Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Pharm Biol ; 57(1): 649-659, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31545916

RESUMEN

Context: Dexmedetomidine (Dex) has been reported to have an anti-inflammatory effect. However, its role on osteoarthritis (OA) has not been explored. Objective: This study investigates the effect of Dex on OA rat model induced by papain. Materials and methods: The OA Wistar rat model was induced by intraluminal injection of 20 mL of papain mixed solution (4% papain 0.2 mL mixed with 0.03 mol L-1 l-cysteine 0.1 mL) into the right knee joint. Two weeks after papain injection, OA rats were treated by intra-articular injection of Dex (5, 10, or 20 µg kg-1) into the right knee (once a day, continuously for 4 weeks). Articular cartilage tissue was obtained after Dex treatment was completed. Results: The gait behavior scores (2.83 ± 0.49), PWMT (15.2 ± 1.78) and PTWL (14.81 ± 0.92) in H-DEX group were higher than that of OA group, while Mankin score (5.5 ± 0.81) was decreased (p < 0.05). Compared with the OA group, the IL-1ß (153.11 ± 16.05 pg mg-1), IL-18 (3.71 ± 0.7 pg mg-1), IL-6 (14.15 ± 1.94 pg/mg) and TNF-α (40.45 ± 10.28 pg mg-1) levels in H-DEX group were decreased (p < 0.05). MMP-13, NLRP3, and caspase-1 p10 expression in Dex groups were significantly lower than that of OA group (p < 0.05), while collagen II was increased (p < 0.05). p65 in the nucleus of Dex groups was significantly down-regulated than that of OA group (p < 0.05). Discussion and Conclusions: Dex can improve pain symptoms and cartilage tissue damage of OA rats, which may be related to its inhibition of the activation of NF-κB and NLRP3 inflammasome.


Asunto(s)
Antiinflamatorios/farmacología , Dexmedetomidina/farmacología , Inflamasomas/efectos de los fármacos , Inflamación/tratamiento farmacológico , FN-kappa B/metabolismo , Osteoartritis/tratamiento farmacológico , Animales , Cartílago Articular/efectos de los fármacos , Masculino , Metaloproteinasa 13 de la Matriz , Modelos Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Osteoartritis/inducido químicamente , Papaína/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
2.
J Pain Res ; 11: 599-609, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29606887

RESUMEN

OBJECTIVE: The analgesic effect on chronic pain of peripheral nerve stimulation (PNS) has been proven, but its underlying mechanism remains unknown. Therefore, this study aimed to assess the analgesic effect of PNS on bone cancer pain in a rat model and to explore the underlying mechanism. MATERIALS AND METHODS: PNS on sciatic nerves with bipolar electrode was performed in both naïve and bone cancer pain model rats. Then, the protein levels of activity-regulated cytoskeleton-associated protein (Arc), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptor 1 (GluA1), and phosphate N-methyl-d-aspartic acid-type glutamate receptor subunit 2B (pGluNR2B) in spinal cord were evaluated by immunohistochemistry and Western blotting. Thermal paw withdraw latency and mechanical paw withdraw threshold were used to estimate the analgesic effect of PNS on bone cancer pain. Intrathecal administration of Arc shRNA was used to inhibit Arc expression in the spinal cord. RESULTS: PNS at 60 and 120 Hz for 20 min overtly induced Arc expression in the spinal cord, increased thermal pain thresholds in naïve rats, and relieved bone cancer pain; meanwhile, 10 Hz PNS did not achieve those results. In addition, PNS at 60 and 120 Hz also reduced the expression of GluA1, but not pGluNR2B, in the spinal cord. Finally, the anti-nociceptive effect and GluA1 downregulation induced by PNS were inhibited by intrathecal administration of Arc shRNA. CONCLUSION: PNS (60 Hz, 0.3 mA) can relieve bone-cancer-induced allodynia and hyperalgesia by upregulating Arc protein expression and then by decreasing GluA1 transcription in the spinal cord dorsal horn.

3.
Mol Pain ; 14: 1744806918767560, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29607715

RESUMEN

Treating bone cancer pain continues to be a clinical challenge and underlying mechanisms of bone cancer pain remain elusive. Here, we reported that sonic hedgehog signaling plays a critical role in the development of bone cancer pain. Tibia bone cavity tumor cell implantation produces bone cancer-related mechanical allodynia, thermal hyperalgesia, and spontaneous and movement-evoked pain behaviors. Production and persistence of these pain behaviors are well correlated with tumor cell implantation-induced up-regulation and activation of sonic hedgehog signaling in primary sensory neurons and spinal cord. Spinal administration of sonic hedgehog signaling inhibitor cyclopamine prevents and reverses the induction and persistence of bone cancer pain without affecting normal pain sensitivity. Inhibiting sonic hedgehog signaling activation with cyclopamine, in vivo or in vitro, greatly suppresses tumor cell implantation-induced increase of intracellular Ca2+ and hyperexcitability of the sensory neurons and also the activation of GluN2B receptor and the subsequent Ca2+-dependent signals CaMKII and CREB in dorsal root ganglion and the spinal cord. These findings show a critical mechanism underlying the pathogenesis of bone cancer pain and suggest that targeting sonic hedgehog signaling may be an effective approach for treating bone cancer pain.


Asunto(s)
Neoplasias Óseas/complicaciones , Dolor en Cáncer/etiología , Dolor en Cáncer/patología , Proteínas Hedgehog/metabolismo , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología , Transducción de Señal , Animales , Calcio/metabolismo , Dolor en Cáncer/metabolismo , Línea Celular Tumoral , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Espacio Intracelular/metabolismo , Trasplante de Neoplasias , Nocicepción , Ratas Sprague-Dawley , Médula Espinal/patología , Regulación hacia Arriba
4.
J Pain ; 19(2): 186-195, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29079540

RESUMEN

Glial cell hyperactivity has been proposed to be responsible for chronic pain, however, the mechanisms remain unclear. Interleukin (IL)-18, released from glial cells, has been reported to be involved in neuropathic pain. In this study, we investigated the role of IL-18 in bone cancer pain. Bone cancer pain was mimicked by injecting Walker-256 mammary gland carcinoma cells into the intramedullary space of the tibia in rats. Expression and location of IL-18 and the IL-18 receptor were tested. To investigate the contribution of IL-18 signaling to bone cancer pain, IL-18 binding protein and recombinant IL-18 were used. To investigate the mechanisms of glial cells effects, MK801, N-methyl-D-aspartate (NMDA) receptor inhibitor, and Src kinase-specific inhibitor PP1 were used. Tumor cell implantation (TCI) treatment increased expression of IL-18 and IL-18 receptor in spinal cord. The time course of IL-18 upregulation was correlated with TCI-induced pain behaviors. Blocking the IL-18 signaling pathway prevented and reversed bone cancer-related pain behaviors. Meanwhile, blocking IL-18 signaling also suppressed TCI-induced glial cell hyperactivity, as well as activation of GluN2B and subsequent Ca2+-dependent signaling. Spinal administration of recombinant IL-18 in naive rat induced significant mechanical allodynia, as well as GluN2B activation. However, intrathecal injection of MK801 failed to suppress recombinant IL-18-induced GluN2B phosphorylation, whereas Src kinase inhibitor PP1 significantly inhibited IL-18-induced GluN2B activation. IL-18-mediated glial-glia and glial-neuron interaction may facilitate bone cancer pain. Blocking IL-18 signaling may effectively prevent and/or suppress bone cancer pain. PERSPECTIVE: IL-18 signaling may be a new target for cancer pain therapy.


Asunto(s)
Dolor en Cáncer/patología , Interleucina-18/metabolismo , Neuroglía/fisiología , Neuronas/fisiología , Médula Espinal/patología , Animales , Neoplasias Óseas/complicaciones , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/etiología , Carcinoma/complicaciones , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Inyecciones Espinales , Interleucina-18/efectos adversos , Ratas , Receptores de Interleucina-18/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mediators Inflamm ; 2015: 752512, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26819501

RESUMEN

OBJECTIVE: The present study is to investigate the analgesic roles of L-THP in rats with bone cancer pain caused by tumor cell implantation (TCI). METHODS: Thermal hyperalgesia and mechanical allodynia were measured at different time points before and after operation. L-THP (20, 40, and 60 mg/kg) were administrated intragastrically at early phase of postoperation (before pain appearance) and later phase of postoperation (after pain appearance), respectively. The concentrations of TNF-α, IL-1ß, and IL-18 in spinal cord were measured by enzyme-linked immunosorbent assay. Western blot was used to test the activation of astrocytes and microglial cells in spinal cord after TCI treatment. RESULTS: TCI treatment induced significant thermal hyperalgesia and mechanical allodynia. Administration of L-THP at high doses significantly prevented and/or reversed bone cancer-related pain behaviors. Besides, TCI-induced activation of microglial cells and the increased levels of TNF-α and IL-18 were inhibited by L-THP administration. However, L-THP failed to affect TCI-induced astrocytes activation and IL-1ß increase. CONCLUSION: This study suggests the possible clinical utility of L-THP in the treatment of bone cancer pain. The analgesic effects of L-THP on bone cancer pain maybe underlying the inhibition of microglial cells activation and proinflammatory cytokines increase.


Asunto(s)
Alcaloides de Berberina/farmacología , Neoplasias Óseas/fisiopatología , Microglía/efectos de los fármacos , Dolor Intratable/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Femenino , Hiperalgesia/tratamiento farmacológico , Interleucina-18/análisis , Microglía/fisiología , Trasplante de Neoplasias , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/análisis
6.
Behav Pharmacol ; 25(4): 267-76, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24978483

RESUMEN

The objective of this study was to explore the role of cyclic adenosine monophosphate-protein kinase A (cAMP-PKA) signaling in the development of bone cancer pain in rats. Female Sprague-Dawley rats (N=48) were divided randomly into four groups: sham (n=8), tumor cell implantation (TCI) (n=16), TCI+saline (n=8), and TCI+PKA inhibitor (n=16). Bone cancer-induced pain behaviors - thermal hyperalgesia and mechanical allodynia - were tested at postoperative days -3, -1, 1, 3, 5, 7, 10, and 14. A PKA inhibitor, Rp-cAMPS (1 mmol/l/20 µl), was injected intrathecally on postoperative days 3, 4, and 5 (early phase) or 7, 8, and 9 postoperative days (late phase). The expression of PKA mRNA in dorsal root ganglia (DRG) was detected by reverse transcription-PCR. The concentration of cAMP and activity of PKA in DRG and spinal cord were measured by enzyme-linked immunosorbent assay. TCI treatment induced significant pain behaviors, manifested as thermal hyperalgesia and mechanical allodynia. Spinal administration of the PKA inhibitor Rp-cAMPS during the early phase and late phase significantly delayed or reversed, respectively, TCI-induced thermal hyperalgesia and mechanical allodynia. TCI treatment also led to obvious tumor growth and bone destruction. The level of PKA mRNA in the DRG, as well as the concentration of cAMP and the activity of PKA, in both the DRG and spinal cord were significantly increased after TCI treatment (P<0.01). We conclude that the inhibition of the cAMP-PKA signaling pathway may reduce bone cancer pain.


Asunto(s)
Neoplasias Óseas/fisiopatología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Ganglios Espinales/fisiopatología , Dolor/fisiopatología , Médula Espinal/fisiopatología , Animales , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Modelos Animales de Enfermedad , Ciclo Estral/fisiología , Femenino , Ganglios Espinales/efectos de los fármacos , Calor , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Trasplante de Neoplasias , Dolor/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Tionucleótidos/farmacología , Factores de Tiempo , Tacto
7.
Spine (Phila Pa 1976) ; 39(19): 1533-41, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24921837

RESUMEN

STUDY DESIGN: A prospective, randomized experimental research. OBJECTIVE: To demonstrate the role of cGMP (cyclic guanosine monophosphate)-cGKI (cGMP-dependent protein kinase I) pathway in dorsal root ganglia (DRG) in bone cancer pain. SUMMARY OF BACKGROUND DATA: Treating bone cancer pain continues to possess a major clinical challenge because the specific cellular and molecular mechanisms underlying bone cancer pain remain elusive. cGMP and cGMP-dependent protein kinases pathway in DRG plays important role in nerve injury-induced hyperexcitability of DRG neurons, as well as neuropathic pain, however, whether this pathway participates in bone cancer pain is unknown. METHODS: The rat model of bone cancer pain was produced by intramedullary injection of rat breast cancer cells (Walker 256) into right tibia. Thermal hyperalgesia and mechanical allodynia were measured before and after administration of inhibitor of cGMP-cGKs pathway (Rp-8-pCPT-cGMPS). Immunofluorescence and reverse transcription-polymerase chain reaction were used to reflect expression of cGKI in DRG neurons, whereas the concentration of cGMP in DRG was tested using enzyme-linked immunosorbent assay method. Whole-cell patch clamp was used to record the hyperexcitability of small neurons in DRG with or without cGKs inhibitor after tumor cell implantation (TCI). RESULTS: TCI treatment significantly increased the concentration of cGMP in DRG and activity of cGKs in DRG and the spinal cord. TCI treatment also induced upregulation of cGKI messenger ribonucleic acid and protein in DRG, as well as enhanced hyperexcitability in DRG neurons. Spinal administration of Rp-8-pCPT-cGMPS, cGMP-cGKs inhibitor, significantly suppressed TCI-induced activation of cGMP-cGKI signaling, and hyperexcitability of DRG neurons. Meanwhile, in vivo intrathecal delivery of the Rp-8-pCPT-cGMPS significantly prevented and suppressed TCI-induced hyperalgesia and allodynia. CONCLUSION: From these results, we confirm that TCI treatment activates cGMP-cGKI signaling pathway and continuing activation of this pathway in DRG is required for hyperalgesia and/or hyperalgesia and allodynia after TCI treatment. LEVEL OF EVIDENCE: N/A.


Asunto(s)
Neoplasias Óseas/secundario , Carcinoma 256 de Walker/secundario , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/fisiología , GMP Cíclico/fisiología , Ganglios Espinales/fisiopatología , Hiperalgesia/fisiopatología , Proteínas de Neoplasias/fisiología , Células Receptoras Sensoriales/fisiología , Tibia , Animales , Neoplasias Óseas/fisiopatología , Carcinoma 256 de Walker/fisiopatología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/biosíntesis , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Inducción Enzimática , Femenino , Calor , Hiperalgesia/etiología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Umbral del Dolor , Técnicas de Placa-Clamp , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Tionucleótidos/farmacología , Tibia/inervación , Tacto
8.
Pain ; 154(12): 2823-2835, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23973554

RESUMEN

Treating bone cancer pain poses a major clinical challenge, and the mechanisms underlying bone cancer pain remain elusive. EphrinB-EphB receptor signaling may contribute to bone cancer pain through N-methyl-d-aspartate receptor neuronal mechanisms. Here, we report that ephrinB-EphB signaling may also act through a Toll-like receptor 4 (TLR4)-glial cell mechanism in the spinal cord. Bone cancer pain was induced by tibia bone cavity tumor cell implantation (TCI) in rats. TCI increased the expression of TLR4 and the EphB1 receptor, the activation of astrocytes and microglial cells, and increased levels of interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α). The increased expression of TLR4 and EphB1 were colocalized with each other in astrocytes and microglial cells. Spinal knockdown of TLR4 suppressed TCI-induced behavioral signs of bone cancer pain. The TCI-induced activation of astrocytes and microglial cells, as well as the increased levels of IL-1ß and TNF-α, were inhibited by intrathecal administration of TLR4-targeting siRNA2 and the EphB receptor antagonist EphB2-Fc, respectively. The administration of EphB2-Fc suppressed the TCI-induced increase of TLR4 expression but siRNA2 failed to affect TCI-induced EphB1 expression. Intrathecal administration of an exogenous EphB1 receptor activator, ephrinB2-Fc, increased the expression of TLR4 and the levels of IL-1ß and TNF-α, activated astrocytes and microglial cells, and induced thermal hypersensitivity. These ephrinB2-Fc-induced alterations were suppressed by spinal knockdown of TLR4. This study suggests that TLR4 may be a potential target for preventing or reversing bone cancer pain and other similar painful processes mediated by ephrinB-EphB receptor signaling.


Asunto(s)
Neoplasias Óseas/metabolismo , Efrina-B2/administración & dosificación , Mediadores de Inflamación/metabolismo , Receptor EphB1/metabolismo , Médula Espinal/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Neoplasias Óseas/patología , Femenino , Inyecciones Espinales , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Receptor EphB1/agonistas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
9.
J Clin Invest ; 123(5): 2268-86, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23585476

RESUMEN

Treating neuropathic pain is a major clinical challenge, and the underlying mechanisms of neuropathic pain remain elusive. We hypothesized that neuropathic pain-inducing nerve injury may elicit neuronal alterations that recapitulate events that occur during development. Here, we report that WNT signaling, which is important in developmental processes of the nervous system, plays a critical role in neuropathic pain after sciatic nerve injury and bone cancer in rodents. Nerve injury and bone cancer caused a rapid-onset and long-lasting expression of WNTs, as well as activation of WNT/frizzled/ß-catenin signaling in the primary sensory neurons, the spinal dorsal horn neurons, and astrocytes. Spinal blockade of WNT signaling pathways inhibited the production and persistence of neuropathic pain and the accompanying neurochemical alterations without affecting normal pain sensitivity and locomotor activity. WNT signaling activation stimulated production of the proinflammatory cytokines IL-18 and TNF-α and regulated the NR2B glutamate receptor and Ca2+-dependent signals through the ß-catenin pathway in the spinal cord. These findings indicate a critical mechanism underlying the pathogenesis of neuropathic pain and suggest that targeting the WNT signaling pathway may be an effective approach for treating neuropathic pain, including bone cancer pain.


Asunto(s)
Neuralgia/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Calcio/metabolismo , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Interleucina-18/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Pain ; 152(11): 2605-2615, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21907492

RESUMEN

Adenosine triphosphate-sensitive potassium (K(ATP)) channels are suggested to be involved in pathogenesis of neuropathic pain, but remain underinvestigated in primary afferents and in the spinal cord. We examined alterations of K(ATP) channels in rat spinal cord and tested whether and how they could contribute to neuropathic pain. The results showed that protein expression for K(ATP) channel subunits SUR1, SUR2, and Kir6.1, but not Kir6.2, were significantly downregulated and associated with thermal hyperalgesia and mechanical allodynia after sciatic nerve injury. Spinal administration of a K(ATP) channel opener cromakalim (CRO, 5, 10, and 20 µg, respectively) prevented or suppressed, in a dose-dependent manner, the hyperalgesia and allodynia. Nerve injury also significantly increased expression and phosphorylation of connexin 43, an astroglial gap junction protein. Such an increase of phosphorylation of connexin 43 was inhibited by CRO treatment. Furthermore, preadministration of an astroglial gap junction decoupler carbenoxolone (10 µg) completely reversed the inhibitory effects of CRO treatment on the hyperalgesia and allodynia and phosphorylation of NR1 and NR2B receptors and the subsequent activation of Ca(2+)-dependent signals Ca(2+)/calmodulin-dependent kinase II and cyclic adenosine monophosphate (cAMP) response element binding protein. These findings suggest that nerve injury-induced downregulation of the K(ATP) channels in the spinal cord may interrupt the astroglial gap junctional function and contribute to neuropathic pain, thus the K(ATP) channels opener can reduce neuropathic pain probably partly via regulating the astroglial gap junctions. This study may provide a new strategy for treating neuropathic pain using K(ATP) channel openers in the clinic.


Asunto(s)
Cromakalim/farmacología , Canales KATP/agonistas , Canales KATP/fisiología , Neuralgia/tratamiento farmacológico , Neuralgia/fisiopatología , Médula Espinal/fisiopatología , Transportadoras de Casetes de Unión a ATP/efectos adversos , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/fisiología , Hiperalgesia/fisiopatología , Masculino , Canales de Potasio de Rectificación Interna/efectos adversos , Canales de Potasio de Rectificación Interna/agonistas , Canales de Potasio de Rectificación Interna/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Droga/fisiología , Ciática/tratamiento farmacológico , Ciática/fisiopatología , Médula Espinal/efectos de los fármacos , Receptores de Sulfonilureas
11.
Cancer Res ; 71(13): 4392-402, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21555368

RESUMEN

Treating bone cancer pain continues to be a clinical challenge and underlying mechanisms of bone cancer pain remain elusive. Here, we report that EphB1 receptor forward signaling in the spinal cord is critical to the development of bone cancer pain and morphine tolerance in treating bone cancer pain. Tibia bone cavity tumor cell implantation (TCI) produces bone cancer-related thermal hyperalgesia, mechanical allodynia, spontaneous and movement-evoked pain behaviors, and bone destruction. Production and persistence of these pain behaviors are well correlated with TCI-induced upregulation of EphB1 receptor and its ligand ephrinB2 in the dorsal horn and primary sensory neurons. Spinal administration of an EphB1 receptor blocking reagent EphB2-Fc prevents and reverses bone cancer pain behaviors and the associated induction of c-Fos and activation of astrocytes and microglial cells, NR1 and NR2B receptors, Src within the N-methyl-D-aspartate receptor complex, and the subsequent Ca(2+)-dependent signals. The exogenous ligand ephrinB2-Fc upregulates level of phosphorylation of NR1 and NR2B receptors depending on the activation of EphB1 receptor. Spinal administration of EphB2-Fc and ephrinB2-Fc induces downregulation of EphB1 and ephrinB2, respectively, accompanied with increased activity of matrix metalloproteinase (MMP)-2/9. Blocking MMP-2 or MMP-9 reverses EphB1-Fc treatment-induced downregulation of EphB1 receptor. In addition, spinal blocking or targeted mutation of EphB1 receptor reverses morphine tolerance in treating bone cancer pain in rats and defensive pain in mice. These findings show a critical mechanism underlying the pathogenesis of bone cancer pain and suggest a potential target for treating bone cancer pain and improving analgesic effect of morphine clinically.


Asunto(s)
Analgésicos Opioides/farmacología , Neoplasias Óseas/metabolismo , Morfina/farmacología , Dolor/tratamiento farmacológico , Dolor/metabolismo , Receptor EphB1/antagonistas & inhibidores , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Animales , Neoplasias Óseas/complicaciones , Neoplasias Óseas/patología , Regulación hacia Abajo , Efrina-B2/biosíntesis , Efrina-B2/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Transgénicos , Dolor/etiología , Ratas , Ratas Sprague-Dawley , Receptor EphB1/biosíntesis , Receptor EphB1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA