CCL2 protein in the control rats had minimal effect on the paw withdrawal threshold (PWT) in response to mechanical stimulation. However, in CFA-treated rats, intraplantar CCL2 led to an increase in pain responses. Patch-clamp recording of acutely dissociated DRG neurons revealed that CCL2 had minimum effect on the excitability of sensory neurons from control rats. However, CCL2 directly depolarized a large proportion of small to medium-sized sensory neurons from CFA-treated rats. In addition, CCL2 was found to enhance whole-cell TTX-sensitive sodium currents without significantly affecting the TTX-resistant sodium currents and the potassium currents. These results are in agreement with previous reports concerning the involvement of CCL2-CCR2 signaling in inflammatory hyperalgesia and further indicate that enhanced TTX-sensitive channel activity may partly underlie the pronociceptive effects of CCL2.


Asunto(s)
Quimiocina CCL2/farmacología , Inflamación/metabolismo , Neuronas Aferentes/efectos de los fármacos , Dolor/metabolismo , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Sinergismo Farmacológico , Adyuvante de Freund , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Masculino , Potenciales de la Membrana/efectos de los fármacos , Neuronas Aferentes/metabolismo , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/genética
20.
Pain ; 159(11): 2383-2393, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30015706

RESUMEN

The exchange proteins activated by cAMP (Epacs) have been shown to play important roles in producing inflammation-induced nociception. Transient receptor potential vanilloid type 1 (TRPV1) is a major receptor processing thermal and chemosensitive nociceptive information. The role of Epacs in modulating the activity of TRPV1 has yet to be determined. Studying the effect of complete Freund adjuvant (CFA)-induced inflammation on capsaicin-activated TRPV1 nociceptive responses in dorsal root ganglia (DRG), we found that CFA produced a large increase in capsaicin-induced responses. The increase was inhibited by Epac1 and Epac2 antagonists. Thus, activation of Epacs is critical in producing enhancement in TRPV1-mediated responses under inflammatory conditions. In addition, the inflammation-induced enhancement of TRPV1 responses was blocked by PKCα and PKCε inhibitors, suggesting the essential roles of these PKCs in enhancing TRPV1 responses. To determine the mechanism underlying the Epac actions on TRPV1, we studied the effects of the Epac activator, 8-(4-chlorophenylthio)-2-O-methyl-cAMP (CPT), on capsaicin-induced nociceptive behavioral responses, capsaicin-activated currents, expression and membrane trafficking of PKC and TRPV1 in DRG. CPT was found to enhance capsaicin-induced nociception and ionic currents. The enhancement was inhibited by PKCα and PKCε inhibitors. In addition, CPT increased the expression of phosphorylated PKCα (pPKCα) and membrane TRPV1 expression in DRG. Studying the colocalization of TRPV1 and pPKCα or pPKCε in DRG slices prepared from CFA-treated rats, we found that pPKCα or pPKCε expressed with TRPV1 in different-sized neurons to exert differential influences on TRPV1 activity. Thus, Epac-PKC signaling is critically important in producing inflammation-induced potentiation of TRPV1 functions.


Asunto(s)
Acetilcisteína/análogos & derivados , Eritromicina/análogos & derivados , Hiperalgesia/fisiopatología , Inflamación/enzimología , Proteína Quinasa C-epsilon/metabolismo , Transducción de Señal/fisiología , Canales Catiónicos TRPV/metabolismo , Acetilcisteína/metabolismo , Acetilcisteína/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Biotinilación , Capsaicina/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Eritromicina/metabolismo , Eritromicina/farmacología , Adyuvante de Freund/toxicidad , Ganglios Espinales/citología , Hiperalgesia/patología , Inflamación/inducido químicamente , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Proteína Quinasa C-alfa/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA