Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 393
1.
Cell Death Dis ; 12(7): 630, 2021 06 18.
Article En | MEDLINE | ID: mdl-34145219

Mitophagy alleviates neuronal damage after cerebral ischemia by selectively removing dysfunctional mitochondria. Phosphatase and tensin homolog (PTEN) induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy is the most well-known type of mitophagy. However, little is known about the role of PINK1/Parkin-mediated mitophagy in ischemic tolerance induced by hypoxic postconditioning (HPC) with 8% O2 against transient global cerebral ischemia (tGCI). Hence, we aimed to test the hypothesis that HPC-mediated PINK1/Parkin-induced mitochondrial ubiquitination and promotes mitophagy, thus exerting neuroprotection in the hippocampal CA1 subregion against tGCI. We found that mitochondrial clearance was disturbed at the late phase of reperfusion after tGCI, which was reversed by HPC, as evidenced by the reduction of the translocase of outer mitochondrial membrane 20 homologs (TOMM20), translocase of inner mitochondrial membrane 23 (TIMM23) and heat shock protein 60 (HSP60) in CA1 after HPC. In addition, HPC further increased the ratio of LC3II/I in mitochondrial fraction and promoted the formation of mitophagosomes in CA1 neurons after tGCI. The administration of lysosome inhibitor chloroquine (CQ) intraperitoneally or mitophagy inhibitor (Mdivi-1) intracerebroventricularly abrogated HPC-induced mitochondrial turnover and neuroprotection in CA1 after tGCI. We also found that HPC activated PINK1/Parkin pathway after tGCI, as shown by the augment of mitochondrial PINK1 and Parkin and the promotion of mitochondrial ubiquitination in CA1. In addition, PINK1 or Parkin knockdown with small-interfering RNA (siRNA) suppressed the activation of PINK1/Parkin pathway and hampered mitochondrial clearance and attenuated neuroprotection induced by HPC, whereas PINK1 overexpression promoted PINK1/Parkin-mediated mitophagy and ameliorated neuronal damage in CA1 after tGCI. Taken together, the new finding in this study is that HPC-induced neuroprotection against tGCI through promoting mitophagy mediated by PINK1/Parkin-dependent pathway.


CA1 Region, Hippocampal/enzymology , Hypoxia/enzymology , Ischemic Attack, Transient/enzymology , Mitochondria/enzymology , Mitophagy , Neurons/enzymology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , CA1 Region, Hippocampal/ultrastructure , Disease Models, Animal , Hypoxia/genetics , Hypoxia/pathology , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/pathology , Male , Mitochondria/genetics , Mitochondria/ultrastructure , Neurons/ultrastructure , Protein Kinases/genetics , Protein Transport , Rats, Wistar , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
Microvasc Res ; 129: 103973, 2020 05.
Article En | MEDLINE | ID: mdl-31891716

Ischemic stroke represents a major cause of adult death and severe neurological disability worldwide. Reperfusion following brain ischemia produces an inflammatory cascade that increases brain damage. In this context, matrix metalloproteinases (MMPs) play an important role as pro-inflammatory mediators. The MMP 2 up-regulation seems to promote matrix degradation, blood-brain barrier (BBB) disruption and facilitates the influx of peripheral inflammatory cells to the brain after stroke. However, there are not studies about MMP-1 in this condition. The aim of this study is to evaluate the association of brain damage, inflammatory response and the immunostaining profile of matrix metalloproteinases 1 and 2 after transient global cerebral ischemia. Mice were submitted to bilateral common carotid arterial occlusion (BCCAo) during 25 min. After three days of reperfusion, the neurological deficit score was evaluated and the animals were euthanized. Brain samples were collected in order to analyze the histopathological damage, MMPs 1 and 2 immunostaining and cytokines and chemokines levels. Ischemic group showed neurological deficits associated with brain lesions, characterized by necrotic core and penumbra zone three days after reperfusion. Higher brain immunostaining of MMP-1 and MMP-2 was observed in BCCAo samples than in sham samples. Ischemic group also exhibited increased brain levels of the cytokines tumoral necrosis factor (TNF) and interleukin 1ß (IL-1ß), chemokine (C-X-C motif) ligand 1 (CXCL1), and chemokine (C-C motif) ligand 5 (CCL5) in comparison to sham group. Our results suggest that the MMP-1 and MMP-2 raise, associated with the up-regulation of inflammatory mediators, contributes to brain damage and neurological deficits after global brain ischemia followed by three days of reperfusion in mice.


Brain/enzymology , Cytokines/metabolism , Ischemic Attack, Transient/enzymology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 2/metabolism , Animals , Brain/pathology , Brain/physiopathology , Chemokine CCL5/metabolism , Chemokine CXCL1/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Male , Mice, Inbred C57BL , Necrosis , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
3.
Cell Death Dis ; 9(6): 635, 2018 05 25.
Article En | MEDLINE | ID: mdl-29802248

Hypoxic postconditioning (HPC) is an innovative neuroprotective strategy with cytoprotective effects on the hippocampal neurons against transient global cerebral ischemia (tGCI) in adult rats. However, its molecular mechanisms have not yet been adequately elucidated. Neuroglobin (Ngb) is an endogenous neuroprotectant with hypoxia-inducible property, and its role in experimental stroke has been increasingly attractive. Hence, the purpose of this study is to explore the involvement of Ngb in HPC-mediated neuroprotection and to further investigate its underlying molecular mechanism. We found that HPC increased Ngb expression in CA1 subregion after tGCI. Also, the inhibition of Ngb expression with Ngb antisense oligodeoxynucleotide (AS-ODNs) eliminated the neuroprotective effect mediated by HPC, whereas overexpression of Ngb ameliorated neuronal damage in CA1 after tGCI, indicating that HPC conferred neuroprotective effects via upregulation of Ngb. We further showed that HPC increased the membranous level of Na+/K+ ATPases ß1 subunit (Atp1b1) in CA1 after tGCI. Furthermore, we demonstrated that Ngb upregulation in CA1 after HPC maintained the membranous level of Atp1b1 through Ngb-Atp1b1 interaction and reduced the glutathionylation of membranous Atp1b1 via suppression of reactive oxygen species (ROS), ultimately preserving the activity of NKA. Taken together, these data indicate that Ngb is involved in the neuroprotection of HPC against tGCI via maintenance of NKA activity in the hippocampal CA1.


Hypoxia/pathology , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Neuroglobin/metabolism , Neuroprotection , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , CA1 Region, Hippocampal/pathology , Glutathione/metabolism , Male , Neurons/metabolism , Neurons/pathology , Rats, Wistar , Reactive Oxygen Species/metabolism , Up-Regulation
4.
Orphanet J Rare Dis ; 11(1): 54, 2016 05 04.
Article En | MEDLINE | ID: mdl-27142856

BACKGROUND: Fabry disease (FD) is an X-linked multisystemic disorder with a heterogeneous phenotype. Especially atypical or late-onset type 2 phenotypes present a therapeutical dilemma. METHODS: To determine the clinical impact of the alpha-Galactosidase A (GLA) p.A143T/ c.427G > A variation, we retrospectively analyzed 25 p.A143T patients in comparison to 58 FD patients with other missense mutations. RESULTS: p.A143T patients suffering from stroke/ transient ischemic attacks had slightly decreased residual GLA activities, and/or increased lyso-Gb3 levels, suspecting FD. However, most male p.A143T patients presented with significant residual GLA activity (~50 % of reference), which was associated with normal lyso-Gb3 levels. Additionally, p.A143T patients showed less severe FD-typical symptoms and absent FD-typical renal and cardiac involvement in comparison to FD patients with other missense mutations. Two tested female p.A143T patients with stroke/TIA did not show skewed X chromosome inactivation. No accumulation of neurologic events in family members of p.A143T patients with stroke/transient ischemic attacks was observed. CONCLUSIONS: We conclude that GLA p.A143T seems to be most likely a neutral variant or a possible modifier instead of a disease-causing mutation. Therefore, we suggest that p.A143T patients with stroke/transient ischemic attacks of unknown etiology should be further evaluated, since the diagnosis of FD is not probable and subsequent ERT or chaperone treatment should not be an unreflected option.


Fabry Disease/enzymology , Mutation/genetics , alpha-Galactosidase/genetics , Adult , Fabry Disease/genetics , Female , Genotype , Humans , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/genetics , Male , Middle Aged , Mutation, Missense/genetics , Retrospective Studies , Stroke/enzymology , Stroke/genetics
5.
Neurobiol Dis ; 91: 236-46, 2016 07.
Article En | MEDLINE | ID: mdl-27001146

New neuroreparative and neuroprotective therapies are being sought to treat stroke patients. One approach is the remodeling of extracellular matrix, which participates in both brain injury and neurovascular repair when matrix metalloproteinases (MMPs) are thought to be key players. Our aim was to investigate the role of MMP-13 (collagenase-3) in the acute (24h and 3days) and delayed (2weeks) phases of stroke. Permanent and transient cerebral ischemia models involving the cortex were induced in MMP-13 knock-out (KO) and wild-type (WT) mice. In the transient model, MMP-13 deficiency reduced the amount of TTC-stained infarct tissue, reduced hemorrhagic events and improved functional outcomes (p<0.01). At two weeks, normal neuroblast (DCX+) migration from the subventricular zone toward the peri-infarct area was observed. However, MMP-13 deficiency significantly reduced the number of newborn neuroblasts (DCX+/BrdU+) in the cortical peri-infarct area (p<0.01). This result occurred in parallel with aberrant cortical vascular remodeling: post-stroke peri-infarct vessel density increased in the WT mice (p<0.01) but this increase was blocked in the MMP-13 KO mice. Prior to these vascular alterations, the levels of pro-angiogenic factors, including G-CSF, VEGF-A and angiopoietin-2, were lower in the ischemic cortex of MMP-13 KO mice than in WT mice (p<0.05). In vitro, gene-silencing of MMP-13 in endothelial progenitor cells (EPCs) confirmed the reduced ability of these cells to build tubulogenic networks in Matrigel™ substrate. Together, our results indicate that MMP-13 is a central protease in infarct development and cortical remodeling during post-stroke neurorepair, which is critical for optimal angiogenic and neurogenic responses.


Ischemic Attack, Transient/enzymology , Matrix Metalloproteinase 13/metabolism , Neuroprotection/physiology , Stroke/enzymology , Animals , Disease Models, Animal , Doublecortin Protein , Infarction, Middle Cerebral Artery/metabolism , Matrix Metalloproteinase 13/genetics , Mice, Knockout , Neurogenesis/physiology
6.
Anesth Analg ; 122(5): 1377-84, 2016 May.
Article En | MEDLINE | ID: mdl-26859875

BACKGROUND: Phosphoinositide-3-kinase (PI3K)/protein kinase B (Akt) pathway activation may promote neuronal survival via neuroprotection during inflammation after cerebral ischemia. In this study, we investigated whether IV pretreatment with emulsified isoflurane (EI) could decrease ischemic brain injury related to the PI3K/Akt pathway. METHODS: Male Sprague-Dawley rats received different doses of IV EI (1, 2, 4, or 8 mL/kg/h) or Intralipid (8 mL/kg/h) for 30 minutes (n = 6-12 per group), followed by middle cerebral artery occlusion (MCAO) for 100 minutes to induce transient focal ischemia. The neurologic score and infarct volume were measured 48 hours after MCAO. Immunostaining, Western blot analysis, and an enzyme-linked immunosorbent assay were used to assess EI effects on the cell inflammatory response, high-mobility group box-1 release, and phosphorylated Akt (expression. LY294002, a PI3K inhibitor, was also infused into the ventricular space before EI to determine the effect of EI. RESULTS: Four milliliters per kilogram per hour of EI reduced the infarct size (21.08 ± 11.24 vs 37.09 ± 10.46, P = 0.006), improved neurologic scores after MCAO (1.13 ± 0.48 vs 1.95 ± 0.65, P = 0.015), significantly reinforced neuronal survival (982.7 ± 364.4 vs 439.8 ± 278.4, P = 0.036), and inhibited CD68 macrophage/macroglial infiltration in the ischemic core (188.2 ± 49.1 vs 282 ± 49.4, P = 0.018) compared with the vehicle group. In the EI pretreatment group, the serum high-mobility group box-1 concentration (3.62 ± 0.72 vs 5.73 ± 0.65, P < 0.001) was decreased, and the cerebral phosphorylated Akt level (50.33 ± 4.73 vs 37.5 ± 3.11, P = 0.007) was increased at 48 hours, which was inhibited by LY294002 compared with the vehicle group (5.31 ± 0.72 vs 5.73 ± 0.65, P = 0.216; 43.00 ± 4.84 vs 37.5 ± 3.11, P = 0.091). CONCLUSIONS: These findings suggest that EI pretreatment protects against ischemic brain injury via the inhibition of cerebral inflammation and is associated with the PI3K-Akt pathway in rats with MCAO. This drug may be a novel therapeutic agent for patients after stroke.


Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/prevention & control , Isoflurane/pharmacology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Administration, Intravenous , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Brain/enzymology , Brain/pathology , Chemistry, Pharmaceutical , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Emulsions , HMGB1 Protein/blood , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Isoflurane/administration & dosage , Isoflurane/chemistry , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Time Factors
7.
Neurology ; 85(18): 1585-91, 2015 Nov 03.
Article En | MEDLINE | ID: mdl-26311748

OBJECTIVE: To determine the association of lipoprotein-associated phospholipase A2 (Lp-PLA2) measured in the acute period and the short-term risk of recurrent vascular events in patients with TIA or minor stroke. METHODS: We measured Lp-PLA2 activity (Lp-PLA2-A) in a subset of 3,201 participants enrolled in the CHANCE (Clopidogrel in High-Risk Patients with Acute Non-disabling Cerebrovascular Events) trial. Participants with TIA or minor stroke were enrolled within 24 hours of symptom onset and randomized to single or dual antiplatelet therapy. In the current analysis, the primary outcome was defined as the composite of ischemic stroke, myocardial infarction, or death within 90 days. RESULTS: The composite endpoint occurred in 299 of 3,021 participants (9.9%). The population average Lp-PLA2-A level was 209 ± 59 nmol/min/mL (95% confidence interval [CI] 207-211). Older age, male sex, and current smoking were associated with higher Lp-PLA2-A levels. Lp-PLA2-A was significantly associated with the primary endpoint (adjusted hazard ratio 1.07, 95% CI 1.01-1.13 for every 30 nmol/min/mL increase). Similar results were seen for ischemic stroke alone. Adjustment for low-density lipoprotein cholesterol attenuated the association between Lp-PLA2-A and the primary endpoint (adjusted hazard ratio 1.04, 95% CI 0.97-1.11 for every 30 nmol/min/mL increase). CONCLUSIONS: Higher levels of Lp-PLA2-A in the acute period are associated with increased short-term risk of recurrent vascular events.


1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Ischemic Attack, Transient/enzymology , Myocardial Infarction/enzymology , Stroke/enzymology , Aged , Biomarkers , Cohort Studies , Female , Humans , Ischemic Attack, Transient/drug therapy , Male , Middle Aged , Myocardial Infarction/mortality , Platelet Aggregation Inhibitors/therapeutic use , Proportional Hazards Models , Prospective Studies , Recurrence , Risk Factors , Secondary Prevention , Severity of Illness Index , Stroke/drug therapy , Stroke/mortality
8.
Mol Med Rep ; 12(2): 2555-62, 2015 Aug.
Article En | MEDLINE | ID: mdl-25955690

The thioredoxin (Trx) and peroxiredoxin (Prx) redox system is associated with neuronal damage and neuroprotective effects via the regulation of oxidative stress in brain ischemia. In the present study, ischemia-induced changes in the protein expression levels of Trx2 and Prx3 in the stratum pyramidale (SP) of the hippocampal CA1 region were investigated in adult and aged gerbils, subjected to 5 min of transient global cerebral ischemia, using immunohistochemistry and western blot analysis. In the adult ischemia-group, minimal Trx2 immunoreactivity was detected in the SP 2 days after ischemia-reperfusion. In the aged animals, the Trx2 immunoreactivity in the sham-group was marginally lower compared with that in the adult sham-group. In the aged ischemia-group, Trx2 immunoreactivity in the SP was significantly higher 1, 2 and 4 days post-ischemia, compared with that in the adult ischemia-group and, in the 5 days post-ischemia group, Trx2 immunoreactivity was significantly decreased in the SP. Prx3 immunoreactivity in the SP of the adult ischemia-group was significantly decreased from 4 days after ischemia-reperfusion. In the aged animals, Prx3 immunoreactivity in the sham-group was also marginally lower compared with that in the adult sham-group. Prx3 immunoreactivity in the aged ischemia-group was also significantly higher 1, 2 and 4 days post-ischemia, compared with the adult ischemia-group; however, the Prx3 immunoreactivity was significantly decreased 5 days post-ischemia. The western blot analyses revealed that the pattern of changes in the protein levels of Trx2 and Prx3 in the adult and aged hippocampal CA1 region following ischemic damage were similar to the results obtained in the immunohistochemical data. These findings indicated that cerebral ischemia lead to different protein expression levels of Trx2 and Prx3 in the hippocampal CA1 region between adult and aged gerbils, and these differences may be associated with more delayed neuronal death in the aged gerbil hippocampus following transient global cerebral ischemia.


Aging/metabolism , CA1 Region, Hippocampal/enzymology , Ischemic Attack, Transient/genetics , Peroxiredoxin III/genetics , Pyramidal Cells/enzymology , Thioredoxins/genetics , Aging/pathology , Animals , CA1 Region, Hippocampal/pathology , Gene Expression Regulation , Gerbillinae , Immunohistochemistry , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Oxidative Stress , Peroxiredoxin III/metabolism , Pyramidal Cells/pathology , Signal Transduction , Thioredoxins/metabolism
9.
Stroke ; 46(2): 492-9, 2015 Feb.
Article En | MEDLINE | ID: mdl-25563647

BACKGROUND AND PURPOSE: Ischemic stroke induces metabolic disarray. A central regulatory site, pyruvate dehydrogeanse complex (PDHC) sits at the cross-roads of 2 fundamental metabolic pathways: aerobic and anaerobic. In this study, we combined ethanol (EtOH) and normobaric oxygen (NBO) to develop a novel treatment to modulate PDHC and its regulatory proteins, namely pyruvate dehydrogenase phosphatase and pyruvate dehydrogenase kinase, leading to improved metabolism and reduced oxidative damage. METHODS: Sprague-Dawley rats were subjected to transient (2, 3, or 4 hours) middle cerebral artery occlusion followed by 3- or 24-hour reperfusion, or permanent (28 hours) middle cerebral artery occlusion without reperfusion. At 2 hours after the onset of ischemia, rats received either an intraperitoneal injection of saline, 1 dose of EtOH (1.5 g/kg) for 2- and 3-hour middle cerebral artery occlusion, 2 doses of EtOH (1.5 g/kg followed by 1.0 g/kg in 2 hours) in 4 hours or permanent middle cerebral artery occlusion, and EtOH+95% NBO (at 2 hours after the onset of ischemia for 6 hours) in permanent stroke. Infarct volumes and neurological deficits were examined. Oxidative metabolism and stress were determined by measuring ADP/ATP ratio and reactive oxygen species levels. Protein levels of PDHC, pyruvate dehydrogenase kinase, and pyruvate dehydrogenase phosphatase were assessed. RESULTS: EtOH induced dose-dependent neuroprotection in transient ischemia. Compared to EtOH or NBO alone, NBO+EtOH produced the best outcomes in permanent ischemia. These therapies improved brain oxidative metabolism by decreasing ADP/ATP ratios and reactive oxygen species levels, in association with significantly raised levels of PDHC and pyruvate dehydrogenase phosphatase, as well as decreased pyruvate dehydrogenase kinase. CONCLUSIONS: Both EtOH and EtOH+NBO treatments conferred neuroprotection in severe stroke by affecting brain metabolism. The treatment may modulate the damaging cascade of metabolic events by bringing the PDHC activity back to normal metabolic levels.


Ethanol/therapeutic use , Ischemic Attack, Transient/therapy , Oxygen Inhalation Therapy/methods , Pyruvate Dehydrogenase Complex/physiology , Severity of Illness Index , Stroke/therapy , Animals , Ischemic Attack, Transient/enzymology , Male , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Stroke/enzymology
10.
Brain Res ; 1587: 127-32, 2014 Oct 31.
Article En | MEDLINE | ID: mdl-25192645

Autophagy is a degradative mechanism for cellular proteins and organelles, but its role in the nervous system is still not clear. In the present study, we found that exercise pretreatment and p38 inhibition had influence on autophagic process after cerebral ischemia, contributing to their neuroprotective effects. We examined the levels of p62 and phosphorylated ERK1/2 as an autophagic marker and cell-survival marker respectively after cerebral ischemic injury. The brain infarction volume after ischemia was measured as well. Both treadmill training pretreatment and p38 inhibition decreased the degradation of p62, promoted the phosphorylation of ERK1/2, and alleviated the brain infarction, indicating that these treatments could provide neuroprotection in cerebral ischemic injury via autophagy suppression.


Autophagy/physiology , Brain Ischemia/pathology , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Physical Conditioning, Animal/physiology , Reperfusion Injury/prevention & control , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Apoptosis Regulatory Proteins , Brain Ischemia/metabolism , Heat-Shock Proteins/metabolism , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/pathology , Male , Nerve Tissue Proteins/physiology , Neurons/enzymology , Neurons/pathology , Phosphorylation , Protein Processing, Post-Translational , Random Allocation , Rats , Rats, Sprague-Dawley , Running , Sequestosome-1 Protein , p38 Mitogen-Activated Protein Kinases/physiology
11.
Eur J Pharmacol ; 728: 93-9, 2014 Apr 05.
Article En | MEDLINE | ID: mdl-24512724

Hyperbaric oxygen (HBO) is emerging as a therapy for brain ischemia, although its benefits are still debated. The present study aimed to investigate the effect of HBO on brain damage in a rat model of transient focal cerebral ischemia and its underlying mechanism of action. Male Wistar rats, which had suffered 1.5h of transient middle cerebral artery occlusion (tMCAO) and had a Longa's neuron score of 3, were given pure oxygen at 3.0 atm absolute, for 60 min after the third hour of reperfusion. After 24h of reperfusion, rat brains were removed and studied. 2,3,5-triphenyltetrazolium chloride (TTC) and hematoxylin and eosin staining revealed that the infarct ratio in the HBO group increased remarkably when compared with the MCAO group. Up-regulation of extracellular signal-regulated kinase 1/2 (ERK1/2) activation was detected in the HBO group because of reactive oxygen species (ROS) generation. Autophagy appeared to be obstructed in the HBO group. Administration of the ERK1/2 inhibitor U0126 decreased the infarct ratio and improved protein clearance by autophagy in the HBO group. Collectively, these results suggest that HBO enlarges the area of brain damage via reactive oxygen species-induced activation of ERK1/2, which interrupts autophagy flux.


Autophagy/physiology , Brain/pathology , Hyperbaric Oxygenation/adverse effects , Infarction, Middle Cerebral Artery/complications , Ischemic Attack, Transient/therapy , MAP Kinase Signaling System/physiology , Animals , Brain/enzymology , Butadienes/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hyperbaric Oxygenation/methods , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/etiology , Ischemic Attack, Transient/pathology , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Up-Regulation
12.
Mol Neurobiol ; 49(3): 1338-49, 2014 Jun.
Article En | MEDLINE | ID: mdl-24352802

Postconditioning has regenerated interest as a mechanical intervention against cerebral ischemia/reperfusion injury, but its molecular mechanisms remain unknown. We previously reported that hypoxic postconditioning (HPC) ameliorated neuronal death induced by transient global cerebral ischemia (tGCI) in hippocampal CA1 subregion of adult rats. This study tested the hypothesis that p38-mitogen-activated protein kinase (p38 MAPK)/mitogen- and stress-response kinase 1 (MSK1) signaling pathway plays a role in the HPC-induced neuroprotection. Male Wistar rats were subjected to 10 min ischemia induced by applying the four-vessel occlusion method. HPC with 120 min was applied at 24 h after reperfusion. Immunohistochemistry and Western blot were used to detect the expression of phosphorylation of p38 MAPK and MSK1, as well as cleaved caspase-3. We found that HPC induced a significant increase of phosphorylated p38 MAPK and MSK1 in neurons of hippocampal CA1 region and a significant decrease in glial cells after tGCI as well. Furthermore, HPC attenuated caspase-3 cleavation triggered by tGCI in CA1 region. Moreover, p38 MAPK inhibition by SB203580 significantly decreased the phosphorylation of MSK1, increased cleaved caspase-3 expression, and abolished the neuroprotection of HPC. These findings suggested that p38 MAPK/MSK1 signaling axis contributed to HPC-mediated neuroprotection against tGCI, at least in part, by regulating the activation of caspase-3.


Hypoxia/enzymology , Ischemic Attack, Transient/enzymology , Ischemic Postconditioning , MAP Kinase Signaling System/physiology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Age Factors , Animals , Hypoxia/pathology , Ischemic Attack, Transient/prevention & control , Ischemic Postconditioning/methods , Male , Rats , Rats, Wistar
13.
Neurochem Res ; 38(12): 2640-9, 2013 Dec.
Article En | MEDLINE | ID: mdl-24146201

Glucose is a main energy source for normal brain functions. Glucokinase (GK) plays an important role in glucose metabolism as a glucose sensor, and GK activity is modulated by glucokinase regulatory protein (GKRP). In this study, we examined the changes of GK and GKRP immunoreactivities in the gerbil hippocampus after 5 min of transient global cerebral ischemia. In the sham-operated-group, GK and GKRP immunoreactivities were easily detected in the pyramidal neurons of the stratum pyramidale of the hippocampus. GK and GKRP immunoreactivities in the pyramidal neurons were distinctively decreased in the hippocampal CA1 region (CA), not CA2/3, 3 days after ischemia-reperfusion (I-R). Five days after I-R, GK and GKRP immunoreactivities were hardly detected in the CA1, not CA2/3, pyramidal neurons; however, at this point in time, GK and GKRP immunoreactivities were newly expressed in astrocytes, not microglia, in the ischemic CA1. In brief, GK and GKRP immunoreactivities are changed in pyramidal neurons and newly expressed in astrocytes in the ischemic CA1 after transient cerebral ischemia. These indicate that changes of GK and GKRP expression may be related to the ischemia-induced neuronal damage/death.


Carrier Proteins/metabolism , Glucokinase/metabolism , Hippocampus/metabolism , Ischemic Attack, Transient/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique , Gerbillinae , Hippocampus/enzymology , Ischemic Attack, Transient/enzymology , Male
14.
Thromb Res ; 132(2): 211-6, 2013 Aug.
Article En | MEDLINE | ID: mdl-23830213

INTRODUCTION: The pharmacological target of aspirin is the inhibition of cyclooxygenase-1 (COX1) and thromboxane-A2 (TX) synthesis. Very few data are available on TX assessment in patients with stroke. We studied platelet TX synthesis, COX1-independent platelet reactivity, the influence of platelet-erythrocyte interactions and the potential association between platelet responses and the severity of stroke, evaluated with a clinical score (NIHSS). MATERIAL AND METHODS: We examined 157 aspirin-treated patients with acute stroke or TIA, 128 aspirin-free and 15 aspirin-treated healthy subjects (HS). Collagen-induced TX, platelet recruitment in whole blood and platelets ± erythrocytes (haematocrit 40%) were assessed in patients on daily-aspirin within three days from onset. Arachidonic-acid-, ADP-, thrombin-receptor activating peptide TRAP-, and collagen-induced aggregation were also evaluated. RESULTS: Partial TX inhibition (<95% inhibition vs aspirin-free controls) was observed in 13% of patients. This was associated with marked increases in COX1-dependent responses (arachidonic-acid- and collagen-induced aggregation and platelet recruitment; P<0.0001) but not with differences in ADP- or TRAP-induced aggregation. Partial TX inhibition was independently associated with severe stroke (NIHSS ≥ 12) at both admission (P<0.05) and discharge (P<0.05). Among patients with fully blocked TX, those with elevated COX1-independent platelet reactivity (mean+2SD of aspirin-treated HS) were most likely to suffer severe stroke (P<0.05). Platelet-erythrocyte interactions enhanced platelet reactivity in these patients by COX1-dependent and -independent mechanisms (P<0.0001). CONCLUSIONS: TX inhibition by aspirin varied across patients. Partial TX inhibition and COX1-independent platelet hyperfunction were associated with more-severe stroke.


Aspirin/therapeutic use , Cyclooxygenase 1/biosynthesis , Ischemic Attack, Transient/blood , Ischemic Attack, Transient/drug therapy , Stroke/blood , Stroke/drug therapy , Thromboxane A2/biosynthesis , Acute Disease , Aged , Case-Control Studies , Cyclooxygenase 1/blood , Cyclooxygenase Inhibitors/therapeutic use , Female , Humans , Ischemic Attack, Transient/enzymology , Male , Platelet Aggregation/drug effects , Stroke/enzymology , Thromboxane A2/blood
15.
J Cereb Blood Flow Metab ; 33(10): 1532-9, 2013 Oct.
Article En | MEDLINE | ID: mdl-23820645

Endothelial nitric oxide synthase (eNOS) dysfunction is related to secondary injury and lesion expansion after cerebral ischemia. To date, there are few reports about postischemic alterations in the eNOS regulatory system. The purpose of the present study was to clarify eNOS expression, Ser1177 phosphorylation, and monomer formation after cerebral ischemia. Male Wistar rats were subjected to transient focal cerebral ischemia. Endothelial nitric oxide synthase messenger RNA (mRNA) and protein expression increased ≈ 8-fold in the ischemic lesion. In the middle cerebral artery core, eNOS-Ser1177 phosphorylation increased 6 hours after ischemia; however, there was an approximately 90% decrease in eNOS-Ser1177 phosphorylation observed 24 hours after ischemia that continued until at least 7 days after ischemia. Endothelial nitric oxide synthase monomer formation also increased 24 and 48 hours after ischemia (P<0.05), and protein nitration progressed in parallel with monomerization. To assess the effect of a neuroprotective agent on eNOS dysfunction, we evaluated the effect of fasudil, a Rho-kinase inhibitor, on eNOS phosphorylation and dimerization. Postischemic treatment with fasudil suppressed lesion expansion and dephosphorylation and monomer formation of eNOS. In conclusion, functional deterioration of eNOS progressed after cerebral ischemia. Rho-kinase inhibitors can reduce ischemic lesion expansion as well as eNOS dysfunction in the ischemic brain.


Endothelium, Vascular/enzymology , Ischemic Attack, Transient/enzymology , Nitric Oxide Synthase Type III/biosynthesis , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Blotting, Western , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/pathology , Immunohistochemistry , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Male , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Multimerization , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
16.
J Ethnopharmacol ; 149(1): 270-80, 2013 Aug 26.
Article En | MEDLINE | ID: mdl-23811213

ETHNOPHARMACOLOGICAL RELEVANCE: Huang-Lian-Jie-Du-Decotion (HLJDD, Hwangryun-Hae-Dok-Decotion in Japan), an ancient antipyretic and detoxifying traditional Chinese medicine formula, was reported to have protective effect on ischemic stroke. AIM OF THE RESEARCH: To investigate the therapeutic effect of HLJDD on ischemic stroke and explore its mode of action. MATERIAL AND METHODS: A model of ischemic stroke in the rat was established after transient middle cerebral artery occlusion (MCAO) followed by reperfusion. Rats were assigned randomly to groups of control, sham, transient ischemia/reperfusion (I/R), and three treatment groups by HLJDD at 2.5, 5.0, 10.0mg/kg. The neurological deficit, the cerebral infarct size, morphology abnormality, biochemical parameters were examined, and the levels of relevant proteins were determined by immunoblotting analysis to evaluate the protective effects of HLJDD on ischemic stroke and explore the underlying mechanism. RESULTS: Compared with I/R group, HLJDD significantly ameliorated neurological deficit and histopathology changes, decreased infarct area, and restored the levels of biochemical indicators including nitric oxide (NO), malondialdehyde (MDA), glutathione (GSH), glutathione disulfide (GSSG), total superoxide dismutase (T-SOD), Cu/Zn-SOD, Mn-SOD and glutathione peroxidase (GSH-PX). HLJDD also notably elevated the levels of microtubule-associated protein 1 light chain 3 (LC3), Beclin-1, and other autophagy related genes (Atgs), promoted the activation of extracellular signal-regulated kinases (ERK), protein kinase B (Akt), 3-phosphoinositide-dependent kinase (PDK1), and inhibited the activation of mammalian target of rapamycin (mTOR), c-Jun N-terminal protein kinases (JNK), p38, phosphatase and tensin homolog (PTEN). CONCLUSION: HLJDD showed neuroprotective effects on ischemic stroke, at least in part to the induced protective autophagy via the regulation of mitogen-activated protein kinase (MAPK) signals. This Akt-independent protective autophagy is favorable in the treatment of stroke, avoiding unfavorable side-effects associated with the inactivation of Akt. The efficacy of HLJDD on ischemic stroke and its safety warranted by its long-term clinical use in traditional Chinese medicine favored further study to develop HLJDD as an effective therapeutic agent to treat ischemic stroke.


Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Autophagy/drug effects , Drugs, Chinese Herbal/therapeutic use , Ischemic Attack, Transient/prevention & control , Mitogen-Activated Protein Kinases/metabolism , Reperfusion Injury/prevention & control , TOR Serine-Threonine Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/isolation & purification , Ethnopharmacology , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Molecular Structure , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction
17.
Atherosclerosis ; 228(2): 508-14, 2013 Jun.
Article En | MEDLINE | ID: mdl-23608248

OBJECTIVE: To investigate the association between the liver enzymes γ-glutamyltransferase (GGT) and (alanine aminotransferase) ALT and risk of stroke, its subtypes including TIA as well as fatal and non-fatal events. METHODS: A case-cohort study within the European Prospective Investigation into Cancer and Nutrition-Potsdam Study comprising 27548 middle-aged subjects was designed. GGT and ALT were measured in plasma of 353 individuals who developed a stroke and in 2110 individuals who remained free of cardiovascular events during a mean follow-up of 8.2 ± 2.2 years. Cox proportional-hazard models were applied to evaluate the association between liver enzymes and stroke risk. RESULTS: After adjustment for established clinical and lifestyle factors, a 1 unit change in naturally logged GGT was related to a 1.20 (95%CI: 1.03-1.40) increased stroke risk. Risk estimates did not significantly differ between fatal (Relative Risk (RR) = 1.35, 95%CI: 1.14-1.61) and non-fatal events (RR = 1.15; 95%CI: 0.97-1.36). ALT was not associated with overall stroke risk (RR = 0.95; 95%CI: 0.71-1.26). However, in subtype analyses we observed in multivariable adjusted models a significant increased risk of hemorrhagic stroke (RR = 2.00; 95% CI: 1.01-3.96), but decreased risk of ischemic stroke (RR = 0.66; 95%CI: 0.44-0.998). CONCLUSIONS: Our data provide further evidence for a link between GGT, but not ALT and overall stroke suggesting that these biomarkers are involved in different pathways of disease development. Further studies are needed to clarify the putative relationships between ALT and subtypes of stroke.


Alanine Transaminase/blood , Ischemic Attack, Transient/etiology , Liver/enzymology , Stroke/etiology , gamma-Glutamyltransferase/blood , Adult , Age Factors , Aged , Biomarkers/blood , Case-Control Studies , Female , Germany , Humans , Ischemic Attack, Transient/blood , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/mortality , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Prognosis , Proportional Hazards Models , Prospective Studies , Risk Assessment , Risk Factors , Stroke/blood , Stroke/enzymology , Stroke/mortality , Time Factors
18.
Neurol Res ; 35(4): 395-405, 2013 May.
Article En | MEDLINE | ID: mdl-23540408

OBJECTIVES: Adenosine monophosphate-activated kinase (AMPK) is an energy-specific sensor within the central nervous system. In this study, we observed AMPK and its phosphorylated form (pAMPK) in the hippocampal CA1 region after 5 minutes of transient forebrain ischemia. In addition, we also investigated the effects of Compound C, an AMPK inhibitor, against ischemic damage in gerbils. METHODS: Adenosine monophosphate-activated kinase and pAMPK immunoreactivity was observed in the hippocampal CA1 region at various time points after ischemia and Compound C was intraperitoneally administered to gerbils immediately after reperfusion and the animals were sacrificed at 5 days after ischemia/reperfusion. RESULTS: Adenosine monophosphate-activated kinase immunoreactivity was transiently increased in the hippocampal CA1 region 1-2 days after ischemia/reperfusion, while AMPK immunoreactivity was almost undetectable in the stratum pyramidale of the CA1 region 4-7 days after ischemia/reperfusion. The administration of Compound C caused a dose-dependent decrease in the ischemia-induced hyperactive behavior, the depletion of ATP, and lactate accumulation in the hippocampal CA1 region within 24 hours after ischemia/reperfusion. In addition, the administration of Compound C decreased reactive gliosis (astrocytes and microglia) and increased the number of cresyl violet-positive neurons when compared to the vehicle-treated group at 5 days post-ischemia/reperfusion. CONCLUSION: These results suggest that AMPK is transiently phosphorylated following forebrain ischemia in the hippocampal CA1 region and inhibition of AMPK has neuroprotective effects against ischemic damage through the reduction of ATP depletion and lactate accumulation in the hippocampal CA1 region.


Adenylate Kinase/metabolism , CA1 Region, Hippocampal/enzymology , Ischemic Attack, Transient/enzymology , Animals , CA1 Region, Hippocampal/drug effects , Enzyme Inhibitors/pharmacology , Gerbillinae , Immunohistochemistry , Male , Neuroprotective Agents/pharmacology , Prosencephalon/blood supply , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Time
19.
Neurochem Res ; 38(1): 74-81, 2013 Jan.
Article En | MEDLINE | ID: mdl-22987057

DNA methylation is a key epigenetic modification of DNA that is catalyzed by DNA methyltransferases (Dnmt). Increasing evidences suggest that DNA methylation in neurons regulates synaptic plasticity as well as neuronal network activity. In the present study, we investigated the changes in DNA methyltransferases 1 (Dnmt1) immunoreactivity and its protein levels in the gerbil hippocampal CA1 region after 5 min of transient global cerebral ischemia. CA1 pyramidal neurons were well stained with NeuN (a neuron-specific soluble nuclear antigen) antibody in the sham-group, Four days after ischemia-reperfusion (I-R), NeuN-positive ((+)) cells were significantly decreased in the stratum pyramidale (SP) of the CA1 region, and many Fluro-Jade B (a marker for neuronal degeneration)(+) cells were observed in the SP. Dnmt1 immunoreactivity was well detected in all the layers of the sham-group. Dnmt1 immunoreactivity was hardly detected only in the stratum pyramidale of the CA1 region from 4 days post-ischemia; however, at these times, Dnmt1 immunoreactivity was newly expressed in GABAergic interneurons or astrocytes in the ischemic CA1 region. In addition, the level of Dnmt1 was lowest at 4 days post-ischemia. In brief, both the Dnmt1 immunoreactivity and protein levels were distinctively decreased in the ischemic CA1 region 4 days after transient cerebral ischemia. These results indicate that the decrease of Dnmt1 expression at 4 days post-ischemia may be related to ischemia-induced delayed neuronal death.


CA1 Region, Hippocampal/enzymology , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , Ischemic Attack, Transient/enzymology , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Blotting, Western , Cell Death/drug effects , DNA (Cytosine-5-)-Methyltransferase 1 , Fluoresceins , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Gerbillinae , Immunohistochemistry , Interneurons/drug effects , Interneurons/enzymology , Male , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology
20.
J Cereb Blood Flow Metab ; 33(1): 106-14, 2013 Jan.
Article En | MEDLINE | ID: mdl-23032483

Rosiglitazone, a synthetic peroxisome proliferator-activated receptor-γ (PPARγ) agonist, prevents cell death after cerebral ischemia in animal models, but the underlying mechanism has not been clarified. In this study, we examined how rosiglitazone protects neurons against ischemia. Mice treated with rosiglitazone were subjected to 60 minutes of focal ischemia followed by reperfusion. Rosiglitazone reduced infarct volume after ischemia and reperfusion. We show that this neuroprotective effect was reversed with a PPARγ antagonist. Western blot analysis showed a significant increase in expression of phosphorylated stress-activated protein kinases (c-Jun N-terminal kinase (JNK) and p38) in ischemic brain tissue. Rosiglitazone blocked this increase. Furthermore, we observed that rosiglitazone increased expression of the dual-specificity phosphatase 8 (DUSP8) protein and messenger RNA in ischemic brain tissue. Dual-specificity phosphatase 8 is a mitogen-activated protein kinase phosphatase that can dephosphorylate JNK and p38. Another key finding of the present study was that knockdown of DUSP8 in primary cultured cortical neurons that were subjected to oxygen-glucose deprivation diminished rosiglitazone's effect on downregulation of JNK phosphorylation. Thus, rosiglitazone's neuroprotective effect after ischemia is mediated by blocking JNK phosphorylation induced by ischemia via DUSP8 upregulation.


Dual-Specificity Phosphatases/metabolism , Ischemic Attack, Transient/drug therapy , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Thiazolidinediones/therapeutic use , Animals , Blotting, Western , Cell Death , Disease Models, Animal , Enzyme Activation , Glucose/metabolism , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Inbred C57BL , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Oxygen/metabolism , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Phosphorylation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology
...