Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
CNS Neurosci Ther ; 28(8): 1259-1267, 2022 08.
Article in English | MEDLINE | ID: mdl-35633059

ABSTRACT

AIMS: Potassium (K+ ) channels have been demonstrated to play a prominent involvement in nociceptive processing. Kir7.1, the newest members of the Kir channel family, has not been extensively studied in the CNS, and its function remains largely unknown. The present study investigated the role of spinal Kir7.1 in the development of pathological pain. METHODS AND RESULTS: Neuropathic pain was induced by spared nerve injury (SNI). The mechanical sensitivity was assessed by von Frey test. Immunofluorescence staining assay revealed that Kir7.1 was predominantly expressed in spinal neurons but not astrocytes or microglia in normal rats. Western blot results showed that SNI markedly decreased the total and membrane expression of Kir7.1 in the spinal dorsal horn accompanied by mechanical hypersensitivity. Blocking Kir7.1 with the specific antagonist ML418 or knockdown kir7.1 by siRNA led to mechanical allodynia. Co-IP results showed that the spinal kir7.1 channels were decorated by SUMO-1 but not SUMO-2/3, and Kir7.1 SUMOylation was upregulated following SNI. Moreover, inhibited SUMOylation by GA (E1 inhibitor) or 2-D08 (UBC9 inhibitor) can increase the spinal surface Kir7.1 expression. CONCLUSION: SUMOylation of the Kir7.1 in the spinal cord might contribute to the development of SNI-induced mechanical allodynia by decreasing the Kir7.1 surface expression in rats.


Subject(s)
Hyperalgesia , Neuralgia , Animals , Hyperalgesia/metabolism , Neuralgia/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord Dorsal Horn/metabolism , Sumoylation
2.
J Neuroinflammation ; 17(1): 310, 2020 Oct 18.
Article in English | MEDLINE | ID: mdl-33070779

ABSTRACT

BACKGROUND: The major dose-limiting toxicity of paclitaxel, one of the most commonly used drugs to treat solid tumor, is painful neuropathy. However, the molecular mechanisms underlying paclitaxel-induced painful neuropathy are largely unclarified. METHODS: Paw withdrawal threshold was measured in the rats following intraperitoneal injection of paclitaxel. The qPCR, western blotting, protein or chromatin immunoprecipitation, ChIP-seq identification of NFATc2 binding sites, and microarray analysis were performed to explore the molecular mechanism. RESULTS: We found that paclitaxel treatment increased the nuclear expression of NFATc2 in the spinal dorsal horn, and knockdown of NFATc2 with NFATc2 siRNA significantly attenuated the mechanical allodynia induced by paclitaxel. Further binding site analysis utilizing ChIP-seq assay combining with gene expression profile revealed a shift of NFATc2 binding site closer to TTS of target genes in dorsal horn after paclitaxel treatment. We further found that NFATc2 occupancy may directly upregulate the chemokine CXCL14 expression in dorsal horn, which was mediated by enhanced interaction between NFATc2 and p300 and consequently increased acetylation of histone H4 in CXCL14 promoter region. Also, knockdown of CXCL14 in dorsal horn significantly attenuated mechanical allodynia induced by paclitaxel. CONCLUSION: These results suggested that enhanced interaction between p300 and NFATc2 mediated the epigenetic upregulation of CXCL14 in the spinal dorsal horn, which contributed to the chemotherapeutic paclitaxel-induced chronic pain.


Subject(s)
Chemokines, CXC/biosynthesis , Epigenesis, Genetic/drug effects , NFATC Transcription Factors/biosynthesis , Neuralgia/chemically induced , Neuralgia/metabolism , Paclitaxel/toxicity , Animals , Antineoplastic Agents, Phytogenic/toxicity , Base Sequence , Chemokines, CXC/genetics , Epigenesis, Genetic/physiology , Male , NFATC Transcription Factors/genetics , Neuralgia/genetics , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Up-Regulation/physiology
3.
Nat Commun ; 10(1): 4119, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511520

ABSTRACT

Circular RNAs are non-coding RNAs, and are enriched in the CNS. Dorsal horn neurons of the spinal cord contribute to pain-like hypersensitivity after nerve injury in rodents. Here we show that spinal nerve ligation is associated with an increase in expression of circAnks1a in dorsal horn neurons, in both the cytoplasm and the nucleus. Downregulation of circAnks1a by siRNA attenuates pain-like behaviour induced by nerve injury. In the cytoplasm, we show that circAnks1a promotes the interaction between transcription factor YBX1 and transportin-1, thus facilitating the nucleus translocation of YBX1. In the nucleus, circAnks1a binds directly to the Vegfb promoter, increases YBX1 recruitment to the Vegfb promoter, thereby facilitating transcription. Furthermore, cytoplasmic circAnks1a acts as a miRNA sponge in miR-324-3p-mediated posttranscriptional regulation of VEGFB expression. The upregulation of VEGFB contributes to increased excitability of dorsal horn neurons and pain behaviour induced by nerve injury. We propose that circAnks1a and VEGFB are regulators of neuropathic pain.


Subject(s)
Hypersensitivity/metabolism , Neuralgia/genetics , Neuralgia/metabolism , RNA, Circular/genetics , Spinal Cord/metabolism , Animals , Base Sequence , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Neurons/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , Rats, Sprague-Dawley , Rodentia , Spinal Cord Dorsal Horn/metabolism , Up-Regulation/genetics , Vascular Endothelial Growth Factor B/metabolism
4.
CNS Neurosci Ther ; 20(8): 728-36, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24629168

ABSTRACT

AIMS: Connexin 43 (Cx43) has been reported to be involved in neuropathic pain, but whether it contributes to morphine antinociceptive tolerance remains unknown. The present study investigated the role of spinal Cx43 in the development of morphine tolerance and its mechanisms in rats. METHODS: Morphine tolerance was induced by intrathecal (i.t.) administration of morphine (15 µg) daily for seven consecutive days. The analgesia effect was assessed by hot-water tail-flick test. Expression of proteins was detected by Western blot and immunohistochemistry assay. RESULTS: Chronic morphine markedly increased the expression of spinal Cx43. Gap26, a specific Cx43 mimic peptide, attenuated not only morphine antinociceptive tolerance, but also the up-regulation of spinal Cx43 expression, the activation of astrocytes, and N-methyl-D-aspartic acid (NMDA) receptors (NR1 and NR2B subunits), as well as the decreased GLT-1 expression induced by chronic morphine. MK-801, a noncompetitive NMDA receptors antagonist, suppressed the chronic morphine-induced spinal Cx43 up-regulation, astrocytes activation and decline of GLT-1 expression. CONCLUSIONS: The spinal astrocytic Cx43 contributes to the development of morphine antinociceptive tolerance by activating astrocytes and NMDA receptors, and inhibiting GLT-1 expression. We also demonstrate that the role of interaction between the spinal astrocytic Cx43 and neuronal NMDA receptors is important in morphine tolerant rats.


Subject(s)
Analgesics, Opioid/pharmacology , Astrocytes/drug effects , Connexin 43/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Morphine/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/cytology , Animals , Astrocytes/metabolism , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Peptides/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects
5.
Neuropsychopharmacology ; 38(6): 1015-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23303066

ABSTRACT

Long-term abolition of a brain arousal system impairs wakefulness (W), but little is known about the consequences of long-term enhancement. The brain histaminergic arousal system is under the negative control of H3-autoreceptors whose deletion results in permanent enhancement of histamine (HA) turnover. In order to determine the consequences of enhancement of the histaminergic system, we compared the cortical EEG and sleep-wake states of H3-receptor knockout (H3R-/-) and wild-type mouse littermates. We found that H3R-/-mice had rich phenotypes. On the one hand, they showed clear signs of enhanced HA neurotransmission and vigilance, i.e., a higher EEG θ power during spontaneous W and a greater extent of W or sleep restriction during behavioral tasks, including environmental change, locomotion, and motivation tests. On the other hand, during the baseline dark period, they displayed deficient W and signs of sleep deterioration, such as pronounced sleep fragmentation and reduced cortical slow activity during slow wave sleep (SWS), most likely due to a desensitization of postsynaptic histaminergic receptors as a result of constant HA release. Ciproxifan (H3-receptor inverse agonist) enhanced W in wild-type mice, but not in H3R-/-mice, indicating a functional deletion of H3-receptors, whereas triprolidine (postsynaptic H1-receptor antagonist) or α-fluoromethylhistidine (HA-synthesis inhibitor) caused a greater SWS increase in H3R-/- than in wild-type mice, consistent with enhanced HA neurotransmission. These sleep-wake characteristics and the obesity phenotypes previously reported in this animal model suggest that chronic enhancement of histaminergic neurotransmission eventually compromises the arousal system, leading to sleep-wake, behavioral, and metabolic disorders similar to those caused by voluntary sleep restriction in humans.


Subject(s)
Histamine/metabolism , Receptors, Histamine H3/deficiency , Sleep Stages/physiology , Synaptic Transmission/physiology , Wakefulness/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sleep/genetics , Sleep/physiology , Sleep Stages/genetics , Synaptic Transmission/genetics , Up-Regulation/genetics , Wakefulness/genetics
6.
Am J Med Sci ; 344(6): 473-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23187120

ABSTRACT

BACKGROUND: The chemokine monocyte chemoattractant protein-1 (MCP-1) has been shown to contribute to neuropathic pain. However, whether MCP-1 is involved in the development of morphine antinociceptive tolerance is incompletely understood. METHODS: Morphine antinociceptive tolerance was induced by intrathecal administration of 15 µg of morphine daily for 7 days. Immunohistochemistry was used to test the changes in the morphology of spinal MCP-1 immunoreactivity and OX-42-IR. The role of MCP-1 in morphine antinociceptive tolerance is explored by hot-water tail-flick test. RESULTS: Our findings showed that intrathecal chronic morphine exposure obviously increased MCP-1 immunoreactivity in the spinal cord. Moreover, the increased MCP-1 immunoreactivity was observed mainly in the spinal neurons. Intrathecal injections of MCP-1-neutralizing antibody significantly reduced the development of morphine antinociceptive tolerance, suggesting that spinal neuronal MCP-1 contributes to tolerance to morphine antinociception. Treatment with MCP-1-neutralizing antibody also reduced the spinal microglial activation induced by chronic morphine treatment. CONCLUSIONS: This study revealed for the first time that spinal neuronal MCP-1 is a key mediator of the spinal microglial activation and that spinal MCP-1 is involved in morphine antinociceptive tolerance. Inhibition of MCP-1 may provide a new therapy for morphine tolerance management.


Subject(s)
Analgesics, Opioid/administration & dosage , Chemokine CCL2/physiology , Drug Tolerance/physiology , Morphine/administration & dosage , Spinal Cord/physiology , Animals , Antibodies, Neutralizing/administration & dosage , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/immunology , Immunohistochemistry , Male , Microglia/drug effects , Microglia/physiology , Nociception/drug effects , Nociception/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Up-Regulation/drug effects
7.
Brain Inj ; 25(6): 604-13, 2011.
Article in English | MEDLINE | ID: mdl-21534738

ABSTRACT

PRIMARY OBJECTIVE: Recent evidence suggests that delayed hypoxic post-conditioning is neuroprotective. The aim of the present study was to test whether early post-conditioning applied immediately after hypoxia could protect cultured neurons from hypoxia/reoxygenation (H/R)-induced injuries. METHODS: Primary cortical neuronal culture depleted of microglia was exposed to H/R. Post-conditioning started immediately after hypoxia and consisted of three cycles of 15-minutes of reoxygenation and 15-minutes of hypoxia. Cell viability assay was performed using Cell Counting Kit-8 (CCK-8). Apoptosis was evaluated by Hoechst 33258 staining, FITC-Annexin V/PI double staining and Western blot assay (testing the cleaved caspase-3 expression). Reactive oxygen species (ROS), intracellular Ca(2+) and mitochondrial membrane potential (MMP) were examined using confocal laser-scanning microscopy. MAIN RESULTS: H/R significantly reduced cell viability and increased neuronal apoptosis and necrosis. Furthermore, the expression of cleaved caspase-3, ROS production and intracellular Ca(2+) were increased. MMP was attenuated. Injuries induced by H/R were substantially attenuated by early hypoxic post-conditioning. Changes in cleaved caspase-3 expression, ROS production, intracellular Ca(2+) level and MMP in response to H/R were significantly decreased by the post-conditioning. CONCLUSIONS: The findings demonstrated that early hypoxic post-conditioning could protect neurons against H/R-induced injuries independent of microglial cells, possibly by inhibiting ROS over-production and intracellular Ca(2+) accumulation and maintaining MMP.


Subject(s)
Apoptosis/physiology , Cell Hypoxia/physiology , Cell Survival/physiology , Cerebral Cortex/metabolism , Neurons/physiology , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cells, Cultured , Cerebral Cortex/physiopathology , Rats , Rats, Sprague-Dawley
8.
Clin Exp Pharmacol Physiol ; 37(3): 316-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19769612

ABSTRACT

1. The aim of the present study was to investigate the effect of hydrogen sulphide (H(2)S) on cobalt chloride (CoCl(2))-induced injury in H9c2 embryonic rat cardiac cells. 2. After 36 h incubation in the presence of 600 micromol/L CoCl(2), reduced cell viability of H9c2 cells was observed, as well as the induction of apoptosis. In addition, CoCl(2) (600 micromol/L) enhanced the production of reactive oxygen species (ROS) and the expression of cleaved caspase 3, induced a loss of mitochondrial membrane potential (MMP) and decreased reduced glutathione (GSH) production. These results suggest that CoCl(2) induces similar responses to hypoxia/ischaemia. 3. Pretreatment of cells with 400 micromol/L NaHS (a H(2)S donor) for 30 min prior to exposure to CoCl(2) (600 micromol/L) significantly protected H9c2 cells against CoCl(2)-induced injury. Specifically, increased cell viability and decreased apoptosis were observed. In addition, NaHS pretreatment blocked the CoCl(2)-induced increases in ROS production and cleaved caspase 3 expression, as well as the decreases in GSH production and loss of MMP. 4. Pretreatment of cells with 2000 micromol/L N-acetylcysteine (NAC), a ROS scavenger, for 1 h prior to CoCl(2) exposure significantly protected H9c2 cells against CoCl(2)-induced injury, specifically enhancing cell viability, decreasing ROS production and preventing loss of MMP. 5. The findings of the present study suggest that H(2)S protects H9c2 cells against CoCl(2)-induced injury by suppressing oxidative stress and caspase 3 activation.


Subject(s)
Cobalt/toxicity , Cytoprotection/drug effects , Cytoprotection/physiology , Hydrogen Sulfide/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Myocardium/cytology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Reactive Oxygen Species/metabolism
9.
Neurosci Lett ; 467(2): 95-9, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19818835

ABSTRACT

N-methyl-d-aspartate (NMDA) receptors and c-Jun N-terminal kinase (JNK) have been shown to be involved in morphine antinociceptive tolerance. However, whether chronic morphine-induced activation of the spinal JNK is NMDA receptor-dependent is unknown. The present study investigated the link between the spinal NMDA receptor NR2B subunit and the JNK activation during morphine antinociceptive tolerance in rats. Our results showed that chronic morphine treatment induced upregulation of the NR2B expression and activation of JNK in the spinal cord. Moreover, the increased NR2B-immunoreactivity (IR) and phosphorylated JNK-IR were observed mainly at the superficial dorsal horn laminae of the spinal cord; the spinal p-JNK was mainly expressed in astrocytes and NR2B in neurons. SP600125, a selective inhibitor of JNK, significantly attenuated morphine tolerance. MK-801, a noncompetitive NMDA receptor antagonist, not only suppressed morphine antinociceptive tolerance and the increase in NR2B, but also reduced the spinal JNK activation induced by chronic morphine treatment. These findings demonstrated for the first time that NMDA receptor-dependent activation of the spinal JNK contributes to morphine antinociceptive tolerance and that MK-801 attenuates morphine tolerance partly due to its inhibition on the spinal JNK activation.


Subject(s)
Analgesics, Opioid/pharmacology , JNK Mitogen-Activated Protein Kinases/physiology , Morphine/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Spinal Cord/metabolism , Animals , Anthracenes/pharmacology , Dizocilpine Maleate/pharmacology , Drug Tolerance , Enzyme Activation , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spinal Cord/drug effects
10.
Clin Exp Pharmacol Physiol ; 36(3): 304-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18986327

ABSTRACT

1. Cytoprotection by H(2)O(2) preconditioning against oxidative stress-induced apoptosis of PC12 cells has been demonstrated previously. In the present study, we investigated the effects of H(2)O(2) preconditioning on nuclear factor (NF)-kappaB activation and the role of NF-kappaB in the adaptive cytoprotection of H(2)O(2) preconditioning in PC12 cells. 2. The PC12 cells were preconditioned with 100 micromol/L H(2)O(2) for 90 min, followed by 24 h recovery and subsequent exposure to 300 micromol/L H(2)O(2) for a further 12 h. 3. The results showed that preconditioning with 100 micromol/L H(2)O(2) upregulated NF-kappaB expression and enhanced its nuclear translocation and DNA binding activity. In addition to its own effects on NF-kappaB expression, H(2)O(2) preconditioning also promoted the overexpression of NF-kappaB induced by a lethal concentration of H(2)O(2) (300 micromol/L). 4. N-Tosyl-l-phenylalanine chloromethyl ketone (TPCK; 20 micromol/L), an inhibitor of NF-kappaB, was administered 20 min before preconditioning with 100 micromol/L H(2)O(2). At this concenteration, TPCK blocked the overexpression of NF-kappaB induced by H(2)O(2) preconditioning, accompanied by attenuation of H(2)O(2) preconditioning-induced cytoprotection. The inhibition of NF-kappaB by TPCK enhanced caspase 3 activity induced by 300 micromol/L H(2)O(2). 5. The findings of the present study provide novel evidence for the effects of preconditioning with H(2)O(2) on constitutive activation of NF-kappaB, which contributes to the adaptive cytoprotection of H(2)O(2) preconditioning against PC12 cells apoptosis.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Apoptosis/drug effects , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Oxidative Stress/drug effects , Pheochromocytoma/metabolism , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus , Adrenal Gland Neoplasms/pathology , Animals , Cytoprotection , DNA/metabolism , Dose-Response Relationship, Drug , Hydrogen Peroxide/toxicity , Oxidants/toxicity , PC12 Cells , Pheochromocytoma/pathology , Rats , Time Factors , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Transcription Factor RelA/antagonists & inhibitors
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 28(8): 1345-8, 2008 Aug.
Article in Chinese | MEDLINE | ID: mdl-18753056

ABSTRACT

OBJECTIVE: To investigate the effects of angiotensin (Ang)-(1-7) on oxidative stress and functional changes in isolated rat hearts with myocardial ischemia-reperfusion (IR) injury. METHODS: IR injury was induced in isolated rat hearts with the Langendorff' apparatus. The left ventricular systolic pressure (LVSP) of the rat heart was measured using a pressure transducer. Malondialdehyde (MDA) level and superoxide dismutase (SOD) activity in the myocardium were detected using commercial kits. RESULTS: Myocardial ischemia (15 min) and reperfusion (30 min) significantly increased myocardial levels of MDA, and reduced the SOD activity and LVSP (P<0.05). Pretreatment with Ang-(1-7) at 1.0 nmol/L 30 min before ischemia obviously inhibited IR-induced MDA increment and lowering of SOD activity and LVSP. Pretreatment of the rats with intraperitoneal injection of 5 mg/kg indomethacin 1 h before isolation of the heart markedly antagonized the effect of Ang-(1-7) on MDA production, SOD activity and LVSP. CONCLUSION: Angiotensin-(1-7) can inhibit IR injury-induced oxidative stress and decrease in cardiac contractile function in isolated rat hearts. The mechanism underlying the effect of Ang-(1-7) may be associated with increased secretion of prostaglandin.


Subject(s)
Angiotensin I/pharmacology , Heart/drug effects , Myocardial Reperfusion Injury/physiopathology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Animals , Heart/physiology , In Vitro Techniques , Male , Myocardial Reperfusion Injury/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
12.
Brain Behav Immun ; 22(1): 114-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17919885

ABSTRACT

We have previously demonstrated that activation of p38 mitogen-activated protein kinase (p38 MAPK) in the spinal microglia mediates morphine antinociceptive tolerance. Minocycline, a selective inhibitor of microglia activation, has been reported to attenuate peripheral inflammation-induced hyperalgesia by depressing p38 MAPK in the spinal microglia. The aim of the present study is to explore the effect of intrathecal minocycline on the development of morphine antinociceptive tolerance and p38 activation in the spinal microglia induced by chronic morphine treatment. Minocycline (20, 50 and 100 microg) was given intrathecally 30 min before each morphine (15 microg) administration for consecutive 7 days. It was shown that minocycline attenuated tolerance to morphine analgesia in a dose-dependent manner. Minocycline administration (50 microg) which was initiated on day 4 followed by another 4 days administration partially reversed the established morphine antinociceptive tolerance. However, minocycline treatment which was started on day 8 followed by its administration for 4 more days failed to reverse the established morphine tolerance. Immunohistochemical analysis showed that chronic intrathecal morphine-induced activation of p38 MAPK in the spinal microglia. Minocycline at a dose that was shown to antagonize tolerance to morphine analgesia significantly inhibited the increase in p38 MAPK activation in the spinal microglia. To our knowledge, this is the first study to demonstrate that minocycline antagonizes morphine antinociceptive tolerance, possibly due to the inhibition of p38 activation in the spinal microglia.


Subject(s)
Analgesics, Opioid/pharmacology , Microglia/enzymology , Minocycline/pharmacology , Morphine/pharmacology , Nociceptors/drug effects , Spinal Cord/enzymology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Tolerance , Enzyme Activation/drug effects , Immunohistochemistry , Injections, Spinal , Male , Minocycline/administration & dosage , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Brain Res ; 1069(1): 235-43, 2006 Jan 19.
Article in English | MEDLINE | ID: mdl-16403466

ABSTRACT

Compelling evidence has suggested that spinal glial cells were activated by chronic morphine treatment and involved in the development of morphine tolerance. However, the mechanisms of glial activation were still largely unknown in morphine tolerance. In present study, we investigated the role of p38 mitogen-activated protein kinase (p38 MAPK) in the spinal cord in the development of chronic morphine antinociceptive tolerance. We found that intrathecal administration of morphine (15 microg) daily for 7 consecutive days significantly induced an increase in number of phospho-p38 (p-p38) immunoreactive cells in the spinal cord compared with chronic saline or acute morphine treated rats. Double immunofluorescence staining revealed that p-p38 immunoreactivity was exclusively restricted in the activated spinal microglia, not in astrocytes or neurons. Repeated intrathecal administration of 4-(4-fluorophenyl)-2-(4-methylsulfonylphenyl)-5-(4-pyridyl)-1H-imidazole (SB203580) (10 microg or 2 microg), a specific p38 inhibitor, 30 min before each morphine injection for 7 consecutive days significantly attenuated tolerance to morphine analgesia assessed by tail flick test. However, a single intrathecal administration of SB203580 (10 microg) did not antagonize the established tolerance to morphine analgesia. Taken together, these findings suggested that p38 MAPK activation in the spinal microglia was involved in the development of morphine antinociceptive tolerance. Inhibition of p38 MAPK by SB203580 in the spinal cord attenuated but not reversed the tolerance to morphine analgesia. The present study provides the first evidence that p38 activation in spinal microglia played an important role in the development of tolerance to morphine analgesia.


Subject(s)
Analgesics, Opioid/administration & dosage , Microglia/physiology , Morphine Dependence , Morphine/administration & dosage , Spinal Cord/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Analgesics, Opioid/adverse effects , Analysis of Variance , Animals , Behavior, Animal , CD11b Antigen/metabolism , Cell Count/methods , Drug Administration Schedule , Drug Interactions , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Imidazoles/pharmacology , Immunohistochemistry/methods , Male , Morphine/adverse effects , Morphine Dependence/metabolism , Morphine Dependence/pathology , Morphine Dependence/physiopathology , Phosphopyruvate Hydratase/metabolism , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
14.
Brain Res ; 1057(1-2): 57-64, 2005 Sep 28.
Article in English | MEDLINE | ID: mdl-16129420

ABSTRACT

The present study is designed to investigate the effects of preconditioning with different doses of hydrogen peroxide (H2O2) on oxidative stress-induced apoptosis and the changes in mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) level, and expression of Bcl-2 during H2O2 preconditioning in rat pheochromocytoma (PC12) cells. It was shown that (1) H2O2 induced apoptosis in PC12 cells in a dose-dependent manner; (2) the preconditioning with 10 micromol L(-1) or 20 micromol L(-1) H2O2 can significantly protect PC12 cells against apoptosis induced by 50 or 100 micromol L(-1) H2O2, low (5 micromol L(-1)) and higher (30 micromol L(-1)) concentrations of H2O2 had no cytoprotections; (3) high concentration (100 micromol L(-1)) of H2O2 reduced MMP and expression of Bcl-2, and increased ROS level, but these effects were blocked by preconditioning with 10 micromol L(-1) H2O2; (4) the preconditioning with 10 micromol L(-1) H2O2 induced overexpression of Bcl-2. These results suggested that the preconditioning with low dose of H2O2 could protect the oxidative stress-induced PC12 cells apoptosis not only by preventing the reduction of MMP and expression of Bcl-2 as well as increase in ROS level, but also through overexpression of Bcl-2. It was indicated that overexpression of Bcl-2 may play a key role in the cytoprotection induced by preconditioning with low dose of H2O2 in PC12 cells.


Subject(s)
Apoptosis/drug effects , Hydrogen Peroxide/pharmacology , Membrane Potentials/drug effects , Oxidative Stress/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/physiology , Blotting, Western/methods , Cell Count/methods , Cell Death , Dose-Response Relationship, Drug , Flow Cytometry/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry/methods , Membrane Potentials/physiology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Oxidative Stress/drug effects , PC12 Cells , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...