Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 876: 173062, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32173379

ABSTRACT

Dezocine is an opioid analgesic widely used in China, occupying over 45% of the domestic market of opioid analgesics. We have recently demonstrated that dezocine produced mechanical antiallodynia and thermal antihyperalgesia through spinal µ-opioid receptor activation and norepinephrine reuptake inhibition in neuropathic pain. This study further explored the dual µ-opioid receptor and norepinephrine reuptake mechanisms underlying dezocine-induced mechanical antiallodynia in bone cancer pain, compared with tapentadol, the first recognized analgesic in this class. Dezocine and tapentadol, given subcutaneously, exerted profound mechanical antiallodynia in bone cancer pain rats in a dose-dependent manner, yielding similar maximal effects but different potencies: ED50s of 0.6 mg/kg for dezocine and 7.5 mg/kg for tapentadol, respectively. Furthermore, their mechanical antiallodynia was partially blocked by intrathecal injection of the specific µ-opioid receptor antagonist CTAP, but not κ-opioid receptor antagonists GNTI and nor-BNI or δ-opioid receptor antagonist naltrindole. Intrathecal administrations of the specific norepinephrine depletor 6-OHDA (but not the serotonin depletor PCPA) for three consecutive days and single injection of the α-adrenoceptor antagonist phentolamine/α2-adrenoceptor antagonist yohimbine partially blocked dezocine- and tapentadol-induced mechanical antiallodynia. Strikingly, the combination of CTAP and yohimbine nearly completely blocked dezocine- and tapentadol-induced mechanical antiallodynia. Our results illustrate that both dezocine and tapentadol exert mechanical antiallodynia in bone cancer pain through dual mechanisms of µ-opioid receptor activation and norepinephrine reuptake inhibition, and suggest that the µ-opioid receptor and norepinephrine reuptake dual-targeting opioids are effective analgesics in cancer pain.


Subject(s)
Analgesics, Opioid/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cancer Pain/drug therapy , Hyperalgesia/prevention & control , Receptors, Opioid, mu/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Tapentadol/pharmacology , Tetrahydronaphthalenes/pharmacology , Animals , Behavior, Animal/drug effects , Bone Neoplasms , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Injections, Spinal , Rats , Rats, Sprague-Dawley , Rats, Wistar
2.
J Neuroinflammation ; 16(1): 84, 2019 Apr 13.
Article in English | MEDLINE | ID: mdl-30981281

ABSTRACT

BACKGROUND: The G protein-coupled receptor 40 (GPR40), broadly expressed in various tissues such as the spinal cord, exerts multiple physiological functions including pain regulation. This study aimed to elucidate the mechanisms underlying GPR40 activation-induced antinociception in neuropathic pain, particularly related to the spinal glial expression of IL-10 and subsequent ß-endorphin. METHODS: Spinal nerve ligation-induced neuropathic pain model was used in this study. ß-Endorphin and IL-10 levels were measured in the spinal cord and cultured primary microglia, astrocytes, and neurons. Double immunofluorescence staining of ß-endorphin with glial and neuronal cellular biomarkers was also detected in the spinal cord and cultured primary microglia, astrocytes, and neurons. RESULTS: GPR40 was expressed on microglia, astrocytes, and neurons in the spinal cords and upregulated by spinal nerve ligation. Intrathecal injection of the GPR40 agonist GW9508 dose-dependently attenuated mechanical allodynia and thermal hyperalgesia in neuropathic rats, with Emax values of 80% and 100% MPE and ED50 values of 6.7 and 5.4 µg, respectively. Its mechanical antiallodynia was blocked by the selective GPR40 antagonist GW1100 but not GPR120 antagonist AH7614. Intrathecal GW9508 significantly enhanced IL-10 and ß-endorphin immunostaining in spinal microglia and astrocytes but not in neurons. GW9508 also markedly stimulated gene and protein expression of IL-10 and ß-endorphin in cultured primary spinal microglia and astrocytes but not in neurons, originated from 1-day-old neonatal rats. The IL-10 antibody inhibited GW9508-stimulated gene expression of the ß-endorphin precursor proopiomelanocortin (POMC) but not IL-10, whereas the ß-endorphin antibody did not affect GW9508-stimulated IL-10 or POMC gene expression. GW9508 increased phosphorylation of mitogen-activated protein kinases (MAPKs) including p38, extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK), and its stimulatory effects on IL-10 and POMC expression were blocked by each MAPK isoform inhibitor. Spinal GW9508-induced mechanical antiallodynia was completely blocked by intrathecal minocycline, IL-10 neutralizing antibody, ß-endorphin antiserum, and µ-opioid receptor-preferred antagonist naloxone. CONCLUSIONS: Our results illustrate that GPR40 activation produces antinociception via the spinal glial IL-10/ß-endorphin antinociceptive pathway.


Subject(s)
Hyperalgesia/etiology , Hyperalgesia/metabolism , Interleukin-10/metabolism , Neuralgia , Neuroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , beta-Endorphin/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Interleukin-10/genetics , Male , Methylamines/therapeutic use , Nerve Tissue Proteins/metabolism , Neuralgia/complications , Neuralgia/metabolism , Neuralgia/pathology , Pain Measurement , Propionates/therapeutic use , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects
3.
Biochem Biophys Res Commun ; 505(1): 113-118, 2018 10 20.
Article in English | MEDLINE | ID: mdl-30241937

ABSTRACT

The transcription factor Gli2 plays crucial roles in the transduction of Hedgehog (Hh) signals, yet the mechanisms that control Gli2 degradation remain unclear. Here we have identified the eubiquitinating enzyme otubain2 (OTUB2) as a regulator of Gli2 protein degradation. We found that OTUB2 was coimmunoprecipitated with Gli2. Knockdown of OTUB2 decreased Gli2 protein level while the proteasome inhibitor MG-132 treatment restored Gli2 expression. Additionally, OTUB2 overexpression stabilized Gli2 protein in U2OS cells and extended the half-life of Gli2. We also found that knockdown of OTUB2 reduced deubiquitination of Gli2 in vivo. In vitro deubiquitination assay showed that ubiquitinated Gli2 was decreased by wild-type OTUB2 but not OTUB2 mutations. We also found that OTUB2 knockdown suppressed the ALP activity and the expression of the common markers BMP2 and RUNX2 during osteogenesis of MSCs in response to Shh and Smo agonists, which indicated OTUB2 may have effect on osteogenic differentiation by regulating Hh signaling.


Subject(s)
Deubiquitinating Enzymes/metabolism , Thiolester Hydrolases/metabolism , Ubiquitination , Zinc Finger Protein Gli2/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , Deubiquitinating Enzymes/genetics , HEK293 Cells , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mutation , Osteogenesis/genetics , Protein Binding , Protein Stability , RNA Interference , Thiolester Hydrolases/genetics , Zinc Finger Protein Gli2/genetics
4.
Brain Behav Immun ; 73: 504-519, 2018 10.
Article in English | MEDLINE | ID: mdl-29928964

ABSTRACT

Interleukin 10 (IL-10) is antinociceptive in various animal models of pain without induction of tolerance, and its mechanism of action was generally believed to be mediated by inhibition of neuroinflammation. Here we reported that intrathecal IL-10 injection dose dependently attenuated mechanical allodynia and thermal hyperalgesiain male and female neuropathic rats, with ED50 values of 40.8 ng and 24 ng, and Emax values of 61.5% MPE and 100% MPE in male rats. Treatment with IL-10 specifically increased expression of the ß-endorphin (but not prodynorphin) gene and protein in primary cultures of spinal microglia but not in astrocytes or neurons. Intrathecal injection of IL-10 stimulated ß-endorphin expression from microglia but not neurons or astrocytes in both contralateral and ipsilateral spinal cords of neuropathic rats. However, intrathecal injection of the ß-endorphin neutralizing antibody, opioid receptor antagonist naloxone, or µ-opioid receptor antagonist CTAP completely blocked spinal IL-10-induced mechanical antiallodynia, while the microglial inhibitor minocycline and specific microglia depletor reversed spinal IL-10-induced ß-endorphin overexpression and mechanical antiallodynia. IL-10 treatment increased spinal microglial STAT3 phosphorylation, and the STAT3 inhibitor NSC74859 completely reversed IL-10-increased spinal expression of ß-endorphin and neuroinflammatory cytokines and mechanical antiallodynia. Silence of the Bcl3 and Socs3 genes nearly fully reversed IL-10-induced suppression of neuroinflammatory cytokines (but not expression of ß-endorphin), although it had no effect on mechanical allodynia. In contrast, disruption of the POMC gene completely blocked IL-10-stimulated ß-endorphin expression and mechanical antiallodynia, but had no effect on IL-10 inhibited expression of neuroinflammatory cytokines. Thus this study revealed that IL-10 produced antinociception through spinal microglial ß-endorphin expression, but not inhibition of neuroinflammation.


Subject(s)
Hyperalgesia/drug therapy , Interleukin-10/pharmacology , beta-Endorphin/metabolism , Analgesics/pharmacology , Animals , Astrocytes , Cytokines/metabolism , Female , Hyperalgesia/metabolism , Injections, Spinal , Interleukin-10/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Microglia/physiology , Minocycline/pharmacology , Naloxone/pharmacology , Neuralgia/metabolism , Neurons , Primary Cell Culture , Rats , Rats, Wistar , Spinal Cord/drug effects , Spinal Cord/metabolism , Spine/drug effects , Spine/metabolism , beta-Endorphin/drug effects
5.
J Neuroimmunol ; 316: 17-22, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29249556

ABSTRACT

GLP-1 receptor agonists, exenatide and GLP-1, promoted M2 type polarization in monocytes/macrophages and microglial cells. This study explored the signal basis underlying exenatide-stimulated expression of M2 microglia-specific genes, including the cytoplasmic marker Arg 1, surface marker CD206, and secretion protein marker IL-4. Treatment with exenatide in cultured primary microglial cells concentration dependently stimulated the expression of Arg 1, CD206 and IL-4, but did not significantly alter LPS-stimulated expression of TNF-α, IL-1ß and IL-6. The stimulatory effects of exenatide were completely prevented by the GLP-1 receptor antagonist exendin(9-39), but not altered by application of LPS. Furthermore, the adenylyl cyclase inhibitor DDA, PKA inhibitor H89 and CREB inhibitor KG501 completely blocked exenatide-induced overexpression of Arg 1, CD206 and IL-4. In addition, exenatide-stimulated expression of Arg 1 and CD206 was totally blocked by the p38 MAPK inhibitor SB203580 and gene silencer siRNA/p38ß (but not siRNA/p38α), whereas the expressed IL-4 was not significantly altered by the p38 inhibitor or other MAPK subtype inhibitors. These findings revealed that both classic Gs-cAMP/PKA/CREB and alternative Gs-cAMP/PKA/p38ß/CREB mediated GLP-1 receptor agonism-induced overexpression of M2 microglial biomarkers.


Subject(s)
Cell Differentiation/physiology , Exenatide/pharmacology , Microglia/drug effects , Microglia/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Glucagon-Like Peptide-1 Receptor/agonists , Incretins/pharmacology , Male , Mitogen-Activated Protein Kinase 11/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects
6.
J Neurosci ; 37(48): 11701-11714, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29084866

ABSTRACT

The glucagon-like peptide-1 (GLP-1) receptor agonist exenatide stimulates microglial ß-endorphin expression and subsequently produces neuroprotection and antinociception. This study illustrated an unrecognized autocrine role of IL-10 in mediation of exenatide-induced ß-endorphin expression. Treatment with exenatide in cultured primary spinal microglia concentration dependently stimulated the expression of the M2 microglial markers IL-10, IL-4, Arg 1, and CD206, but not the M1 microglial markers TNF-α, IL-1ß, IL-6, or CD68. Intrathecal exenatide injection also significantly upregulated spinal microglial expression of IL-10, IL-4, Arg 1, and CD206, but not TNF-α, IL-1ß, IL-6, or CD68. Intrathecal injection of exenatide stimulated spinal microglial expression of IL-10 and ß-endorphin in neuropathic rats. Furthermore, treatment with IL-10 (but not IL-4) stimulated ß-endorphin expression in cultured primary microglia, whereas treatment with ß-endorphin failed to increase IL-10 expression. The IL-10-neutralizing antibody entirely blocked exenatide-induced spinal microglial expression of ß-endorphin in vitro and in vivo and fully blocked exenatide mechanical antiallodynia in neuropathic rats. Moreover, specific cAMP/PKA/p38 signal inhibitors and siRNA/p38ß, but not siRNA/p38α, completely blocked exenatide-induced IL-10 expression in cultured primary microglia. Knock-down of IL-10 receptor-α mRNA using siRNA fully inhibited exenatide-induced spinal microglial ß-endorphin expression and mechanical antiallodynia in neuropathy. Exenatide also markedly stimulated phosphorylation of the transcription factor STAT3 in cultured primary microglia and ß-endorphin stimulation was completely inhibited by the specific STAT3 activation inhibitor. These results revealed that IL-10 in microglia mediated ß-endorphin expression after GLP-1 receptor activation through the autocrine cAMP/PKA/p38ß/CREB and subsequent IL-10 receptor/STAT3 signal pathways.SIGNIFICANCE STATEMENT Activation of GLP-1 receptors specifically and simultaneously stimulates the expression of anti-inflammatory cytokines IL-10 and IL-4, as well as the neuroprotective factor ß-endorphin from microglia. GLP-1 receptor agonism induces ß-endorphin expression and antinociception through autocrine release of IL-10. Activation of GLP-1 receptors stimulates IL-10 and ß-endorphin expression subsequently through the Gs-cAMP/PKA/p38ß/CREB and IL-10/IL-10 receptor-α/STAT3 signal transduction pathways.


Subject(s)
Autocrine Communication/physiology , Glucagon-Like Peptide-1 Receptor/biosynthesis , Interleukin-10/biosynthesis , Microglia/metabolism , Spinal Cord/metabolism , beta-Endorphin/biosynthesis , Animals , Animals, Newborn , Autocrine Communication/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Exenatide , Gene Expression , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Interleukin-10/genetics , Interleukin-10/pharmacology , Male , Microglia/drug effects , Peptides/pharmacology , Rats , Rats, Wistar , Spinal Cord/cytology , Spinal Cord/drug effects , Venoms/pharmacology , beta-Endorphin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...