Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
J Neurotrauma ; 35(3): 508-520, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29048243

ABSTRACT

After spinal cord injury (SCI), blood-spinal cord barrier (BSCB) disruption and progressive hemorrhage lead to secondary injury, subsequent apoptosis and/or necrosis of neurons and glia, causing permanent neurological deficits. Growing evidence indicates that mithramycin A (MA), an anti-cancer drug, has neuroprotective effects in ischemic brain injury and Huntington's disease (HD). However, the precise mechanism underlying its protective effects is largely unknown. Here, we examined the effect of MA on BSCB breakdown and hemorrhage as well as subsequent inflammation after SCI. After moderate spinal cord contusion injury at T9, MA (150 µg/kg) was immediately injected intraperitoneally (i.p.) and further injected once a day for 5 days. Our data show that MA attenuated BSCB disruption and hemorrhage, and inhibited the infiltration of neutrophils and macrophages after SCI. Consistent with these findings, the expression of inflammatory mediators was significantly alleviated by MA. MA also inhibited the expression and activation of matrix metalloprotease-9 (MMP-9) after injury, which is known to disrupt BSCB and the degradation of tight junction (TJ) proteins. In addition, the expression of sulfonylurea receptor 1 (SUR1) and transient receptor potential melastatin 4 (TRPM4), which are known to mediate hemorrhage at an early stage after SCI, was significantly blocked by MA treatment. Finally, MA inhibited apoptotic cell death and improved functional recovery after injury. Thus, our results demonstrated that MA improves functional recovery by attenuating BSCB disruption and hemorrhage through the downregulation of SUR1/TRPM4 and MMP-9 after SCI.


Subject(s)
Capillary Permeability/drug effects , Neuroprotective Agents/pharmacology , Plicamycin/analogs & derivatives , Recovery of Function/drug effects , Spinal Cord Injuries/pathology , Animals , Hemorrhage/pathology , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/drug effects , Plicamycin/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonylurea Receptors/biosynthesis , Sulfonylurea Receptors/drug effects , TRPM Cation Channels/biosynthesis , TRPM Cation Channels/drug effects
2.
J Cereb Blood Flow Metab ; 37(12): 3695-3708, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28338398

ABSTRACT

The blood-brain barrier (BBB) exhibits a highly selective permeability to support the homeostasis of the central nervous system (CNS). The tight junctions in the BBB microvascular endothelial cells seal the paracellular space to prevent diffusion. Thus, disruption of tight junctions results in harmful effects in CNS diseases and injuries. It has recently been demonstrated that glucocorticoids have beneficial effects on maintaining tight junctions in both in vitro cell and in vivo animal models. In the present study, we found that dexamethasone suppresses the expression of JMJD3, a histone H3K27 demethylase, via the recruitment of glucocorticoid receptor α (GRα) and nuclear receptor co-repressor (N-CoR) to the negative glucocorticoid response element (nGRE) in the upstream region of JMJD3 gene in brain microvascular endothelial cells subjected to TNFα treatment. The decreased JMJD3 gene expression resulted in the suppression of MMP-2, MMP-3, and MMP-9 gene activation. Dexamethasone also activated the expression of the claudin 5 and occludin genes. Collectively, dexamethasone attenuated the disruption of the tight junctions in the brain microvascular endothelial cells subjected to TNFα treatment. Therefore, glucocorticoids may help to preserve the integrity of the tight junctions in the BBB via transcriptional and post-translational regulation following CNS diseases and injuries.


Subject(s)
Brain/blood supply , Dexamethasone/pharmacology , Down-Regulation/drug effects , Endothelial Cells/drug effects , Glucocorticoids/pharmacology , Jumonji Domain-Containing Histone Demethylases/genetics , Tight Junctions/drug effects , Animals , Brain/drug effects , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Mice , Microvessels/cytology , Microvessels/drug effects , Microvessels/metabolism , Receptors, Glucocorticoid/metabolism , Tight Junctions/metabolism , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism
3.
Neurobiol Dis ; 95: 66-81, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27425890

ABSTRACT

The disruption of the blood-spinal cord barrier (BSCB) by matrix metalloprotease (MMP) activation is a detrimental event that leads to blood cell infiltration, inflammation, and apoptosis, thereby contributing to permanent neurological disability after spinal cord injury (SCI). However, the molecular mechanisms underlying Mmp gene regulation have not been fully elucidated. Here, we demonstrated the critical role of histone H3K27 demethylase Jmjd3 in the regulation of Mmp gene expression and BSCB disruption using in vitro cellular and in vivo animal models. We found that Jmjd3 up-regulation, in cooperation with NF-κB, after SCI is required for Mmp-3 and Mmp-9 gene expressions in injured vascular endothelial cells. In addition, Jmjd3 mRNA depletion inhibited Mmp-3 and Mmp-9 gene expressions and significantly attenuated BSCB permeability and the loss of tight junction proteins. These events further led to improved functional recovery, along with decreased hemorrhage, blood cell infiltration, inflammation, and cell death of neurons and oligodendrocytes after SCI. Thus, our findings suggest that Jmjd3 regulation may serve as a potential therapeutic intervention for preserving BSCB integrity following SCI.


Subject(s)
Gene Expression Regulation/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Animals , Blood-Brain Barrier , Capillary Permeability/genetics , Endothelial Cells/metabolism , Male , NF-kappa B/metabolism , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord/metabolism , Up-Regulation
4.
Neuropharmacology ; 109: 78-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27256500

ABSTRACT

Recently we reported that fluoxetine (10 mg/kg) improves functional recovery by attenuating blood spinal cord barrier (BSCB) disruption after spinal cord injury (SCI). Here we investigated whether a low-dose of fluoxetine (1 mg/kg) and vitamin C (100 mg/kg), separately not possessing any protective effect, prevents BSCB disruption and improves functional recovery when combined. After a moderate contusion injury at T9 in rat, a low-dose of fluoxetine and vitamin C, or the combination of both was administered intraperitoneally immediately after SCI and further treated once a day for 14 d. Co-treatment with fluoxetine and vitamin C significantly attenuated BSCB permeability at 1 d after SCI. When only fluoxetine or vitamin C was treated after injury, however, there was no effect on BSCB disruption. Co-treatment with fluoxetine and vitamin C also significantly inhibited the expression and activation of MMP-9 at 8 h and 1 d after injury, respectively, and the infiltration of neutrophils (at 1 d) and macrophages (at 5 d) and the expression of inflammatory mediators (at 2 h, 6 h, 8 h or 24 h after injury) were significantly inhibited by co-treatment with fluoxetine and vitamin C. Furthermore, the combination of fluoxetine and vitamin C attenuated apoptotic cell death at 1 d and 5 d and improved locomotor function at 5 weeks after SCI. These results demonstrate the synergistic effect combination of low-dose fluoxetine and vitamin C on BSCB disruption after SCI and furthermore support the effectiveness of the combination treatment regimen for the management of acute SCI.


Subject(s)
Ascorbic Acid/administration & dosage , Blood-Brain Barrier/drug effects , Fluoxetine/administration & dosage , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Animals , Antioxidants/administration & dosage , Blood-Brain Barrier/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Selective Serotonin Reuptake Inhibitors/administration & dosage , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
5.
J Ethnopharmacol ; 186: 169-180, 2016 Jun 20.
Article in English | MEDLINE | ID: mdl-27036628

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Anti-inflammatory therapy has been intensively investigated as a potential strategy for treatment of cerebral stroke. However, despite many positive outcomes reported in animal studies, anti-inflammatory treatments have not proven successful in humans as yet. Although immunomodulatory activity and safety of Cordyceps species (Chinese caterpillar fungi) have been proven in clinical trials and traditional Asian prescriptions for inflammatory diseases, its anti-ischemic effect remains elusive. AIM OF THE STUDY: In the present study, therefore, we investigated the potential therapeutic efficacy of WIB801C, the standardized extract of Cordyceps militaris, for treatment of cerebral ischemic stroke. MATERIALS AND METHODS: The anti-chemotactic activity of WIB801C was assayed in cultured rat microglia/macrophages. Sprague-Dawley rats were subjected to ischemic stroke via either transient (1.5-h tMCAO and subsequent 24-h reperfusion) or permanent middle cerebral artery occlusion (pMCAO for 24-h without reperfusion). WIB801C was orally administered twice at 3- and 8-h (50mg/kg each) after the onset of MCAO. Infarct volume, edema, blood brain barrier and white matter damages, neurological deficits, and long-term survival rates were investigated. The infiltration of inflammatory cells into ischemic lesions was assayed by immunostaining. RESULTS: WIB801C significantly decreased migration of cultured microglia/macrophages. This anti-chemotactic activity of WIB-801C was not mediated via adenosine A3 receptors, although cordycepin, the major ingredient of WIB801C, is known as an adenosine receptor agonist. Post-ischemic treatment with WIB801C significantly reduced the infiltration of ED-1-and MPO-positive inflammatory cells into ischemic lesions in tMCAO rats. WIB801C-treated rats exhibited significantly decreased infarct volume and cerebral edema, less white matter and blood-brain barrier damages, and improved neurological deficits. WIB801C also improved survival rates over 34 days after ischemia onset. A significant reduction in infarct volume and neurobehavioral deficits by WIB801C was also observed in rats subjected to pMCAO. CONCLUSIONS: In summary, post-ischemic treatment of WIB801C reduced infiltration of inflammatory cells into ischemic lesions via inhibition of chemotaxis, which confers long-lasting histological and neurological protection in ischemic brain. WIB801C may be a promising anti-ischemic drug candidate with clinically relevant therapeutic time window and safety.


Subject(s)
Brain Ischemia/drug therapy , Cell Movement/drug effects , Cordyceps/chemistry , Plant Extracts/pharmacology , Animals , Cells, Cultured , Chemotaxis/drug effects , Male , Microglia/drug effects , Plant Extracts/administration & dosage , Rats , Rats, Sprague-Dawley
6.
Neurobiol Dis ; 82: 141-151, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26079709

ABSTRACT

The activation of microglia after spinal cord injury (SCI) contributes to secondary damage by producing pro-inflammatory cytokines and mediators, leading to cell death of oligodendrocytes and neurons. Here, we show that matrix metalloprotease-3 (MMP-3) produced and secreted in the endothelial cells of blood vessels after SCI mediates microglial activation. MMP-3 was produced and secreted in bEnd.3 cells, a mouse brain-derived endothelial cell line, by oxygen-glucose deprivation/reoxygenation (OGD/RO). OGD/RO-induced MMP-3 expression and activity was also significantly inhibited by ghrelin, which was dependent on the ghrelin receptor GHS-R1a. Furthermore, the secreted MMP-3 from OGD/RO-induced bEnd.3 cells activated BV-2 cells, a murine microglial cell line. We also found that microglial activation after SCI was attenuated in MMP-3 knockout (KO) mice compared with wild type (WT) mice. Both p38 mitogen-activated protein kinase (MAPK) activation and pro-nerve growth factor (proNGF) production were more inhibited in MMP-3 KO than WT mice at 5d after injury. When WT mice were treated with Mmp-3 siRNA after injury, MMP-3 activity, microglial activation, p38MAPK activation and proNGF expression were significantly inhibited. Ghrelin treatment also significantly inhibited MMP-3 expression and activation after SCI, which was dependent on GHS-R1a. Finally, RhoA activation and oligodendrocyte cell death after injury were attenuated by Mmp-3 siRNA or ghrelin treatment compared with vehicle control. Thus, our study indicates that MMP-3 produced in blood vessel endothelial cells after SCI serves as an endogenous molecule for microglial activation followed by p38MAPK activation and proNGF production, and further indicates that the protective effect of ghrelin on oligodendrocytes cell death may be at least partly mediated by the inhibition of MMP-3-induced microglial activation after SCI.


Subject(s)
Cell Death/physiology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Matrix Metalloproteinase 3/metabolism , Microglia/metabolism , Oligodendroglia/metabolism , Spinal Cord Injuries/metabolism , Animals , Cell Death/drug effects , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Ghrelin/pharmacology , Mice , Microglia/drug effects , Microglia/pathology , Nerve Growth Factors/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Spinal Cord Injuries/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Endocrinology ; 156(5): 1838-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25763638

ABSTRACT

Blood-spinal cord barrier (BSCB) disruption and progressive hemorrhage after spinal cord injury (SCI) lead to secondary injury and the subsequent apoptosis and/or necrosis of neuron and glia, causing permanent neurological deficits. In this study, we examined the effect of 17ß-estradiol (E2) on BSCB breakdown and hemorrhage as well as subsequent inflammation after SCI. After a moderate contusion injury at the 9th thoracic segment of spinal cord, E2 (300 µg/kg) was administered by iv injection immediately after SCI, and the same dose of E2 was then administered 6 and 24 hours after injury. Our data show that E2 attenuated BSCB permeability and hemorrhage and reduced the infiltration of neutrophils and macorphages after SCI. Consistent with this finding, the expression of inflammatory mediators was significantly reduced by E2. Furthermore, E2 treatment significantly inhibited the expression of sulfonylurea receptor 1 and transient receptor potential melastatin 4 after injury, which are known to mediate hemorrhage at an early stage after SCI. Moreover, the expression and activation of matrix metalloprotease-9 after injury, which is known to disrupt BSCB, and the degradation of tight junction proteins, such as zona occludens-1 and occludin, were significantly inhibited by E2 treatment. Furthermore, the protective effects of E2 on BSCB disruption and functional improvement were abolished by an estrogen receptor antagonist, ICI 182780 (3 mg/kg). Thus, our study provides evidence that the neuroprotective effect of E2 after SCI is, in part, mediated by inhibiting BSCB disruption and hemorrhage through the down-regulation of sulfonylurea receptor 1/transient receptor potential melastatin 4 and matrix metalloprotease-9, which is dependent on estrogen receptor.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Estradiol/pharmacology , Hemorrhage/metabolism , Matrix Metalloproteinase 9/drug effects , Spinal Cord Injuries/metabolism , Sulfonylurea Receptors/drug effects , TRPM Cation Channels/drug effects , Animals , Apoptosis , Blood-Brain Barrier/metabolism , Down-Regulation/drug effects , Estradiol/analogs & derivatives , Estrogen Receptor Antagonists/pharmacology , Fulvestrant , Male , Matrix Metalloproteinase 9/metabolism , Rats , Rats, Sprague-Dawley , Sulfonylurea Receptors/metabolism , TRPM Cation Channels/metabolism
8.
J Neurotrauma ; 32(9): 633-44, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25366938

ABSTRACT

Oligodendrocyte cell death and axon demyelination after spinal cord injury (SCI) are known to be important secondary injuries contributing to permanent neurological disability. Thus, blocking oligodendrocyte cell death should be considered for therapeutic intervention after SCI. Here, we demonstrated that fluoxetine, an antidepressant drug, alleviates oligodendrocyte cell death by inhibiting microglia activation after SCI. After injury at the T9 level with a Precision Systems and Instrumentation (Lexington, KY) device, fluoxetine (10 mg/kg, intraperitoneal) was administered once a day for the indicated time points. Immunostaining with CD11b (OX-42) antibody and quantification analysis showed that microglia activation was significantly inhibited by fluoxetine at 5 days after injury. Fluoxetine also significantly inhibited activation of p38 mitogen-activated protein kinase (p38-MAPK) and expression of pro-nerve growth factor (pro-NGF), which is known to mediate oligodendrocyte cell death through the p75 neurotrophin receptor after SCI. In addition, fluoxetine attenuated activation of Ras homolog gene family member A and decreased the level of phosphorylated c-Jun and, ultimately, alleviated caspase-3 activation and significantly reduced cell death of oligodendrocytes at 5 days after SCI. Further, the decrease of myelin basic protein, myelin loss, and axon loss in white matter was also significantly blocked by fluoxetine, as compared to vehicle control. These results suggest that fluoxetine inhibits oligodendrocyte cell death by inhibiting microglia activation and p38-MAPK activation, followed by pro-NGF production after SCI, and provide a potential usage of fluoxetine for a therapeutic agent after acute SCI in humans.


Subject(s)
Fluoxetine/therapeutic use , Microglia/drug effects , Oligodendroglia/drug effects , Selective Serotonin Reuptake Inhibitors/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Animals , Apoptosis/drug effects , Disease Models, Animal , Drug Administration Schedule , Injections, Intraperitoneal , Macrophage Activation/drug effects , Male , Mice, Inbred C57BL , Nerve Growth Factor/metabolism , Spinal Cord Injuries/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Biochim Biophys Acta ; 1842(12 Pt A): 2403-12, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25261791

ABSTRACT

Blood spinal cord barrier (BSCB) disruption after spinal cord injury (SCI) leads to secondary injury and results in apoptosis of neurons and glia, leading to permanent neurological deficits. Here, we examined the effect of ghrelin on BSCB breakdown and hemorrhage after SCI. After moderate weight-drop contusion injury at T9 spinal cord, ghrelin (80µg/kg) was administered via intraperitoneal injection immediately after SCI and then the same dose of ghrelin was treated every 6h for 1d. Our data showed that ghrelin treatment significantly inhibited the expression and activation of matrix metalloprotease-9 (MMP-9) at 1d after SCI. The increases of sulfonylurea receptor 1 (SUR1) and transient receptor potential melastatin 4 (TrpM4) expressions at 1h and 8h after SCI respectively were also alleviated by ghrelin treatment. In addition, both BSCB breakdown and hemorrhage at 1d after injury were significantly attenuated by ghrelin. In parallel, the infiltration of blood cells such as neutrophils and macrophages was inhibited by ghrelin treatment at 1d and 5d after SCI respectively. We also found that ghrelin receptor, growth hormone secretagogue receptor-1a (GHS-R1a), was expressed in the blood vessel of normal spinal tissue. Furthermore, the inhibitory effects of ghrelin on hemorrhage and BSCB disruption at 1d after SCI were blocked by GHS-R1a antagonist, [D-Lys-3]-GHRP-6 (3mg/kg). Thus, these results indicate that the neuroprotective effect by ghrelin after SCI is mediated in part by blocking BSCB disruption and hemorrhage through the down-regulation of SUR1/TrpM4 and MMP-9, which is dependent on GHS-R1a.


Subject(s)
Ghrelin/pharmacology , Matrix Metalloproteinase 9/genetics , Spinal Cord Injuries/genetics , Spinal Cord/drug effects , Sulfonylurea Receptors/genetics , TRPM Cation Channels/genetics , Animals , Blotting, Western , Capillary Permeability/drug effects , Gene Expression/drug effects , Ghrelin/administration & dosage , Hemorrhage/prevention & control , Immunohistochemistry , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Oligopeptides/pharmacology , Rats, Sprague-Dawley , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/blood supply , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Sulfonylurea Receptors/metabolism , TRPM Cation Channels/metabolism , Time Factors
10.
Neuropharmacology ; 79: 161-71, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24316161

ABSTRACT

Ischemia induces blood-brain barrier (BBB) disruption by matrix metalloproteases (MMPs) activation, leading to neuronal cell death. Here, we show that fluoxetine inhibits apoptotic cell death of hippocampal neuron and memory impairment by blocking BBB disruption after transient global ischemia. Fluoxetine treatment (10 mg/kg) after global ischemia significantly inhibited mRNA expression of MMP-2 and -9 and reduced MMP-9 activity. By Evan blue assay, fluoxetine reduced ischemia-induced BBB permeability. In parallel, fluoxetine significantly attenuated the loss of occludin and laminin in the hippocampal area after ischemia. By immunostaining with occludin antibody, fluoxetine preserved the integrity of vascular networks, especially in hippocampal areas after injury. Fluoxetine also prevented the infiltration of macrophages and inhibited the mRNA expression of inflammatory mediators after injury. In addition, the activation of microglia and astrocyte in hippocampal regions was significantly attenuated by fluoxetine. Finally, fluoxetine reduced apoptotic cell death of hippocampal neurons as well as vascular endothelial cell death and improved learning and memory. Thus, our study suggests that the neuroprotective effect of fluoxetine is likely mediated by blocking MMP activation followed BBB disruption after transient global ischemia, and the drug may represent a potential therapeutic agent for preserving BBB integrity following ischemic brain injury in humans.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Cell Death/drug effects , Fluoxetine/pharmacology , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Blood-Brain Barrier/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Death/physiology , Gene Expression/drug effects , Hippocampus/blood supply , Hippocampus/pathology , Hippocampus/physiopathology , Laminin/metabolism , Macrophages/drug effects , Macrophages/physiology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred Strains , Microglia/drug effects , Microglia/physiology , Neurons/drug effects , Neurons/physiology , Occludin/metabolism , RNA, Messenger/metabolism
11.
J Neurotrauma ; 31(6): 582-94, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24294888

ABSTRACT

Both oxidative stress and endoplasmic reticulum (ER) stress are known to contribute to secondary injury, ultimately leading to cell death after spinal cord injury (SCI). Here, we showed that valproic acid (VPA) reduced cell death of motor neurons by inhibiting cytochrome c release mediated by oxidative stress and ER stress after SCI. After SCI, rats were immediately injected with VPA (300 mg/kg) subcutaneously and further injected every 12 h for an indicated time period. Motor neuron cell death at an early time after SCI was significantly attenuated by VPA treatment. Superoxide anion (O2-) production and inducible NO synthase (iNOS) expression linked to oxidative stress was increased after injury, which was inhibited by VPA. In addition, VPA inhibited c-Jun N-terminal kinase (JNK) activation, which was activated and peaked at an early time after SCI. Furthermore, JNK activation and c-Jun phosphorylation were inhibited by a broad-spectrum reactive oxygen species (ROS) scavenger, Mn (III) tetrakis (4-benzoic acid) porphyrin (MnTBAP), indicating that ROS including O2- increased after SCI probably contribute to JNK activation. VPA also inhibited cytochrome c release and caspase-9 activation, which was significantly inhibited by SP600125, a JNK inhibitor. The levels of phosphorylated Bim and Mcl-1, which are known as downstream targets of JNK, were significantly reduced by SP600125. On the other hand, VPA treatment inhibited ER stress-induced caspase-12 activation, which is activated in motor neurons after SCI. In addition, VPA increased the Bcl-2/Bax ratio and inhibited CHOP expression. Taken together, our results suggest that cell death of motor neurons after SCI is mediated through oxidative stress and ER stress-mediated cytochrome c release and VPA-inhibited cytochrome c release by attenuating ROS-induced JNK activation followed by Mcl-1 and Bim phosphorylation and ER stress-coupled CHOP expression.


Subject(s)
Cell Death/drug effects , Cytochromes c/metabolism , Endoplasmic Reticulum Stress/drug effects , Motor Neurons/drug effects , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Spinal Cord Injuries/drug therapy , Valproic Acid/therapeutic use , Animals , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Motor Neurons/metabolism , Motor Neurons/pathology , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Valproic Acid/pharmacology
12.
PLoS One ; 8(9): e73948, 2013.
Article in English | MEDLINE | ID: mdl-24040124

ABSTRACT

Acupuncture (AP) has been used worldwide to relieve pain. However, the mechanism of action of AP is poorly understood. Here, we found that AP relieved neuropathic pain (NP) by inhibiting Jun-N-terminal kinase (JNK) activation in astrocytes after spinal cord injury (SCI). After contusion injury which induces the below-level (L4-L5) NP, Shuigou (GV26) and Yanglingquan (GB34) acupoints were applied. At 31 d after injury, both mechanical allodynia and thermal hyperalgesia were significantly alleviated by AP applied at GV26 and GB34. Immunocytochemistry revealed that JNK activation was mainly observed in astrocytes after injury. AP inhibited JNK activation in astrocytes at L4-L5 level of spinal cord. The level of p-c-Jun known, a downstream molecule of JNK, was also decreased by AP. In addition, SCI-induced GFAP expression, a marker for astrocytes, was decreased by AP as compared to control groups. Especially, the number of hypertrophic, activated astrocytes in laminae I-II of dorsal horn at L4-5 was markedly decreased by AP treatment when compared with vehicle and simulated AP-treated groups. When animals treated with SP600125, a specific JNK inhibitor, after SCI, both mechanical allodynia and thermal hyperalgesia were significantly attenuated by the inhibitor, suggesting that JNK activation is likely involved in SCI-induced NP. Also, the expression of chemokines which is known to be mediated through JNK pathway was significantly decreased by AP and SP600125 treatment. Therefore, our results indicate that analgesic effect of AP is mediated in part by inhibiting JNK activation in astrocytes after SCI.


Subject(s)
Acupuncture Therapy , Anesthesia/methods , Astrocytes/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Enzyme Activation , Gene Expression Regulation , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/therapy , Proto-Oncogene Proteins c-jun/metabolism , Rats , Spinal Cord Injuries/genetics
13.
Glia ; 61(11): 1807-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038428

ABSTRACT

Inflammation induced by microglial activation plays a pivotal role in progressive degeneration after traumatic spinal cord injury (SCI). Voltage-gated sodium channels (VGSCs) are also implicated in microglial activation following injury. However, direct evidence that VGSCs are involved in microglial activation after injury has not been demonstrated yet. Here, we show that the increase in VGSC inward current elicited microglial activation followed inflammatory responses, leading to cell death after injury in vitro and in vivo. Isoforms of sodium channel, Nav 1.1, Nav 1.2, and Nav 1.6 were expressed in primary microglia, and the inward current of VGSC was increased by LPS treatment, which was blocked by a sodium channel blocker, tetrodotoxin (TTX). TTX inhibited LPS-induced NF-κB activation, expression of TNF-α, IL-1ß and inducible nitric oxide synthase, and NO production. LPS-induced p38MAPK activation followed pro-nerve growth factor (proNGF) production was inhibited by TTX, whereas LPS-induced JNK activation was not. TTX also inhibited caspase-3 activation and cell death of primary cortical neurons in neuron/microglia co-cultures by inhibiting LPS-induced microglia activation. Furthermore, TTX attenuated caspase-3 activation and oligodendrocyte cell death at 5 d after SCI by inhibiting microglia activation and p38MAPK activation followed proNGF production, which is known to mediate oligodendrocyte cell death. Our study thus suggests that the increase in inward current of VGSC appears to be an early event required for microglia activation after injury.


Subject(s)
Microglia/metabolism , Neurons/metabolism , Sodium Channels/metabolism , Spinal Cord Injuries/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Inflammation/metabolism , Microglia/drug effects , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Spinal Cord Injuries/pathology , Tetrodotoxin/pharmacology , Tumor Necrosis Factor-alpha/metabolism
14.
Brain ; 135(Pt 8): 2375-89, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22798270

ABSTRACT

After spinal cord injury, the disruption of blood-spinal cord barrier by activation of matrix metalloprotease is a critical event leading to infiltration of blood cells, inflammatory responses and neuronal cell death, contributing to permanent neurological disability. Recent evidence indicates that fluoxetine, an anti-depressant drug, is shown to have neuroprotective effects in ischaemic brain injury, but the precise mechanism underlying its protective effects is largely unknown. Here, we show that fluoxetine prevented blood-spinal cord barrier disruption via inhibition of matrix metalloprotease activation after spinal cord injury. After a moderate contusion injury at the T9 level of spinal cord with an infinite horizon impactor in the mouse, fluoxetine (10 mg/kg) was injected intraperitoneally and further administered once a day for indicated time points. Fluoxetine treatment significantly inhibited messenger RNA expression of matrix metalloprotease 2, 9 and 12 after spinal cord injury. By zymography and fluorimetric enzyme activity assay, fluoxetine also significantly reduced matrix metalloprotease 2 and matrix metalloprotease 9 activities after injury. In addition, fluoxetine inhibited nuclear factor kappa B-dependent matrix metalloprotease 9 expression in bEnd.3, a brain endothelial cell line, after oxygen-glucose deprivation/reoxygenation. Fluoxetine also attenuated the loss of tight junction molecules such as zona occludens 1 and occludin after injury in vivo as well as in bEnd.3 cultures. By immunofluorescence staining, fluoxetine prevented the breakdown of the tight junction integrity in endothelial cells of blood vessel after injury. Furthermore, fluoxetine inhibited the messenger RNA expression of chemokines such as Groα, MIP1α and 1ß, and prevented the infiltration of neutrophils and macrophages, and reduced the expression of inflammatory mediators after injury. Finally, fluoxetine attenuated apoptotic cell death and improved locomotor function after injury. Thus, our results indicate that fluoxetine improved functional recovery in part by inhibiting matrix metalloprotease activation and preventing blood-spinal cord barrier disruption after spinal cord injury. Furthermore, our study suggests that fluoxetine may represent a potential therapeutic agent for preserving blood-brain barrier integrity following ischaemic brain injury and spinal cord injury in humans.


Subject(s)
Blood-Brain Barrier/drug effects , Fluoxetine/therapeutic use , Matrix Metalloproteinase Inhibitors , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord/drug effects , Animals , Blood-Brain Barrier/enzymology , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fluoxetine/pharmacology , Male , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Spinal Cord/enzymology , Spinal Cord Injuries/enzymology
15.
Endocrinology ; 153(8): 3815-27, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700771

ABSTRACT

A delayed oligodendrocyte cell death after spinal cord injury (SCI) contributes to chronic demyelination of spared axons, leading to a permanent neurological deficit. Therefore, therapeutic approaches to prevent oligodendrocyte cell death after SCI should be considered. Estrogens are well known to have a broad neuroprotective effect, but the protective effect of estrogens on oligodendrocytes after injury is largely unknown. Here, we demonstrated that 17ß-estradiol attenuates apoptosis of oligodendrocytes by inhibiting RhoA and c-Jun-N-terminal kinase activation after SCI. Estrogen receptor (ER)-α and -ß were expressed in oligodendrocytes of the spinal cord, and 17ß-estradiol treatment significantly inhibited oligodendrocyte cell death at 7 d after injury as compared with vehicle (cyclodextrin) control. 17ß-Estradiol also attenuated caspase-3 and -9 activation at 7 d and reduced the loss of axons from progressive degeneration. In addition, 17ß-estradiol inhibited RhoA and JNK3 activation, which were activated and peaked at 3 and/or 5 d after injury. Furthermore, administration of Rho inhibitor, PEP-1-C3 exoenzyme, inhibited RhoA and JNK3 activation, and decreased phosphorylated c-Jun level at 5 d after injury. Additionally, the attenuation of RhoA and JNK3 activation as well as oligodendrocyte cell death by 17ß-estradiol was reversed by ER antagonist, ICI182780. Our results thus indicate that 17ß-estradiol treatment improves functional recovery after SCI in part by reducing oligodendrocyte cell death via inhibition of RhoA and JNK3 activation, which were ER dependent. Furthermore, improvement of hindlimb motor function by posttreatment of 17ß-estradiol suggests its potential as a therapeutic agent for SCI patients.


Subject(s)
Estradiol/pharmacology , Estradiol/therapeutic use , Mitogen-Activated Protein Kinase 10/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Apoptosis/drug effects , Behavior, Animal , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , In Situ Nick-End Labeling , Male , Mitogen-Activated Protein Kinase 10/genetics , Oligodendroglia/drug effects , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , rhoA GTP-Binding Protein/genetics
16.
Exp Neurol ; 236(2): 268-82, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22634758

ABSTRACT

Acupuncture (AP) is currently used worldwide to relieve pain. However, little is known about its mechanisms of action. We found that after spinal cord injury (SCI), AP inhibited the production of superoxide anion (O(2)·), which acted as a modulator for microglial activation, and the analgesic effect of AP was attributed to its anti-microglial activating action. Direct injection of a ROS scavenger inhibited SCI-induced NP. After contusion injury which induces the below-level neuropathic pain (NP), Shuigou and Yanglingquan acupoints were applied. AP relieved mechanical allodynia and thermal hyperalgesia, while vehicle and simulated AP did not. AP also decreased the proportion of activated microglia, and inhibited both p38MAPK and ERK activation in microglia at the L4-5. Also, the level of prostaglandin E(2) (PGE2), which is produced via ERK signaling and mediates the below-level pain through PGE2 receptor, was reduced by AP. Injection of p38MAPK or ERK inhibitors attenuated NP and decreased PGE2 production. Furthermore, ROS produced after injury-induced p38MAPK and ERK activation in microglia, and mediated mechanical allodynia and thermal hyperalgesia, which were inhibited by AP or a ROS scavenger. AP also inhibited the expression of inflammatory mediators. Therefore, our results suggest that the analgesic effect of AP may be partly mediated by inhibiting ROS-induced microglial activation and inflammatory responses after SCI and provide the possibility that AP can be used effectively as a non-pharmacological intervention for SCI-induced chronic NP in patients.


Subject(s)
Acupuncture Therapy/methods , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Microglia/enzymology , Neuralgia/enzymology , Reactive Oxygen Species/antagonists & inhibitors , Spinal Cord Injuries/enzymology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Chronic Disease , Disease Models, Animal , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Microglia/pathology , Neuralgia/etiology , Neuralgia/therapy , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/administration & dosage , Spinal Cord Injuries/complications , Spinal Cord Injuries/therapy , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Neurochem ; 121(5): 818-29, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22409448

ABSTRACT

The disruption of blood-spinal cord barrier (BSCB) after spinal cord injury (SCI) elicits an intensive local inflammation by the infiltration of blood cells such as neutrophils and macrophages, leading to cell death and permanent neurological disability. SCI activates matrix metalloprotease-9 (MMP-9), which is known to induce BSCB disruption. Here, we examined whether valproic acid (VPA), a histone deacetylase inhibitor, would attenuate BSCB disruption by inhibiting MMP-9 activity, leading to improvement of functional outcome after SCI. After moderate spinal cord contusion injury at T9, VPA (300 mg/kg) were immediately injected subcutaneously and further injected every 12 h for 5 days. Our data show that VPA inhibited MMP-9 activity after injury, and attenuated BSCB permeability and degradation of tight junction molecules such as occludin and ZO-1. In addition, VPA reduced the expression of inflammatory mediators including tumor necrosis factor-α. Furthermore, VPA increased the levels of acetylated histone 3, pAkt, and heat-shock protein 27 and 70, which have anti-apoptotic functions after SCI. Finally, VPA inhibited apoptotic cell death and caspase 3 activation, reduced the lesion volume and improved functional recovery after injury. Thus, our results demonstrated that VPA improves functional recovery by attenuating BSCB disruption via inhibition of MMP-9 activity after SCI.


Subject(s)
Capillary Permeability/drug effects , Matrix Metalloproteinase 9/metabolism , Neuroprotective Agents/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/enzymology , Valproic Acid/pharmacology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Histone Deacetylase Inhibitors/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/pathology
18.
Endocrinology ; 152(6): 2377-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21467197

ABSTRACT

Here, we examined the protective effect of ghrelin on apoptotic cell death induced by hydrogen peroxide (H2O2) in primary oligodendrocyte cultures. Ghrelin receptor, growth hormone secretagogue receptor 1a, was expressed in mature oligodendrocytes. H2O2 (1 mm) treatment induced apoptotic cell death of oligodendrocytes, which was significantly inhibited by ghrelin treatment. Ghrelin also reduced cytochrome c release, and caspase-3 activation increased by H2O2 treatment. Furthermore, the protective effect of ghrelin against H2O2-induced oligodendrocyte cell death was mediated through growth hormone secretagogue receptor 1a. Both ERK and p38MAPK were activated (peaked at 8 h in ERK and 1 h in p38MAPK) by H2O2 treatment, whereas c-Jun N-terminal kinase and Akt were not. Interestingly, ghrelin further increased ERK activation and decreased p38MAPK activation after H2O2 treatment. Next, we tried to elucidate the role of ERK and p38MAPK activation in H2O2-induced apoptotic cell death of oligodendrocytes using pharmacological inhibitors. We found that the inhibition of apoptotic cell death of oligodendrocytes by ghrelin was abolished by ERK inhibitor, PD98059 (20 µM), whereas cell survival was increased by p38MAPK inhibitor, SB203580 (10 µM). These results thus indicate that ghrelin inhibits H2O2 -induced oligodendrocytes cell death in part by increasing ERK activation and decreasing p38MAPK activation, and ghrelin may represent a potential therapeutic agent for protecting oligodendrocytes in central nervous system injuries.


Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Ghrelin/metabolism , Hydrogen Peroxide/toxicity , MAP Kinase Signaling System , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Protective Agents/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , MAP Kinase Signaling System/drug effects , Oligodendroglia/cytology , Oligodendroglia/enzymology , Rats , Rats, Sprague-Dawley
19.
Endocrinology ; 151(8): 3815-26, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20444938

ABSTRACT

Spinal cord injury (SCI) induces massive cell death, leading to permanent neurological disability. No satisfactory treatment is currently available. Ghrelin, a gastric hormone, is known to stimulate GH release from the hypothalamus and pituitary gland. Here, we report that ghrelin administration improves functional recovery after SCI in part by inhibiting apoptosis of neurons and oligodendrocytes. Ghrelin was not detected in normal, uninjured spinal cords, but spinal cord neurons and oligodendrocytes expressed the ghrelin receptor. Ghrelin significantly inhibited apoptotic cell death of neurons and oligodendrocytes, release of mitochondrial cytochrome c, and activation of caspase-3 after moderate contusion SCI. Ghrelin also significantly increased the level of phosphorylated ERK but decreased the level of phosphorylated p38MAPK. In addition, ghrelin increased the level of ERK-dependent brain-derived neurotrophic factor expression and decreased the level of pronerve growth factor expression. Furthermore, the neuroprotective effects of ghrelin were mediated through the ghrelin receptor. Finally, ghrelin significantly improved functional recovery and reduced the size of the lesion volume and the loss of axons and myelin after injury. These results suggest that ghrelin may represent a potential therapeutic agent after acute SCI in humans.


Subject(s)
Apoptosis/drug effects , Ghrelin/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/rehabilitation , Animals , Caspase 3/metabolism , Caspase Inhibitors , Cell Death/drug effects , Cytochromes c/metabolism , Down-Regulation/drug effects , Ghrelin/therapeutic use , Male , Mice , Neurons/drug effects , Neurons/pathology , Oligodendroglia/drug effects , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
20.
Neurobiol Dis ; 39(3): 272-82, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20382225

ABSTRACT

Here, we first demonstrated the neuroprotective effect of acupuncture after SCI. Acupuncture applied at two specific acupoints, Shuigou (GV26) and Yanglingquan (GB34) significantly alleviated apoptotic cell death of neurons and oligodendrocytes, thereby leading to improved functional recovery after SCI. Acupuncture also inhibited caspase-3 activation and reduced the size of lesion cavity and extent of loss of axons. We also found that the activation of both p38 mitogen-activated protein kinase and resident microglia after injury are significantly attenuated by acupuncture. In addition, acupuncture significantly reduced the expression or activation of pro-nerve growth factor, proinflammatory factors such as tumor necrosis factor-alpha, interleukin-1beta, interleukin-6, nitric oxide synthase, cycloxygenase-2, and matrix metalloprotease-9 after SCI. Thus, our results suggest that the neuroprotection by acupuncture may be partly mediated via inhibition of inflammation and microglial activation after SCI and acupuncture can be used as a potential therapeutic tool for treating acute spinal injury in human.


Subject(s)
Acupuncture Therapy , Inflammation/therapy , Motor Activity/physiology , Spinal Cord Injuries/therapy , Spinal Cord/metabolism , Analysis of Variance , Animals , Blotting, Western , Caspase 3/metabolism , Cytokines/metabolism , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Matrix Metalloproteinase 9/metabolism , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Recovery of Function , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL