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.
Brain Behav Immun ; 119: 36-50, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555991

ABSTRACT

This study aimed to elucidate the opioid mechanisms underlying dexamethasone-induced pain antihypersensitive effects in neuropathic rats. Dexamethasone (subcutaneous and intrathecal) and membrane-impermeable Dex-BSA (intrathecal) administration dose-dependently inhibited mechanical allodynia and thermal hyperalgesia in neuropathic rats. Dexamethasone and Dex-BSA treatments increased expression of dynorphin A in the spinal cords and primary cultured microglia. Dexamethasone specifically enhanced dynorphin A expression in microglia but not astrocytes or neurons. Intrathecal injection of the microglial metabolic inhibitor minocycline blocked dexamethasone-stimulated spinal dynorphin A expression; intrathecal minocycline, the glucocorticoid receptor antagonist Dex-21-mesylate, dynorphin A antiserum, and κ-opioid receptor antagonist GNTI completely blocked dexamethasone-induced mechanical antiallodynia and thermal antihyperalgesia. Additionally, dexamethasone elevated spinal intracellular cAMP levels, leading to enhanced phosphorylation of PKA, p38 MAPK and CREB. The specific adenylate cyclase inhibitor DDA, PKA inhibitor H89, p38 MAPK inhibitor SB203580 and CREB inhibitor KG-501 completely blocked dexamethasone-induced anti-neuropathic pain and increased microglial dynorphin A exprression. In conclusion, this study reveal that dexamethasone mitigateds neuropathic pain through upregulation of dynorphin A in spinal microglia, likely involving the membrane glucocorticoid receptor/cAMP/PKA/p38 MAPK/CREB signaling pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Dexamethasone , Dynorphins , Microglia , Neuralgia , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord , p38 Mitogen-Activated Protein Kinases , Animals , Microglia/metabolism , Microglia/drug effects , Cyclic AMP/metabolism , Spinal Cord/metabolism , Spinal Cord/drug effects , Male , Neuralgia/metabolism , Neuralgia/drug therapy , Dynorphins/metabolism , Rats , Cyclic AMP-Dependent Protein Kinases/metabolism , Dexamethasone/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Hyperalgesia/metabolism , Hyperalgesia/drug therapy
2.
Bioorg Chem ; 131: 106328, 2023 02.
Article in English | MEDLINE | ID: mdl-36542986

ABSTRACT

Epigenetic regulation and Focal adhesion kinase (FAK) are considered to be two important targets for the development of antitumor drugs. Studies have shown that the combination of FAK and HDAC inhibitors could exhibit synergistic effects in a subset of cancer cells in vitro and in vivo. At present, there are few reports on dual target inhibitors of FAK and HDAC. Here, we first reported a new compound MY-1259 as a dual FAK and HDAC6 inhibitor, which exhibited efficient treatment effects on gastric cancers in vitro and in vivo. MY-1259 exhibited potent inhibitory activities against FAK (IC50 = 132 nM) and HDAC6 (IC50 = 16 nM). Notably, MY-1259 showed selective inhibitory potency on HDAC6 over HDAC1, HDAC2 and HDAC3. In addition, MY-1259 could potently inhibit the proliferative activities of MGC-803 and BGC-823 cells (IC50 = 3.91 and 15.46 nM, respectively, using flow cytometry counting), induce cell apoptosis, and cellular senescence. MY-1259 could effectively down-regulate the levels of Ac-Histone H3 and Ac-α-tubulin, and also inhibit the phosphorylation of FAK at three phosphorylation sites Y397, Y576/577 and Y925, thereby inhibiting the activation of ERK and AKT/mTOR. MY-1259 exhibited more effective antitumor effect in vivo than the HDAC inhibitor SAHA and FAK inhibitor TAE-226 alone or in combination, showing the advantages of FAK/HDAC dual inhibitors in the treatment of gastric cancers. Therefore, the results in this work suggested that inhibition of FAK and HDAC by MY-1259 might represent a promising strategy for the treatment of gastric cancers.


Subject(s)
Antineoplastic Agents , Focal Adhesion Protein-Tyrosine Kinases , Histone Deacetylase Inhibitors , Stomach Neoplasms , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation , Epigenesis, Genetic , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Stomach Neoplasms/drug therapy , Structure-Activity Relationship
3.
Biochem Pharmacol ; 192: 114727, 2021 10.
Article in English | MEDLINE | ID: mdl-34390739

ABSTRACT

Thalidomide is an antiinflammatory, antiangiogenic and immunomodulatory agent which has been used for the treatment of erythema nodosum leprosum and multiple myeloma. It has also been employed in treating complex regional pain syndromes. The current study aimed to reveal the molecular mechanisms underlying thalidomide-induced pain antihypersensitive effects in neuropathic pain. Thalidomide gavage, but not its more potent analogs lenalidomide and pomalidomide, inhibited mechanical allodynia and thermal hyperalgesia in neuropathic pain rats induced by tight ligation of spinal nerves, with ED50 values of 44.9 and 23.5 mg/kg, and Emax values of 74% and 84% MPE respectively. Intrathecal injection of thalidomide also inhibited mechanical allodynia and thermal hyperalgesia in neuropathic pain. Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFα, IL-1ß and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. In contrast, treatment with thalidomide, but not lenalidomide or pomalidomide, stimulated spinal expressions of IL-10 and ß-endorphin in neuropathic rats. Particularly, thalidomide specifically stimulated IL-10 and ß-endorphin expressions in microglia but not astrocytes or neurons. Furthermore, pretreatment with the IL-10 antibody blocked upregulation of ß-endorphin in neuropathic rats and cultured microglial cells, whereas it did not restore thalidomide-induced downregulation of proinflammatory cytokine expression. Importantly, pretreatment with intrathecal injection of the microglial metabolic inhibitor minocycline, IL-10 antibody, ß-endorphin antiserum, and preferred or selective µ-opioid receptor antagonist naloxone or CTAP entirely blocked thalidomide gavage-induced mechanical antiallodynia. Our results demonstrate that thalidomide, but not lenalidomide or pomalidomide, alleviates neuropathic pain, which is mediated by upregulation of spinal microglial IL-10/ß-endorphin expression, rather than downregulation of TNFα expression.


Subject(s)
Interleukin-10/biosynthesis , Microglia/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Thalidomide/therapeutic use , beta-Endorphin/biosynthesis , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interleukin-10/agonists , Male , Microglia/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Thalidomide/pharmacology , beta-Endorphin/agonists
4.
Biochem Pharmacol ; 190: 114600, 2021 08.
Article in English | MEDLINE | ID: mdl-33992630

ABSTRACT

Cynandione A, an acetophenone isolated from Cynanchum Wilfordii Radix, attenuates inflammation. The present study aimed to study the mechanisms underlying cynandione A-induced antiinflammation. Treatment with cynandione A and the specific α7 nicotinic acetylcholine receptor (α7 nAChR) agonist PHA-543613 remarkably reduced overexpression of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1ß in lipopolysaccharide (LPS)-treated RAW264.7 cells and primary peritoneal macrophages, and endotoxemic mice. Both cynandione A and PHA-543613 also stimulated IL-10 expression in naïve and LPS-treated macrophages and endotoxemic mice. Cynandione A- and PHA-543613-inhibited proinflammatory cytokine expression was completely blocked by the α7 nAChR antagonist methyllycaconitine and the IL-10 antibody. The stimulatory effect of cynandione A and PHA-543613 on IL-10 expression were suppressed by methyllycaconitine and knockdown of α7 nAChRs using siRNA/α7 nAChR. Cynandione A significantly stimulated STAT3 phosphorylation, which was attenuated by methyllycaconitine and the IL-10 neutralizing antibody. The STAT3 activation inhibitor NSC74859 also blocked cynandione A-inhibited proinflammatory cytokine expression. Taken together, our results, for the first time, demonstrate that cynandione A and PHA-543613 inhibit inflammation through macrophageal α7 nAChR activation and subsequent IL-10 expression.


Subject(s)
Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Inflammation Mediators/antagonists & inhibitors , Interleukin-10/agonists , Macrophages/drug effects , Quinuclidines/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Biphenyl Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cells, Cultured , Cynanchum , Dose-Response Relationship, Drug , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-10/biosynthesis , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Quinuclidines/therapeutic use , RAW 264.7 Cells , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
Brain Behav Immun ; 95: 344-361, 2021 07.
Article in English | MEDLINE | ID: mdl-33862171

ABSTRACT

Gabapentinoids are recommended first-line treatments for neuropathic pain. They are neuronal voltage-dependent calcium channel α2δ-1 subunit ligands and have been suggested to attenuate neuropathic pain via interaction with neuronal α2δ-1 subunit. However, the current study revealed their microglial mechanisms underlying antineuropathic pain. Intrathecal injection of gabapentin, pregabalin and mirogabalin rapidly inhibited mechanical allodynia and thermal hyperalgesia, with projected ED50 values of 30.3, 6.2 and 1.5 µg (or 176.9, 38.9 and 7.2 nmol) and Emax values of 66%, 61% and 65% MPE respectively for mechanical allodynia. Intrathecal gabapentinoids stimulated spinal mRNA and protein expression of IL-10 and ß-endorphin (but not dynorphin A) in neuropathic rats with the time point parallel to their inhibition of allodynia, which was observed in microglia but not astrocytes or neurons in spinal dorsal horns by using double immunofluorescence staining. Intrathecal gabapentin alleviated pain hypersensitivity in male/female neuropathic but not male sham rats, whereas it increased expression of spinal IL-10 and ß-endorphin in male/female neuropathic and male sham rats. Treatment with gabapentin, pregabalin and mirogabalin specifically upregulated IL-10 and ß-endorphin mRNA and protein expression in primary spinal microglial but not astrocytic or neuronal cells, with EC50 values of 41.3, 11.5 and 2.5 µM and 34.7, 13.3 and 2.8 µM respectively. Pretreatment with intrathecal microglial metabolic inhibitor minocycline, IL-10 antibody, ß-endorphin antiserum or µ-opioid receptor antagonist CTAP (but not κ- or δ-opioid receptor antagonists) suppressed spinal gabapentinoids-inhibited mechanical allodynia. Immunofluorescence staining exhibited specific α2δ-1 expression in neurons but not microglia or astrocytes in the spinal dorsal horns or cultured primary spinal cells. Thus the results illustrate that gabapentinoids alleviate neuropathic pain through stimulating expression of spinal microglial IL-10 and consequent ß-endorphin.


Subject(s)
Gabapentin/pharmacology , Interleukin-10 , Microglia/metabolism , Neuralgia , beta-Endorphin , Animals , Female , Hyperalgesia/drug therapy , Interleukin-10/metabolism , Male , Neuralgia/drug therapy , Rats , Rats, Wistar , Spinal Cord , beta-Endorphin/metabolism
6.
Biochem Biophys Res Commun ; 525(4): 1087-1094, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32184015

ABSTRACT

Lemairamin (also known as wgx-50), is isolated from the pericarps of the Zanthoxylum plants. As an agonist of α7 nicotinic acetylcholine receptors (α7nAChRs), it can reduce neuroinflammation in Alzheimer's disease. This study evaluated its antinociceptive effects in pain hypersensitivity and explored the underlying mechanisms. The data showed that subcutaneous lemairamin injection dose-dependently inhibited formalin-induced tonic pain but not acute nociception in mice and rats, while intrathecal lemairamin injection also dose-dependently produced mechanical antiallodynia in the ipsilateral hindpaws of neuropathic and bone cancer pain rats without affecting mechanical thresholds in the contralateral hindpaws. Multiple bi-daily lemairamin injections for 7 days did not induce mechanical antiallodynic tolerance in neuropathic rats. Moreover, the antinociceptive effects of lemairamin in formalin-induced tonic pain and mechanical antiallodynia in neuropathic pain were suppressed by the α7nAChR antagonist methyllycaconitine. In an α7nAChR antagonist-reversible manner, intrathecal lemairamin also stimulated spinal expression of IL-10 and ß-endorphin, while lemairamin treatment induced IL-10 and ß-endorphin expression in primary spinal microglial cells. In addition, intrathecal injection of a microglial activation inhibitor minocycline, anti-IL-10 antibody, anti-ß-endorphin antiserum or µ-opioid receptor-preferred antagonist naloxone was all able to block lemairamin-induced mechanical antiallodynia in neuropathic pain. These data demonstrated that lemairamin could produce antinociception in pain hypersensitivity through the spinal IL-10/ß-endorphin pathway following α7nAChR activation.


Subject(s)
Acrylamides/pharmacology , Analgesics/pharmacology , Cancer Pain/drug therapy , Hyperalgesia/drug therapy , Microglia/drug effects , Neuralgia/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/agonists , Aconitine/analogs & derivatives , Aconitine/pharmacology , Acrylamides/administration & dosage , Acrylamides/therapeutic use , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Female , Formaldehyde , Hyperalgesia/genetics , Hyperalgesia/metabolism , Injections, Spinal , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice , Microglia/metabolism , Minocycline/administration & dosage , Naloxone/administration & dosage , Rats , Rats, Wistar , Spinal Cord/metabolism , Zanthoxylum/chemistry , Zanthoxylum/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , beta-Endorphin/genetics , beta-Endorphin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...