Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biochem Biophys Res Commun ; 522(2): 499-505, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31780264

RESUMO

BACKGROUND: Pulmonary microvascular endothelial cells (PMVECs) are initial targets of sepsis-induced acute lung injury (ALI). During the apoptosis of PMVECs, tight junctions (TJ) and adherens junctions (AJ) are firstly damaged. Previous studies have suggested hydrogen treatment can protect lung microvasculature of mice from sepsis-induced endothelial dysfunction and maintain the coherence of pulmonary endothelium, but the underlying mechanism remains unclear. METHODS: We investigated the role of hydrogen-rich medium on regulating intercellular junction proteins under lipopolysaccharide (LPS) treatment which mimicked sepsis in vitro. Changes of cytoskeleton regulatory protein ROCK and RhoA as well as PMVEC apoptotic rate were examined. RESULTS: LPS treatment reduced the expression levels of occludin and VE-cadherin in PMVECs, while hydrogen-rich medium can recover these changes. Furthermore, H2 can significantly ameliorate the excessive expression of ROCK and RhoA under sepsis-mimicking condition. The application of RhoA activator U-46619 resulted in a more significant elevation in cell apoptotic rate as well as reduction in the expression of junctional proteins. Using H2 can almost completely inhibit the effects of RhoA activator. CONCLUSIONS: Our findings suggest that RhoA is a crucial protein in the signaling pathway of LPS-induced endothelial cell dysfunction. Hydrogen treatment can prevent LPS-induced junctional injury and cell death by inhibiting the activity of RhoA.


Assuntos
Células Endoteliais/patologia , Hidrogênio/farmacologia , Lipopolissacarídeos/efeitos adversos , Pulmão/patologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Antígenos CD , Apoptose/efeitos dos fármacos , Caderinas , Células Endoteliais/efeitos dos fármacos , Camundongos , Modelos Biológicos , Ocludina/metabolismo , Inibidores de Proteínas Quinases/farmacologia
2.
Cell Mol Neurobiol ; 37(4): 643-653, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27380044

RESUMO

Up-regulation of GluN2B-containing N-methyl-D-aspartate receptors (NMDARs) expression and trafficking is the key mechanism for remifentanil-induced hyperalgesia (RIH), nevertheless, the signaling pathway and pivotal proteins involved in RIH remain equivocal. PKMζ, an isoform of protein kinase C (PKC), maintains pain memory storage in neuropathic pain and inflammatory pain, which plays a parallel role regulated by NMDARs in long-term memory trace. In the present study, Zeta Inhibitory Peptide (ZIP), a PKMζ inhibitor, and a selective GluN2B antagonist Ro-256981 are injected intrathecally before remifentanil infusion (1 µg kg-1 min-1 for 1 h, iv) in order to detect whether GluN2B contributes to RIH through affecting synthesis and activity of PKMζ in spinal dorsal horn. Nociceptive tests are measured by Paw withdrawal mechanical threshold (PWT) and paw withdrawal thermal latency (PWL). The L4-L6 segments of dorsal horn taken from rats with RIH are for determining expression of PKMζ and pPKMζ by Western blot and immunohistochemistry. Our data suggest that remifentanil infusion causes an increase of PKMζ in expression and phosphorylation in rats with nociceptive sensitization, beginning at 2 h, peaked at 2 days, and returned to basal level at 7 days. ZIP (10 ng) could block behavioral sensitization induced by remifentanil. Ro25-6981 dosage-dependently attenuated mechanical and thermal hyperalgesia and reversed expression of PKMζ and pPKMζ, indicating that GluN2B-containing NMDA receptor facilitates development of RIH through mediating expression and activity of spinal PKMζ in rats. Although detailed mechanisms require further comprehensive study, the preventive role of Ro25-6981 and ZIP provide novel options for the effective precaution of RIH in clinics.


Assuntos
Hiperalgesia/metabolismo , Neuralgia/tratamento farmacológico , Piperidinas/farmacologia , Proteína Quinase C/metabolismo , Animais , Hiperalgesia/induzido quimicamente , Memória de Longo Prazo/efeitos dos fármacos , Neuralgia/metabolismo , Fosforilação , Células do Corno Posterior/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Remifentanil , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Regulação para Cima/efeitos dos fármacos
3.
Anesthesiology ; 122(4): 908-20, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25501899

RESUMO

BACKGROUND: Hyperalgesia is one of the negative consequences following intraoperative analgesia with remifentanil. Peroxynitrite is a critical determinant in nociceptive process. Peroxynitrite inactivates iron-sulfur cluster that results in mitochondrial dysfunction and the release of iron, leading to mitochondrial iron accumulation. Iron accumulation mediated by divalent metal transporter 1 (DMT1) plays a key role in N-methyl-D-aspartate neurotoxicity. This study aims to determine whether peroxynitrite contributes to remifentanil-induced postoperative hyperalgesia via DMT1-mediated iron accumulation. METHODS: Behavior testing was performed in rat model at different time points. Three-nitrotyrosine, nitrated manganese superoxide dismutase, and DMT1 with/without iron-responsive element [DMT1(+)IRE and DMT1(-)IRE] in spinal cord were detected by Western blot and immunohistochemistry. Spinal iron concentration was measured using the Perl stain and atomic absorption spectrophotometer. Hydrogen-rich saline imparting selectivity for peroxynitrite decomposition and iron chelator was applied in mechanistic study on the roles of peroxynitrite and iron, as well as the prevention of hyperalgesia. RESULTS: Remifentanil induced thermal and mechanical hyperalgesia at postoperative 48 h. Compared with control, there were higher levels of 3-nitrotyrosine (mean ± SD, hyperalgesia vs. control, 1.22 ± 0.18 vs. 0.25 ± 0.05, n = 4), nitrated manganese superoxide dismutase (1.01 ± 0.1 vs. 0.19 ± 0.03, n = 4), DMT1(-)IRE (1.42 ± 0.19 vs. 0.33 ± 0.06, n = 4), and iron concentration (12.87 ± 1.14 vs. 5.26 ± 0.61 µg/g, n = 6) in remifentanil-induced postoperative hyperalgesia, while DMT1(+)IRE was unaffected. Eliminating peroxynitrite with hydrogen-rich saline protected against hyperalgesia and attenuated DMT1(-)IRE overexpression and iron accumulation. Iron chelator prevented hyperalgesia in a dose-dependent manner. CONCLUSIONS: Our study identifies that spinal peroxynitrite activates DMT1(-)IRE, leading to abnormal iron accumulation in remifentanil-induced postoperative hyperalgesia, while providing the rationale for the development of molecular hydrogen and "iron-targeted" therapies.


Assuntos
Proteínas de Transporte de Cátions/biossíntese , Hiperalgesia/metabolismo , Proteínas Reguladoras de Ferro/biossíntese , Ácido Peroxinitroso/biossíntese , Piperidinas/toxicidade , Medula Espinal/metabolismo , Animais , Hiperalgesia/induzido quimicamente , Ferro/metabolismo , Masculino , Dor Pós-Operatória/metabolismo , Ratos , Ratos Sprague-Dawley , Remifentanil
4.
Shock ; 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38010088

RESUMO

BACKGROUND: Sepsis is a type of life-threatening organ dysfunction that is caused by a dysregulated host response to infection. The lung is the most vulnerable target organ under septic conditions. Pulmonary microvascular endothelial cells (PMVECs) play a critical role in acute lung injury (ALI) caused by severe sepsis. The impairment of PMVECs during sepsis is a complex regulatory process involving multiple mechanisms, in which the imbalance of calcium (Ca2+) homeostasis of endothelial cells is a key factor in its functional impairment. Our preliminary results indicated that hydrogen gas (H2) treatment significantly alleviates lung injury in sepsis, protects PMVECs from hyperpermeability, and decreases the expression of plasma membrane stromal interaction molecule 1 (STIM1), but the underlying mechanism by which H2 maintains Ca2+ homeostasis in endothelial cells in septic models remains unclear. Thus, the purpose of the present study was to investigate the molecular mechanism of STIM1 and Ca2+-release-activated- Ca2+ channel protein1 (Orai1) regulation by H2 treatment and explore the effect of H2 treatment on Ca2+ homeostasis in lipopolysaccharide (LPS)-induced PMVECs and LPS-challenged mice. METHODS: We observed the role of H2 on LPS-induced ALI of mice in vivo. The lung wet/dry (W/D) weight ratio, total protein in the bronchoalveolar lavage (BAL) fluid and Evans blue dye (EBD) assay were used to evaluate the pulmonary endothelial barrier damage of LPS-challenged mice. The expression of STIM1 and Orai1 were also detected using epifluorescence microscopy. Moreover, we also investigated the role of H2- rich medium in regulating PMVECs under LPS treatment, which induced injury similar to sepsis in vitro. The expression of STIM1 and Orai1 as well as the Ca2+ concentration in PMVECs were examined. RESULTS: In vivo, we found that H2 alleviated ALI of mice through decreasing lung W/D weight ratio, total protein in the BAL fluid and permeability of lung. In addition, H2 also decreased the expression of STIM1 and Orai1 in pulmonary microvascular endothelium. In vitro, LPS treatment increased the expression levels of STIM1 and Orai1 in PMVECs, while H2 reversed these changes. Furthermore, H2 ameliorated Ca2+ influx under sepsis-mimicking conditions. Treatment with the sarco/endoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA) inhibitor, thapsigargin (TG), resulted in a significant reduction in cell viability as well as a reduction in the expression of junctional proteins, including VE-cadherin and occludin. Treatment with the store-operated Ca2+ entry (SOCE) inhibitor, YM-58483 (BTP2), increased the cell viability and expression of junctional proteins. CONCLUSIONS: The present study suggested that H2 treatment alleviates LPS-induced PMVEC dysfunction by inhibiting SOCE mediated by STIM1 and Orai1 in vitro and in vivo.

5.
Ann Transl Med ; 10(2): 56, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35282080

RESUMO

Background: The nuclear receptor REV-ERBα (nuclear receptor subfamily 1, Group D member 1, NR1D1) is one of the essential components of the circadian clock which modulates cell proliferation, glucose metabolism, inflammation, and many other biological processes. Modulation of these processes are also relevant to cancer development. Previous studies have suggested that activation of REV-ERBα correlates with cancer cell senescence and death, but how REV-ERBα play roles in tumor progression require further elucidation. Methods: We investigated the expression of REV-ERBα in clinical samples by immunohistochemistry (IHC). REV-ERBα is downregulated by shorth hairpin RNA (shRNA). The gene expression level of each group was detected by Western blot analysis. The effects of REV-ERBα downregulation on apoptosis and cell cycles was assessed by flow cytometry assay. A549 cell growth curve under different treatments measured by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. Cell invasion ability under different treatments was measured by Transwell assay. Immunostaining analysis was also used for evaluating the effects of downregulation of REV-ERBα on nuclear factor-κB (NF-κB). Results: Compared to 81.8% (54/66) of samples exhibiting a lower expression level of REV-ERBα in cancer tissue than in paired normal tissue, only 18.2% (12/66) were higher or equally expressed in lung cancer tissue. Furthermore, downregulation of REV-ERBα by RNA interference can significantly enhance the transcription of nuclear factor-κB (NF-κB), while the expression of p53 remained the same. Downregulation of REV-ERBα was also shown to stimulate the invasion and promote the proliferation of lung adenocarcinoma cell line A549. Conclusions: Our findings suggest that tumorigenesis and progression of lung carcinoma is relevant to downregulation or inhibition of REV-ERBα. This pathophysiological process also correlates with regulation of the NF-κB signaling pathway, indicating that REV-ERBα is a potential target of lung cancer therapy.

6.
J Pain ; 22(1): 32-47, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32574785

RESUMO

N-methyl-D-aspartate (NMDA) receptor activation is known to be critical in remifentanil-induced hyperalgesia. Evidence indicates that iron accumulation participates in NMDA neurotoxicity. This study aims to investigate the role of iron accumulation in remifentanil-induced hyperalgesia. Remifentanil was delivered intravenously in rats to induce hyperalgesia. The NMDA receptor antagonist MK-801 was intrathecally administrated. The levels of divalent metal transporter 1 without iron-responsive element [DMT1(-)IRE] and iron were detected. Behavior testing was performed in DMT1(-)IRE knockdown rats and rats treated with iron chelator DFO. Meanwhile, the spinal dorsal horn neurons were cultured and transfected with DMT1(-)IRE siRNA, and then respectively incubated with remifentanil and MK-801. The levels of intracellular Ca2+ and iron were assessed by fluorescence imaging. Our data revealed that spinal DMT1(-)IRE and iron content significantly increased in remifentanil-treated rats, and MK-801 inhibited the enhancements. DMT1(-)IRE knockdown and DFO prevented against remifentanil-induced hyperalgesia. Notably, the levels of Ca2+ and iron increased in remifentanil-incubated neurons, and these growths can be blocked by MK-801. DMT1(-)IRE knockdown attenuated iron accumulation but did not influence Ca2+ influx. This study suggests that DMT1(-)IRE-mediated iron accumulation is likely to be the downstream event following NMDA receptor activation and Ca2+ influx, contributing to remifentanil-induced hyperalgesia. PERSPECTIVE: Remifentanil-induced hyperalgesia is common even when used within clinical accepted doses. This study presents that aberrant iron accumulation is involved in the development of remifentanil-induced hyperalgesia in vivo and in vitro. Iron chelation may be a potential therapeutic strategy for the prevention of hyperalgesia in populations at high risk.


Assuntos
Analgésicos Opioides/farmacologia , Proteínas de Transporte de Cátions/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/prevenção & controle , Quelantes de Ferro/farmacologia , Ferro/metabolismo , Células do Corno Posterior/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Remifentanil/farmacologia , Coluna Vertebral/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Células Cultivadas , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Masculino , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores Opioides mu/agonistas , Remifentanil/administração & dosagem , Transdução de Sinais/fisiologia
7.
Clin J Pain ; 33(5): 435-442, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27518488

RESUMO

BACKGROUND: Acute remifentanil exposure during intraoperative analgesia might enhance sensitivity to noxious stimuli and nociceptive responses to innocuous irritation. Cyclooxygenase inhibition was demonstrated to attenuate experimental remifentanil-induced hyperalgesia (RIH) in rodents and human volunteers. The study aimed to compare the effects of preoperative and postoperative flurbiprofen axetil (FA) on RIH after surgery. MATERIALS AND METHODS: Ninety patients undergoing elective laparoscopic gynecologic surgery were randomly assigned to receive either intravenous placebo before anesthesia induction (Group C); or intravenous FA (1.0 mg/kg) before anesthesia induction (Group F1) or before skin closure (Group F2). Anesthesia consisted off sevoflurane and remifentanil (0.30 µg/kg/min). Postoperative pain was managed by sufentanil titration in the postanesthetic care unit, followed by sufentanil infusion via patient-controlled analgesia. Mechanical pain threshold (primary outcome), pain scores, sufentanil consumption, and side-effects were documented for 24 hours postoperatively. RESULTS: Postoperative pain score in Group F1 was lower than Group C. Time of first postoperative sufentanil titration was prolonged in Group F1 than Group C (P=0.021). Cumulative sufentanil consumption in Group F1 was lower than Group C (P<0.001), with a mean difference of 8.75 (95% confidence interval, 5.21-12.29) µg. Mechanical pain threshold on the dominant inner forearm was more elevated in Group F1 than Group C (P=0.005), with a mean difference of 17.7 (95% confidence interval, 5.4-30.0) g. Normalized hyperalgesia area was decreased in Group F1 compared to Group C (P=0.007). No statistically significant difference was observed between Group F2 and Group C. CONCLUSIONS: Preoperative FA reduces postoperative RIH in patients undergoing laparoscopic gynecologic surgery under sevoflurane-remifentanil anesthesia.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Flurbiprofeno/análogos & derivados , Procedimentos Cirúrgicos em Ginecologia , Hiperalgesia/tratamento farmacológico , Laparoscopia , Piperidinas/efeitos adversos , Adulto , Analgésicos Opioides/administração & dosagem , Anestésicos Intravenosos/efeitos adversos , Anestésicos Intravenosos/uso terapêutico , Anti-Inflamatórios não Esteroides/efeitos adversos , Feminino , Flurbiprofeno/administração & dosagem , Flurbiprofeno/efeitos adversos , Antebraço/fisiopatologia , Humanos , Hiperalgesia/induzido quimicamente , Éteres Metílicos/efeitos adversos , Éteres Metílicos/uso terapêutico , Pessoa de Meia-Idade , Limiar da Dor/efeitos dos fármacos , Dor Pós-Operatória/tratamento farmacológico , Piperidinas/uso terapêutico , Cuidados Pós-Operatórios , Cuidados Pré-Operatórios , Remifentanil , Sevoflurano , Sufentanil/uso terapêutico , Tato , Resultado do Tratamento
8.
Brain Res Bull ; 135: 8-24, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28867384

RESUMO

BACKGROUND: Cold hyperalgesia is an intractable sensory abnormality commonly seen in peripheral neuropathies. Although glial cell line-derived neurotrophic factor family receptor alpha3 (GFRα3) is required for the formation of pathological cold pain has been revealed, potential transduction mechanism is poorly elucidated. We have previously demonstrated the contribution of enhanced activity of transient receptor potential melastatin 8 (TRPM8) to cold hyperalgesia in neuropathic pain using a rat model of chronic constriction injury (CCI) to the sciatic nerve. Recently, the enhancement of TRPM8 activity is attributed to the increased TRPM8 plasma membrane trafficking. In addition, TRPM8 can be sensitized by the activation of GFRα3, leading to increased cold responses in vivo. The aim of this study was to investigate whether GFRα3 could influence cold hyperalgesia of CCI rats via modulating TRPM8 expression and plasma membrane trafficking in dorsal root ganglion (DRG). METHODS: Mechanical allodynia, cold and heat hyperalgesia were measured on 1day before CCI and the 1st, 4th, 7th, 10th and 14th day after CCI. TRPM8 total expression and membrane trafficking as well as GFRα3 expression in DRG were detected by immunofluorescence and western blot. Furthermore, GFRα3 small interfering RNA (siRNA) was intrathecally administrated to reduce GFRα3 expression in DRG, and the effects of GFRα3 knockdown on CCI-induced behavioral sensitization as well as TRPM8 total expression and membrane trafficking in both mRNA and protein levels were investigated, and the change in coexpression of TRPM8 with GFRα3 was also evaluated. Then, the effect of GFRα3 activation with artemin on pain behavior of CCI rats pretreated with the selective TRPM8 antagonist RQ-00203078 was observed. RESULTS: Here we found that TRPM8 total expression and plasma membrane trafficking as well as GFRα3 expression in DRG were initially increased on the 4th day after CCI, and maintained at the peak level from the 10th to the 14th day, which entirely conformed with the induction and maintenance of behavioral-reflex facilitation following CCI. The coexpression of TRPM8 with GFRα3, which was mainly located in peptidergic C-fibers DRG neurons, was also increased after CCI. Downregulation of GFRα3 protein in DRG attenuated CCI-induced cold hyperalgesia without affecting mechanical allodynia and heat hyperalgesia, and reduced the upregulations of TRPM8 total expression and plasma membrane trafficking as well as coexpression of TRPM8 with GFRα3 induced by CCI. Additionally, the inhibition of TRPM8 abolished the influence of GFRα3 activation on cold hyperalgesia after CCI. CONCLUSION: Our results demonstrate that GFRα3 knockdown specially inhibits cold hyperalgesia following CCI via decreasing the expression level and plasma membrane trafficking of TRPM8 in DRG. GFRα3 and its downstream mediator, TRPM8, represent a new analgesia axis which can be further exploited in sensitized cold reflex under the condition of chronic pain.


Assuntos
Gânglios Espinais/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Hiperalgesia/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Dor Crônica/metabolismo , Lesão por Frio/genética , Lesão por Frio/metabolismo , Constrição , Gânglios Espinais/fisiologia , Regulação da Expressão Gênica , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/deficiência , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Hiperalgesia/genética , Masculino , Neuralgia/metabolismo , Neurônios/metabolismo , Manejo da Dor , Transporte Proteico/genética , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo , Canais de Cátion TRPM/biossíntese , Canais de Cátion TRPM/genética
9.
Clin J Pain ; 32(8): 702-10, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26550961

RESUMO

BACKGROUND: Several mechanisms of remifentanil-induced hyperalgesia in spinal cord mainly have been explained such as N-methyl-D-aspartate receptors activation, but the mechanism in dorsal root ganglion (DRG) is poorly understood. It has been reported that CCL3 may be a regulator in both inflammatory pain and hyperalgesia. In this paper we explored whether CCL3 and CCR5, the mainly receptor of CCL3, play a role in the remifentanil-induced hyperalgesia in DRG by using a rat model with remifentanil administration. MATERIALS AND METHODS: The von Frey test and hot plate test were performed to measure the different threshold to evaluate mechanical and thermal hyperalgesia. Real-time polymerase chain reaction and Western blot analysis were used to evaluate time course of CCL3 and CCR5 expression in DRG after remifentanil infusion. The changes of glial cells and the expression of CCL3 and CCR5 were detected by immunofluorescence assay. Finally, intrathecal injection of CCL3-neutralizing antibody and maraviroc, the CCR5 antagonists, were used sevoflurane to confirm the role of CCL3/CCR5 signaling in the rat model of remifentanil-induced hyperalgesia. RESULTS: Remifentanil infusion profoundly increased thermal and mechanical hyperalgesia from 2 to 48 hours according to paw withdrawal latency (PWL) and paw withdrawal thresholds (PWT) (mean±SD, hyperalgesia vs. control, 17.4±0.91 vs. 8.5±1.46 s; 20.1±0.32 vs. 9.6±0.5 g, n=8). Moreover, the expression of mRNAs and proteins of CCL3 and its receptor CCR5 in DRG were dramatically increased after remifentanil infusion as compared with the normal saline group. We also found that CCL3 and CCR5 were colocalized with glial cells or neurons. Furthermore, intrathecal injection of CCL3-neutralizing antibody (mean±SD, hyperalgesia vs. control, 17.4±0.91 vs. 8.5±1.46 s; 20.1±0.32 vs. 9.6±0.5 g, n=5) and maraviroc(mean±SD, hyperalgesia vs. control, 17.4±0.91 vs. 8.5±1.46 s; 20.1±0.32 vs. 9.6±0.5 g, n=5) were able to suppress remifentanil-related thermal and mechanical hyperalgesia according to behavioral test. CONCLUSIONS: The results highlighted the fact that CCL3 and its receptor CCR5 in DRG might contribute to remifentanil-induced hyperalgesia. Thus CCL3/CCR5 signaling may be further considered in the development of new therapeutic strategies.


Assuntos
Analgésicos Opioides/toxicidade , Quimiocina CCL3/metabolismo , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Piperidinas/toxicidade , Receptores CCR5/metabolismo , Animais , Antagonistas dos Receptores CCR5 , Cicloexanos/farmacologia , Modelos Animais de Doenças , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Temperatura Alta , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Maraviroc , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Remifentanil , Tato , Triazóis/farmacologia
10.
Brain Res Bull ; 110: 30-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25498394

RESUMO

BACKGROUND: Several studies have demonstrated that intraoperative remifentanil infusions have been associated with opioid-induced hyperalgesia (OIH). Activation of delta opioid receptor (DOR) and augmentation of N-methyl-d-aspartate (NMDA) receptor expression and function may play an important role in the development of OIH. The aim of this study was to investigate whether DOR inhibition could prevent remifentanil-induced hyperalgesia via regulating spinal NMDA receptor expression and function in vivo and in vitro. METHODS: A rat model of remifentanil-induced postoperative hyperalgesia was performed with the DOR agonist deltorphin-deltorphin II or the DOR antagonist naltrindole injected intrathecally 10 min before remifentanil infusion. Mechanical and thermal hyperalgesia were measured at -24h, 2, 6, 24 and 48 h after remifentanil infusion. Western blot was applied to detect the membrane and total expression of DOR and NMDA receptor subunits (NR1, NR2A and NR2B) in spinal cord L4-L6 segments. In addition, whole-cell patch-clamp recording was used to investigate the effect of DOR inhibition on NMDA receptor-induced current in spinal cord slices in vitro. RESULTS: We found that membrane trafficking of DOR, NR1 and NR2B subunits in the spinal cord increased after remifentanil administration and surgery. The DOR antagonist naltrindole could attenuate mechanical and thermal hyperalgesia without affecting baseline nociceptive threshold, reduce membrane expression of DOR and decrease the membrane and total expressions of NR1 and NR2B subunits. Furthermore, the amplitude and the frequency of NMDA receptor-induced current were significantly increased by remifentanil incubation in neurons of the dorsal horn, which was reversed by the application of naltrindole. CONCLUSION: The above results indicate that inhibition of DOR could significantly inhibit remifentanil-induced hyperalgesia via modulating the total protein level, membrane trafficking and function of NMDA receptors in the dorsal horn of spinal cord, suggesting that naltrindole could be a potential anti-hyperalgesic agent for treating OIH.


Assuntos
Anestésicos Intravenosos/toxicidade , Hiperalgesia/prevenção & controle , Dor Pós-Operatória/prevenção & controle , Piperidinas/toxicidade , Receptores Opioides delta/antagonistas & inibidores , Medula Espinal/efeitos dos fármacos , Anestésicos Intravenosos/uso terapêutico , Animais , Modelos Animais de Doenças , Temperatura Alta , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Vértebras Lombares , Masculino , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Dor Pós-Operatória/induzido quimicamente , Piperidinas/uso terapêutico , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Remifentanil , Medula Espinal/metabolismo , Técnicas de Cultura de Tecidos , Tato
11.
Brain Res Bull ; 106: 47-55, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24951883

RESUMO

BACKGROUND: Although NMDAR trafficking mediated by GSK-3ß involvement in transmission of pronociceptive messages in the spinal cord has been confirmed by our previous studies, whether NMDAR trafficking is implicated in peripheral sensitization remains equivocal. It is demonstrated that inflammation is associated with spinal NMDAR-containing nociceptive neurons activation and the maintenance of opioid induced pain hypersensitivity. However, whether and how hydrogen-rich saline, as an effective anti-inflammatory drug, could prevent hyperalgesia through affecting peripheral sensitization caused by NMDAR activation remains to be explored. METHODS: To test these effects, hydrogen-rich saline (2.5, 5 or 10 ml/kg) was administrated intraperitoneally after remifentanil infusion, NMDAR antagonist MK-801 or GSK-3ß inhibitor TDZD-8 was administrated intravenously before remifentanil infusion in rats. We examined time course of hydrogen concentration in blood after hydrogen-rich saline administration. Mechanical and thermal hyperalgesia were evaluated by measuring PWT and PWL for 48 post-infusion hours, respectively. Western blotting and real-time qPCR assay were applied to analyze the NR1 membrane trafficking, GSK-3ß expression and activity in DRG. Inflammatory mediators (TNF-α, IL-1ß, and IL-6) expressions in DRG were also analyzed. RESULTS: We found that NR1 membrane trafficking in DRG increased, possibly due to GSK-3ß activation after remifentanil infusion. We also discovered that hydrogen-rich saline not 2.5 ml/kg but 5 and 10 ml/kg could dose-dependently attenuate mechanical and thermal hyperalgesia without affecting baseline nociceptive threshold, reduce expressions of inflammatory mediators (TNF-α, IL-1ß, and IL-6) and decrease NR1 trafficking mediated by GSK-3ß, and minimal effective concentration was observed to be higher than 10 µmol/L, namely peak concentration in arterial blood after administration of HRS 2.5 ml/kg without any influence on hyperalgesia. CONCLUSION: Our results indicated that antihyperalgesic effect of hydrogen-rich saline might depend predominantly on its ability to reverse NR1 trafficking via inhibition of GSK-3ß activity in DRG in a dose-dependent manner.


Assuntos
Analgésicos Opioides/toxicidade , Gânglios Espinais/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Hidrogênio/uso terapêutico , Hiperalgesia/tratamento farmacológico , Piperidinas/toxicidade , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Membrana Celular/metabolismo , Gânglios Espinais/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta , Hidrogênio/sangue , Hidrogênio/farmacologia , Concentração de Íons de Hidrogênio , Hiperalgesia/induzido quimicamente , Masculino , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Ratos , Ratos Sprague-Dawley , Remifentanil , Cloreto de Sódio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA