Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Neurosci ; 55(3): 661-674, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35075718

RESUMO

Bone cancer pain (BCP) is the most frequently observed chronic cancer pain, and its development remains largely unexplored. Dysregulation of non-coding RNAs greatly contributes to the pathogenesis of BCP. In the present study, we found a new long noncoding RNA (lncRNA), NONRATT009773.2, and investigated its role in the spinal cord of BCP rats. Our results showed that NONRATT009773.2 was significantly up-regulated in BCP model rats, whereas depletion of NONRATT009773.2 attenuated BCP. In contrast, overexpression of NONRATT009773.2 triggered pain-like symptoms in normal animals. Moreover, NONRATT009773.2 functioned as a microRNA (miRNA) sponge to absorb miR-708-5p and up-regulated miRNA downstream target CXCL13, which plays fundamental roles in the initiation and maintenance of neuroinflammation and hyperalgesia. Collectively, our current findings indicated that NONRATT009773.2 could be employed as a new therapeutic target for BCP.


Assuntos
Dor do Câncer , MicroRNAs , Neoplasias , RNA Longo não Codificante , Animais , Dor do Câncer/genética , Dor do Câncer/patologia , Hiperalgesia/genética , MicroRNAs/genética , Neoplasias/patologia , RNA Longo não Codificante/genética , Ratos , Medula Espinal/patologia
2.
Ann Transl Med ; 10(24): 1377, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36660643

RESUMO

Background: Vascular endothelial barrier disruption is pivotal in the development of acute and chronic pain. Here, we demonstrate a previously unidentified molecular mechanism in which activation of the peripheral Epac1/p-Cav-1 pathway accelerated the disruption of the vascular endothelial barrier, thereby promoting chronic postsurgical pain (CPSP). Methods: We established a rat model of CPSP induced by skin/muscle incision and retraction (SMIR). Pain behaviors were assessed by the mechanical withdrawal threshold (MWT) at different times. Local muscle tissues around the incision were isolated to detect the vascular permeability and the expression of Epac1 and Cav-1. They were assessed by western blot and immunofluorescence staining. Results: SMIR increased vascular endothelial permeability and the number of macrophages and endothelial cells in the muscle tissues around the incision. The peripheral upregulation of Epac1 was macrophage-derived, whereas that of p-Cav-1 was both macrophage and endothelial cell-derived in the SMIR model. Moreover, the Epac1 agonist 8-pCPT could induce mechanical sensitivity, increase the expression of p-Cav-1, and disrupt vascular endothelial barrier in normal rats. The Epac1 inhibitor CE3F4 attenuated established SMIR-induced mechanical hyperalgesia, the upregulation of p-Cav-1 and vascular endothelial barrier. Finally, we showed that intrathecal injection of Cav-1siRNA relieved SMIR-induced mechanical allodynia, but had no effects of the expression of Epac1. Conclusions: Collectively, these results revealed a molecular mechanism for modulating CPSP through the peripheral Epac1/Cav-1 pathway. Importantly, targeting Epac1/Cav-1 signaling might be a potential treatment for CPSP.

3.
Exp Ther Med ; 22(5): 1289, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34630644

RESUMO

Chronic postsurgical pain (CPSP) has a high incidence, but the underlying mechanisms remain elusive. Previous studies have indicated that caveolin-1 (Cav-1) plays a notable role in pain modulation. To study the role of Cav-1 in CPSP in the present study, a rat model of skin/muscle incision and retraction (SMIR) was established. Under anesthesia, skin and superficial muscle of the medial thigh were incised and a small pair of retractors inserted. It was revealed that SMIR increased the expression of Cav-1 in the dorsal root ganglion (DRG) and the injured tissue around the incision. Furthermore, the infiltration of endothelial cells and macrophages in the injured tissue around the incision increased constantly, and the vascular permeability increased due to the destruction of the vascular endothelial barrier function around the injured tissue. Cav-1 was mainly expressed by CD68-positive macrophages and CD34-positive endothelial cells in the injured tissues around the incision, while it was also primarily localized in the medium and large neurofilament 200-positive neurons and a small number of calcitonin gene-related peptide- and isolectin B4-positive small and medium-sized neurons in the DRG. The results demonstrated that the sustained high expression levels of Cav-1 in the injured tissue around the incision could lead to the dysfunction of the vascular endothelial barrier and, thus, could induce the inflammatory response through the lipoprotein transport of endothelial cells, thereby resulting in peripheral sensitization. In addition, the sustained high expression levels of Cav-1 in the DRG could sensitize large-sized neurons and change the transmission mode of noxious stimuli. The findings of the present study indicated that a Cav-1-mediated process could participate in neuronal transmission pathways associated with pain modulation.

4.
Ann Transl Med ; 9(22): 1656, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34988165

RESUMO

BACKGROUND: Gliosis and inflammation are pivotal in the development of acute and chronic pain. Here, we demonstrated a previously unidentified molecular mechanism in which the activation of exchange proteins directly activated by cyclic adenosine monophosphate (Epac)1 accelerated the activation of astrocytes in the spinal cord, thereby promoting chronic postsurgical pain (CPSP). METHODS: We established a rat model of CPSP induced by skin/muscle incision and retraction (SMIR). Pain behaviors were assessed using mechanical withdrawal threshold (MWT) at different times. The lumbosacral enlargement of the spinal cord was isolated to detect the expression of Epac1 and the activity of astrocytes. They were assessed using western blot and immunofluorescence staining. RESULTS: SMIR induced persistent mechanical hyperalgesia after surgery. This hyperalgesia response was prolonged to more than 21 d after surgery. The time course of spinal Epac1 upregulation was correlated with SMIR-induced pain behaviors. Meanwhile, Epac1 immunoreactivity was colocalized primarily with astrocytes but not with microglial cells or neurons on 7 d after surgery. Intrathecal injection of Epac1 inhibitor CE3F4 significantly suppressed SMIR-induced mechanical allodynia and activation of astrocytes in the spinal cord. This analgesic effect of single-dose administration of CE3F4 lasted up to 6 h and wore off at 12 h after injection. CONCLUSIONS: Spinal Epac1-mediated activation of astrocytes may facilitate CPSP. Inhibition of Epac1 may effectively prevent CPSP.

5.
Mol Med Rep ; 22(6): 4821-4827, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33173987

RESUMO

Chronic postsurgical pain (CPSP) has a high incidence, but the underlying mechanism is not well understood. Accumulating evidence has suggested that central sensitization is the main mechanism of pain. To study the role of p120 in CPSP, a skin/muscle incision and retraction (SMIR) model was established, and immunofluorescence staining and western blotting were performed to analyze the expression of p120 in the spinal cord and dorsal root ganglion (DRG). The results demonstrated that SMIR increased the expression of p120 in the DRG and the spinal cord compared with the naive group. Furthermore, it was demonstrated that p120 was mainly distributed in the glial fibrillary acidic protein­positive astrocytes in the spinal cord, and in the neurofilament 200­positive medium and large neurons in the DRG. Our previous studies have shown that adenosine triphosphate­sensitive potassium channel (KATP) agonists can reduce postoperative pain in rats. Therefore, the changes in p120 were observed in the DRG and spinal cord of rats following the intraperitoneal injection of nicorandil, a KATP agonist. It was demonstrated that nicorandil administration could relieve mechanical pain experienced following SMIR in rats, and decrease the expression of p120 in the DRG and spinal cord. The results revealed that p120 may contribute to the prophylactic analgesic effect of nicorandil, thus providing a novel insight into the mechanism of CPSP prevention.


Assuntos
Cateninas/metabolismo , Dor Crônica/tratamento farmacológico , Nicorandil/farmacologia , Dor Pós-Operatória/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Dor Crônica/metabolismo , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Nicorandil/uso terapêutico , Medição da Dor , Dor Pós-Operatória/metabolismo , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , delta Catenina
6.
J Pain Res ; 12: 2009-2026, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31308730

RESUMO

BACKGROUND: Preoperative anxiety is associated with postoperative hyperalgesia; however, few studies have investigated the mechanism underlying this association in female surgical patients. Research has suggested that ON cells in the rostral ventromedial medulla (RVM) receive nerve impulses via cholecystokinin 2 (CCK2) receptors, facilitating hyperalgesia. Additionally, the downstream serotonergic projection system from the RVM to the spinal cord has a dual regulating effect on pain responses, and the 5-hydoxytryptophan 2B (5-HT2B) receptor in spinal dorsal horn neurons is critically involved in mechanical allodynia. METHODS: Ovariectomized rats were treated with estrogen replacement, single prolonged stress (SPS), and plantar incision. Various receptor agonists and antagonists were then administered into the RVM and spinal cord to study the mechanism underlying postoperative hyperalgesia caused by preoperative anxiety in female rats. RESULTS: Behavioral testing revealed that preoperative SPS induced postoperative hyperalgesia, as well as the expression of the CCK2 receptor in the RVM and the expression of the 5-HT2B receptor, protein kinase Cγ (PKCγ), and phosphorylation of the N-methyl-d-aspartate receptor1 (p-NR1) in the spinal cord increased confirmed by Western blot. RVM microinjection of the CCK2 receptor agonist CCK-8 and intrathecal injection of the 5-HT2B receptor agonist BW723C86 both produced hyperalgesia in female rats after plantar incision, whereas the CCK2 receptor antagonist YM022, the 5-HT2B receptor antagonist RS127445, and the PKCγ inhibitor C37H65N9O13 decreased the rats' sensitivity to the same stimulus. Additionally, electrophysiological analysis suggested that activation of the 5-HT2B receptor increased the whole-cell current (IBa) in superficial dorsal horn neurons through the PKCγ pathway. CONCLUSION: Our study demonstrated that preoperative anxiety-induced postoperative hyperalgesia in female rats is associated with descending pain pathways. The CCK2 receptor in the RVM and spinal 5-HT2B receptor may play a role in this hyperalgesic effect.

7.
Pain Res Manag ; 2019: 9017931, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30863475

RESUMO

Chronic postsurgical pain (CPSP) is a chronic pain state that is difficult to be treated clinically. A series of complicated changes have been produced from nociceptive stimulation to the occurrence and development of postsurgical pain. Many mechanisms remain unclear. In order to study the role of intercellular gap junctions in inducing inflammatory microenvironment at the beginning of nociceptor after operation, the model of skin/muscle incision and retraction (SMIR) was established. We observed the changes of the expression of exchange proteins directly activated by cAMP-1 (Epac1) and p120 catenin (p120), the quantities of macrophages and endothelial cells, vascular endothelial permeability, and mechanical withdrawal threshold (MWT). It was found that macrophages and endothelial cells were functionally coupled through Epac1-p120. Adhesive linkage disorder remodeled the chronic, inflammatory, and eutrophic microenvironment at the beginning of nociceptor after operation through macrophages, endothelial cells, and endothelial paracellular pathways. It might be an early event and a key step in peripheral sensitization of CPSP. The expression of p120 in muscle tissue around the incision might become a prognostic marker for the conversion of acute postsurgical pain into CPSP. Targeted intervention of Epac1-p120 might be a clinical strategy for inhibiting the conversion of acute postsurgical pain into CPSP.


Assuntos
Cateninas/metabolismo , Dor Crônica/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Dor Pós-Operatória/metabolismo , Animais , Masculino , Ratos , Ratos Sprague-Dawley , delta Catenina
8.
Mol Pain ; 13: 1744806917725636, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28825338

RESUMO

Background Accumulating studies have suggested that remifentanil, the widely-used opioid analgesic in clinical anesthesia, can activate the pronociceptive systems and enhance postoperative pain. Glial cells are thought to be implicated in remifentanil-induced hyperalgesia. Electroacupuncture is a complementary therapy to relieve various pain conditions with few side effects, and glial cells may be involved in its antinociceptive effect. In this study, we investigated whether intraoperative electroacupuncture could relieve remifentanil-induced postoperative hyperalgesia by inhibiting the activation of spinal glial cells, the production of spinal proinflammatory cytokines, and the activation of spinal mitogen-activated protein kinases. Methods A rat model of remifentanil-induced postoperative hyperalgesia was used in this study. Electroacupuncture during surgery was conducted at bilateral Zusanli (ST36) acupoints. Behavior tests, including mechanical allodynia and thermal hyperalgesia, were performed at different time points. Astrocytic marker glial fibrillary acidic protein, microglial marker Iba1, proinflammatory cytokines, and phosphorylated mitogen-activated protein kinases in the spinal cord were detected by Western blot and/or immunofluorescence. Results Mechanical allodynia and thermal hyperalgesia were induced by both surgical incision and remifentanil infusion, and remifentanil infusion significantly exaggerated and prolonged incision-induced pronociceptive effects. Glial fibrillary acidic protein, Iba1, proinflammatory cytokines (interleukin-1ß and tumor necrosis factor-α), and phosphorylated mitogen-activated protein kinases (p-p38, p-JNK, and p-ERK1/2) were upregulated after surgical incision, remifentanil infusion, and especially after their combination. Intraoperative electroacupuncture significantly attenuated incision- and/or remifentanil-induced pronociceptive effects, spinal glial activation, proinflammatory cytokine upregulation, and phosphorylated mitogen-activated protein kinase upregulation. Conclusions Our study suggests that remifentanil-induced postoperative hyperalgesia can be relieved by intraoperative electroacupuncture via inhibiting the activation of spinal glial cells, the upregulation of spinal proinflammatory cytokines, and the activation of spinal mitogen-activated protein kinases.


Assuntos
Eletroacupuntura , Hiperalgesia/etiologia , Hiperalgesia/terapia , Neuroglia/patologia , Dor Pós-Operatória/etiologia , Dor Pós-Operatória/terapia , Piperidinas/efeitos adversos , Medula Espinal/patologia , Animais , Biomarcadores/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Mediadores da Inflamação/metabolismo , Cuidados Intraoperatórios , Masculino , Proteínas dos Microfilamentos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuroglia/metabolismo , Fosforilação , Ratos Sprague-Dawley , Remifentanil , Corno Dorsal da Medula Espinal/metabolismo , Corno Dorsal da Medula Espinal/patologia , Regulação para Cima
9.
Mol Neurobiol ; 54(6): 4316-4328, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27339881

RESUMO

Clinically, preoperative anxiety adversely affected postoperative hyperalgesia. As stress-induced glucocorticoids (GCs) were reported to sensitize the activation of microglia, the present study investigated whether and how GCs and microglia played in the process of preoperative anxiety-induced postoperative hyperalgesia. The study used an animal model that exposed rats to single prolonged stress (SPS) procedure to induce preoperative anxiety-like behaviors 24 h before the plantar incisional surgery. Behavioral testing revealed that preoperative SPS enhanced the mechanical allodynia induced by plantar incision. SPS was also found to induce elevated circulating corticosterone levels, potentiate the activation of spinal microglia, and increase the expression of spinal proinflammatory cytokines. Inhibition of microglia by pretreatment with minocycline attenuated the SPS-enhanced mechanical allodynia, and this was accompanied by decreased activation of spinal microglia and expression of proinflammatory cytokines. Another experiment was conducted by administering RU486, the GC receptor (GR) antagonist, to rats. The results showed that RU486 suppressed SPS-induced and SPS-potentiated proinflammatory activation of spinal microglia and revealed analgesic effects. Together, these data indicated that inhibition of stress-induced GR activation attenuated the preoperative anxiety-induced exacerbation of postoperative pain, and the suppression of spinal microglia activation may underlie this anti-hyperalgesia effect. Pending further studies, these findings suggested that GR and spinal microglia may play important roles in the development of preoperative anxiety-induced postoperative hyperalgesia and may serve as novel targets to prevent this phenomenon.


Assuntos
Ansiedade/complicações , Glucocorticoides/farmacologia , Hiperalgesia/etiologia , Microglia/patologia , Animais , Ansiedade/sangue , Ansiedade/patologia , Corticosterona/sangue , Citocinas/metabolismo , Hiperalgesia/sangue , Hiperalgesia/patologia , Mediadores da Inflamação/metabolismo , Masculino , Mifepristona/farmacologia , Minociclina/farmacologia , Cuidados Pós-Operatórios , Cuidados Pré-Operatórios , Ratos Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Medula Espinal/patologia , Estresse Psicológico/sangue , Estresse Psicológico/complicações
10.
Cell Mol Neurobiol ; 37(1): 101-109, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26935064

RESUMO

The objective of this study was to explore the potential role of G-protein-coupled receptor kinase 2 (GRK2) in the progression of cannabinoid 2 receptor (CB2) agonist-induced analgesic effects of bone cancer pain. Female Sprague-Dawley rats, weighing 160-180 g, were utilized to establish a model of bone cancer pain induced by intra-tibia inoculation of Walker 256 mammary gland carcinoma cells. JWH-015, a selective CB2 agonist, was injected intrathecally or intraperitoneally on postoperative day 10. Bone cancer-induced pain behaviors-mechanical allodynia and ambulatory pain-were assessed on postoperative days -1 (baseline), 4, 7, and 10 and at post-treatment hours 2, 6, 24, 48, and 72. The expressions of spinal CB2 and GRK2 protein were detected by Western Blotting on postoperative days -1 (baseline), 4, 7, and 10 and at post-treatment hours 6, 24, and 72. The procedure produced prolonged mechanical allodynia, ambulatory pain, and different changes in spinal CB2 and GRK2 expression levels. Intrathecal or intraperitoneal administration of JWH-015 alleviated the induced mechanical allodynia and ambulatory pain, and inhibited the downregulation of spinal GRK2 expression. These effects were in a time-dependent manner and reversed by pretreatment of CB2 selective antagonist AM630. The results affirmed CB2 receptor agonists might serve as new treatment targets for bone cancer pain. Moreover, spinal GRK2 was an important regulator of CB2 receptor agonist-analgesia pathway.


Assuntos
Neoplasias Ósseas/metabolismo , Dor do Câncer/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/biossíntese , Indóis/administração & dosagem , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/metabolismo , Animais , Neoplasias Ósseas/tratamento farmacológico , Dor do Câncer/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Injeções Intraperitoneais , Injeções Espinhais , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
11.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27852966

RESUMO

The high comorbidity rates of posttraumatic stress disorder and chronic pain have been widely reported, but the underlying mechanisms remain unclear. Emerging evidence suggested that an excess of inflammatory immune activities in the hippocampus involved in the progression of both posttraumatic stress disorder and chronic pain. Considering that microglia are substrates underlying the initiation and propagation of the neuroimmune response, we hypothesized that stress-induced activation of hippocampal microglia may contribute to the pathogenesis of posttraumatic stress disorder-pain comorbidity. We showed that rats exposed to single prolonged stress, an established posttraumatic stress disorder model, exhibited persistent mechanical allodynia and anxiety-like behavior, which were accompanied by increased activation of microglia and secretion of pro-inflammatory cytokines in the hippocampus. Correlation analyses showed that hippocampal activation of microglia was significantly correlated with mechanical allodynia and anxiety-like behavior. Our data also showed that both intraperitoneal and intra-hippocampal injection of minocycline suppressed single prolonged stress-induced microglia activation and inflammatory cytokines accumulation in the hippocampus, and attenuated both single prolonged stress-induced mechanical allodynia and anxiety-like behavior. Taken together, the present study suggests that stress-induced microglia activation in the hippocampus may serve as a critical mechanistic link in the comorbid relationship between posttraumatic stress disorder and chronic pain. The novel concept introduces the possibility of cotreating chronic pain and posttraumatic stress disorder.


Assuntos
Dor Crônica , Hipocampo , Microglia , Transtornos de Estresse Pós-Traumáticos , Estresse Fisiológico/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Dor Crônica/tratamento farmacológico , Citocinas/metabolismo , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Hiperalgesia/tratamento farmacológico , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Minociclina/farmacologia , Ratos Sprague-Dawley , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico
12.
Reg Anesth Pain Med ; 41(6): 744-749, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27429048

RESUMO

BACKGROUND AND OBJECTIVES: Administration of resiniferatoxin (RTX) can mimic the clinical symptoms of postherpetic neuralgia. However, it is unclear whether activated glia contribute to the pathogenesis of RTX-induced neuropathic pain; furthermore, the relationship between p38, N-methyl-D-aspartate receptor type 2B (NR2B) as well as proinflammatory cytokines and activated glia remains unknown. METHODS: Intraperitoneal injection of RTX was performed to induce neuropathic pain in rats. Mechanical allodynia and thermal hyperalgesia were assessed by von Frey filaments and a radiant heat stimulus, respectively. Western blot and immunofluorescence labeling examined the expression of NR2B, activated glia markers, p38, and proinflammatory cytokines in the spinal cord. We further investigated the effect of the glial inhibitors, fluorocitrate and minocycline, on nociceptive behaviors and expression of p38, NR2B, and proinflammatory cytokines. RESULTS: Resiniferatoxin leads to an increase of paw withdrawal latency to a heat stimulus and caused a mechanical allodynia within 2 weeks. The expression of tumor necrosis factor α, IL-1ß, p38, and NR2B was up-regulated in RTX-induced neuropathic pain rat model and lasted for at least 49 days. Microglia were activated at the early phase of the disease, whereas activated astrocytes were detected in the sustainment phase. Both minocycline and fluorocitrate attenuated the nociceptive behaviors and expression of related proteins. CONCLUSIONS: Activated glia participate in the pathogenesis of RTX-induced neuropathic pain and are likely to be the source of proinflammatory cytokines. Inhibition of glia contributes to an analgesic effect. These findings provide a novel strategy for the treatment of postherpetic neuralgia.


Assuntos
Citocinas/metabolismo , Neuralgia/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Modelos Animais de Doenças , Diterpenos , Hiperalgesia , Neuroglia , Ratos , Ratos Sprague-Dawley , Medula Espinal
13.
Am J Transl Res ; 8(2): 1144-54, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27158400

RESUMO

OBJECTIVES: In the present study, we investigate the effects of Mas oncogene-related gene (Mrg) C receptors (MrgC) on the expression and activation of spinal Gi protein, N-methyl-D-aspartate receptor subunit 2B (NR2B), and neuronal nitric oxide synthase (nNOS) in mouse model of bone cancer pain. METHODS: The number of spontaneous foot lift (NSF) and paw withdrawal mechanical threshold (PWMT) were measured after inoculation of tumor cells and intrathecal injection of MrgC agonist bovine adrenal medulla 8-22 (BAM8-22) or MrgC antagonist anti-MrgC for 14 days after operation. Expression of spinal MrgC, Gi protein, NR2B and nNOS and their phosphorylated forms after inoculation was examined by immunohistochemistry and Western blotting. Double labeling was used to identify the co-localization of NR2B or nNOS with MrgC in spinal cord dorsal horn (SCDH) neurons. The effects of intrathecal injection of BAM8-22 or anti-MrgC on nociceptive behaviors and the corresponding expression of spinal MrgC, Gi protein, NR2B and nNOS were also investigated. RESULTS: The expression of spinal MrgC, Gi protein, NR2B, and nNOS was higher in tumor-bearing mice in comparison to sham mice or normal mice. Intrathecal injection of MrgC agonist BAM8-22 significantly alleviated bone cancer pain, up-regulated MrgC and Gi protein expression, and down-regulated the expression of spinal p-NR2B, t-nNOS and p-nNOS in SCDH on day 14 after operation, whereas administration of anti-MrgC produced the opposite effect. Meanwhile, MrgC-like immunoreactivity (IR) co-localizes with NR2B-IR or nNOS-IR in SCDH neurons. CONCLUSIONS: The present study demonstrates that MrgC-activated spinal Gi-NR2B-nNOS signaling pathway plays important roles in the development of bone cancer pain. These findings may provide a novel strategy for the treatment of bone cancer pain.

14.
PLoS One ; 11(5): e0154851, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27152740

RESUMO

OBJECTIVE: This study is to investigate the role of Mas-related gene (Mrg) C in the pathogenesis and treatment of bone cancer pain (BCP). METHODS: BCP mouse model was established by osteosarcoma cell inoculation. Pain-related behaviors were assessed with the spontaneous lifting behavior test and mechanical allodynia test. Expression levels of MrgC, Gi, and NR2B in the spinal cord were detected with Western blot analysis and immunohistochemistry. RESULTS: Pain-related behavior tests showed significantly increased spontaneous flinches (NSF) and decreased paw withdrawal mechanical threshold (PWMT) in mouse models of BCP. Western blot analysis showed that, compared with the control group and before modeling, all the expression levels of MrgC, Gi, and NR2B in the spinal cord of BCP mice were dramatically elevated, which were especially increased at day 7 after operation and thereafter, in a time-dependent manner. Moreover, the treatment of MrgC agonist BAM8-22 significantly up-regulated Gi and down-regulated NR2B expression levels, in the spinal cord of BCP mice, in a time-dependent manner. On the other hand, anti-MrgC significantly down-regulated Gi expression, while dramatically up-regulated NR2B expression, in the BCP mice. Similar results were obtained from the immunohistochemical detection. Importantly, BAM8-22 significantly attenuated the nociceptive behaviors in the BCP mice. CONCLUSION: Our results indicated the MrgC-mediated Gi and NR2B expression alterations in the BCP mice, which might contribute to the pain hypersensitivity. These findings may provide a novel strategy for the treatment of BCP in clinic.


Assuntos
Neoplasias Ósseas/complicações , Dor do Câncer/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Comportamento Animal , Dor do Câncer/etiologia , Dor do Câncer/terapia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Medula Espinal/metabolismo , Regulação para Cima
15.
Inflammation ; 38(5): 1880-90, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25896633

RESUMO

Cannabinoid receptor type 2 (CB2) agonists display potential analgesic effects in acute and neuropathic pain. However, its complex cellular and molecular mechanisms in bone cancer pain remain unclear. And less relevant reports concerned its time-dependent effects on the long-lasting modifications of behavior, spinal inflammatory cytokines levels, astrocytes activity induced by bone cancer pain. A rat model of bone cancer pain induced by intra-tibia inoculation of Walker 256 mammary gland carcinoma cells was utilized. Pain behaviors at different time points were assessed by ambulatory pain scores and paw withdrawal mechanical threshold (PWMT). Pro-inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, IL-18, and tumor necrosis factor alpha (TNF-α), were quantitated by Western blots. Glial activity was assessed by immunohistochemistry. Intra-tibia inoculation of Walker 256 mammary gland carcinoma cells induced progressive bone cancer pain; a long-term up-regulation of IL-1ß, IL-6, IL-18, and TNF-α; and the activation of glia in spinal cord. Activation of microglia was first evident on day 4 after surgery and reached to a peak on day 7 while activation of astrocytes was on day 10. A single intrathecal injection of JWH-015 attenuated bone cancer induced spontaneous pain and mechanical allodynia, reduced the expression of pro-inflammatory cytokines, and inhibited the activity of astrocytes. All the modifications were transient and peaked at 24 h after JWH-015 administration. Furthermore, the protective effects of JWH-015 were reversed in the presence of CB2-selective antagonist AM630. Overall, our results provided evidences for the persistent participation of inflammation reaction in the progression of bone cancer pain, and demonstrated that JWH-015 reduced the expression of IL-1ß, IL-6, IL-18, and TNF-α and inhibited astrocytes activation in a time-dependent manner, thereby displaying an analgesic effect.


Assuntos
Astrócitos/metabolismo , Neoplasias Ósseas/metabolismo , Citocinas/biossíntese , Indóis/administração & dosagem , Mediadores da Inflamação/metabolismo , Dor/metabolismo , Medula Espinal/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Neoplasias Ósseas/complicações , Neoplasias Ósseas/tratamento farmacológico , Citocinas/antagonistas & inibidores , Feminino , Mediadores da Inflamação/antagonistas & inibidores , Injeções Espinhais , Dor/tratamento farmacológico , Dor/etiologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Fatores de Tempo
16.
J Neurol Sci ; 352(1-2): 48-52, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25819118

RESUMO

BACKGROUND: The ultra-short-acting mu-opioid receptor (MOR) agonist remifentanil induces postoperative hyperalgesia both in preclinical and clinical research studies. However, the precise mechanisms remain unclear, although changes in opioid receptor expression might be a correlative feature. Neuron-restrictive silencer factor (NRSF) functions as a crucial regulator of MOR expression in specific neuronal cells. Using a mouse model of incisional postoperative pain, we assessed the expression of MOR and NRSF and investigated whether disruption of NRSF expression could prevent the postoperative nociceptive sensitization induced by surgical incision and subcutaneous infusion of remifentanil. METHODS: Paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL) were independently used to assess mechanical allodynia and thermal hyperalgesia after surgery and cerebral ventricle injection of NRSF antisense oligonucleotide. Western blotting analyses were preformed to assess the expression levels of MOR and NRSF. RESULTS: NRSF expression levels were enhanced after intraoperative infusion of remifentanil, resulting in repression of MOR expression in the periaqueductal gray (PAG). NRSF blockade with an NRSF antisense oligonucleotide significantly enhanced the expression levels of MOR and alleviated mechanical allodynia and thermal hyperalgesia induced by intraoperative infusion of remifentanil. CONCLUSION: NRSF functions as a negative regulator of MOR in PAG and contributes to remifentanil-induced postoperative hyperalgesia. NRSF in PAG may be a potential target for this pain therapy.


Assuntos
Hiperalgesia/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Receptores Opioides mu/metabolismo , Proteínas Repressoras/metabolismo , Animais , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Piperidinas/toxicidade , Remifentanil , Proteínas Repressoras/antagonistas & inibidores , Resultado do Tratamento
17.
Int J Clin Exp Med ; 8(11): 20178-87, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26884930

RESUMO

OBJECTIVES: The aim of this study is to investigate the effects of Mas-related G-protein-coupled receptor C (MrgC) agonist bovine adrenal medulla 8-22 (BAM8-22) on bone cancer pain and mirror-image pain. METHODS: Bone cancer pain was induced by intramedullary injection of NC2472 fibrosarcoma cells in the mice. BAM8-22 and/or anti-MrgC antibody were injected intrathecally at day 14 after bone cancer induction and their effects on pain behaviors were detected. The pain behaviours were assessed by the number of spontaneous foot lifts and paw withdrawal mechanical threshold (PWMT) tests. MrgC expression was detected using western blot analysis and immunofluorescence assay. RESULTS: There were increased bone cancer pain and mirror-image pain in the tumor-bearing mice while not in the sham-treated mice. BAM8-22 attenuated bone cancer pain in mice dose dependently with the highest effects at 2 hr after BAM8-22 administration, and anti-MrgC antibody reversed the effects of BAM8-22. However, intrathecal administration of BAM8-22 did not affect the mirror-image pain. Furthermore, BAM8-22 stimulated the expression of MrgC in the spinal dorsal horn. CONCLUSIONS: MrgC agonist BAM8-22 could attenuate bone cancer pain in mice. This study may provide a novel strategy for the treatment of bone cancer pain.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...