Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
J Biol Chem ; 298(3): 101721, 2022 03.
Article in English | MEDLINE | ID: mdl-35151685

ABSTRACT

Hypoxia-inducible factor prolyl 4-hydroxylases (HIF-P4Hs) regulate the hypoxic induction of >300 genes required for survival and adaptation under oxygen deprivation. Inhibition of HIF-P4H-2 has been shown to be protective in focal cerebral ischemia rodent models, while that of HIF-P4H-1 has no effects and inactivation of HIF-P4H-3 has adverse effects. A transmembrane prolyl 4-hydroxylase (P4H-TM) is highly expressed in the brain and contributes to the regulation of HIF, but the outcome of its inhibition on stroke is yet unknown. To study this, we subjected WT and P4htm-/- mice to permanent middle cerebral artery occlusion (pMCAO). Lack of P4H-TM had no effect on lesion size following pMCAO, but increased inflammatory microgliosis and neutrophil infiltration was observed in the P4htm-/- cortex. Furthermore, both the permeability of blood brain barrier and ultrastructure of cerebral tight junctions were compromised in P4htm-/- mice. At the molecular level, P4H-TM deficiency led to increased expression of proinflammatory genes and robust activation of protein kinases in the cortex, while expression of tight junction proteins and the neuroprotective growth factors erythropoietin and vascular endothelial growth factor was reduced. Our data provide the first evidence that P4H-TM inactivation has no protective effect on infarct size and increases inflammatory microgliosis and neutrophil infiltration in the cortex at early stage after pMCAO. When considering HIF-P4H inhibitors as potential therapeutics in stroke, the current data support that isoenzyme-selective inhibitors that do not target P4H-TM or HIF-P4H-3 would be preferred.


Subject(s)
Blood-Brain Barrier , Infarction, Middle Cerebral Artery , Neuroinflammatory Diseases , Prolyl Hydroxylases , Stroke , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/metabolism , Cell Membrane Permeability , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/metabolism , Mice , Neuroinflammatory Diseases/enzymology , Neuroinflammatory Diseases/metabolism , Permeability , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Stroke/enzymology , Stroke/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
Oxid Med Cell Longev ; 2021: 5173035, 2021.
Article in English | MEDLINE | ID: mdl-34712383

ABSTRACT

Cerebral ischemic stroke (IS) is still a difficult problem to be solved; energy metabolism failure is one of the main factors causing mitochondrion dysfunction and oxidation stress damage within the pathogenesis of cerebral ischemia, which produces considerable reactive oxygen species (ROS) and opens the blood-brain barrier. Dichloroacetic acid (DCA) can inhibit pyruvate dehydrogenase kinase (PDK). Moreover, DCA has been indicated with the capability of increasing mitochondrial pyruvate uptake and promoting oxidation of glucose in the course of glycolysis, thereby improving the activity of pyruvate dehydrogenase (PDH). As a result, pyruvate flow is promoted into the tricarboxylic acid cycle to expedite ATP production. DCA has a protective effect on IS and brain ischemia/reperfusion (I/R) injury, but the specific mechanism remains unclear. This study adopted a transient middle cerebral artery occlusion (MCAO) mouse model for simulating IS and I/R injury in mice. We investigated the mechanism by which DCA regulates glycolysis and protects the oxidative damage induced by I/R injury through the PDK2-PDH-Nrf2 axis. As indicated from the results of this study, DCA may improve glycolysis, reduce oxidative stress and neuronal death, damage the blood-brain barrier, and promote the recovery of oxidative metabolism through inhibiting PDK2 and activating PDH. Additionally, DCA noticeably elevated the neurological score and reduced the infarct volume, brain water content, and necrotic neurons. Moreover, as suggested from the results, DCA elevated the content of Nrf2 as well as HO-1, i.e., the downstream antioxidant proteins pertaining to Nrf2, while decreasing the damage of BBB and the degradation of tight junction proteins. To simulate the condition of hypoxia and ischemia in vitro, HBMEC cells received exposure to transient oxygen and glucose deprivation (OGD). The DCA treatment is capable of reducing the oxidative stress and blood-brain barrier of HBMEC cells after in vitro hypoxia and reperfusion (H/R). Furthermore, this study evidenced that HBMEC cells could exhibit higher susceptibility to H/R-induced oxidative stress after ML385 application, the specific inhibitor of Nrf2. Besides, the protection mediated by DCA disappeared after ML385 application. To sum up, as revealed from the mentioned results, DCA could exert the neuroprotective effect on oxidative stress and blood-brain barrier after brain I/R injury via PDK2-PDH-Nrf2 pathway activation. Accordingly, the PDK2-PDH-Nrf2 pathway may play a key role and provide a new pharmacology target in cerebral IS and I/R protection by DCA.


Subject(s)
Antioxidants/pharmacology , Brain/drug effects , Dichloroacetic Acid/pharmacology , Glycolysis/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Stroke/drug therapy , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Reperfusion Injury/prevention & control , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/ultrastructure , Brain/enzymology , Brain/physiopathology , Brain/ultrastructure , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/ultrastructure , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Ischemic Stroke/enzymology , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Male , Mice, Inbred C57BL , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction
3.
Transl Stroke Res ; 12(1): 147-163, 2021 02.
Article in English | MEDLINE | ID: mdl-32221863

ABSTRACT

Some researchers have previously shown that RNAi knockdown of arginyl-tRNA synthetase (ArgRS) before or after a hypoxic injury can rescue animals from death, based on the model organism, C. elegans. However, there has been no study on the application of arginyl-tRNA synthetase knockdown in treating mammalian ischemic stroke, and its potential mechanism and effect on ischemic brain damage are still unknown. Here, we focused on the Rars gene, which encodes an arginyl-tRNA synthetase, and examined the effects of Rars knockdown in a permanent middle cerebral artery occlusion model in rats. To achieve this aim, adult male Sprague-Dawley (SD) rats were given right cerebral cortex injections of short hairpin RNA (shRNA) adenovirus (AV) particles to knock down arginyl-tRNA synthetase, and a non-targeting control (NTC) vector or phosphate-buffered solution served as the controls. After 4 days, the rats were exposed to permanent middle cerebral artery occlusion (pMCAO). Then, the right cerebral cortex level of arginyl-tRNA synthetase was examined, and the effects of the Rars knockdown were evaluated by differences in infarction volume, oxidative stress, blood-brain barrier, mitochondrial function, and glucose metabolism at 1 day and 3 days after MCAO. The injection of shRNA adenovirus particles successfully suppressed the expression of arginyl-tRNA synthetase in the cerebral cortex. We observed an improvement in oxidative stress, mitochondrial function, and glucose utilization and a reduction in brain edema compared with the non-targeting control rats with suppressed expression of arginyl-tRNA synthetase mRNA in the ipsilateral ischemic cortex of the brain. Our findings indicate that knockdown of arginyl-tRNA synthetase in the cerebral cortex exerted neuroprotective effects, which were achieved not only by the improvement of oxidative stress and glucose utilization but also by the maintenance of mitochondrial morphological integrity and the preservation of mitochondrial function. Knockdown of ArgRS administration could be a promising approach to protect ischemic stroke.


Subject(s)
Arginine-tRNA Ligase/antagonists & inhibitors , Brain Ischemia/enzymology , Cerebral Cortex/enzymology , Gene Knockdown Techniques/methods , Infarction, Middle Cerebral Artery/enzymology , Animals , Arginine-tRNA Ligase/genetics , Brain Ischemia/diagnostic imaging , Brain Ischemia/genetics , Cerebral Cortex/diagnostic imaging , Infarction, Middle Cerebral Artery/diagnostic imaging , Infarction, Middle Cerebral Artery/genetics , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley
4.
J Ethnopharmacol ; 267: 113491, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33091490

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Gardenia jasminoides J. Ellis (Fructus Gardenia) is a traditional Chinese medicine with diverse pharmacological functions, such as anti-inflammation, anti-depression, as well as improvement of cognition and ischemia brain injury. GJ-4 is a natural extract from Gardenia jasminoides J. Ellis (Fructus Gardenia) and has been proved to improve memory impairment in Alzheimer's disease (AD) mouse model in our previous studies. AIM OF THE STUDY: This study aimed to evaluate the therapeutic effects of GJ-4 on vascular dementia (VD) and explore the potential mechanisms. MATERIAL AND METHODS: In our experiment, a focal cerebral ischemia and reperfusion rat model was successfully developed by the middle cerebral artery occlusion and reperfusion (MCAO/R). GJ-4 (10 mg/kg, 25 mg/kg, 50 mg/kg) and nimodipine (10 mg/kg) were orally administered to rats once a day for consecutive 12 days. Learning and memory behavioral performance was assayed by step-down test and Morris water maze test. The neurological scoring test was performed to evaluate the neurological function of rats. 2,3,5-Triphenyltetrazolium chloride (TTC) staining and Nissl staining were respectively employed to determine the infarct condition and neuronal injury of the brain. Iba1 immunohistochemistry was used to show the activation of microglia. Moreover, the synaptic damage and inflammatory level were detected by Western blot. RESULTS: GJ-4 could significantly improve memory impairment, cerebral infraction, as well as neurological deficits of VD rats induced by MCAO/R. Further research indicated VD-induced neuronal injury was alleviated by GJ-4. In addition, GJ-4 could protect synapse of VD rats by upregulating synaptophysin (SYP) expression, post synaptic density 95 protein (PSD95) expression, and downregulating N-Methyl-D-Aspartate receptor 1 (NMDAR1) expression. Subsequent investigation of the underlying mechanisms identified that GJ-4 could suppress neuroinflammatory responses, supported by inhibited activation of microglia and reduced expression of inflammatory proteins, which ultimately exerted neuroprotective effects on VD. Further mechanistic study indicated that janus kinase 2 (JAK2)/signal transducer and activator of transcription 1 (STAT1) pathway was inhibited by GJ-4 treatment. CONCLUSION: These results suggested that GJ-4 might serve as a potential drug to improve VD. In addition, our study indicated that inhibition of neuroinflammation might be a promising target to treat VD.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Dementia, Vascular/prevention & control , Infarction, Middle Cerebral Artery/drug therapy , Janus Kinase 2/metabolism , Memory Disorders/prevention & control , Memory/drug effects , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Plant Extracts/pharmacology , Reperfusion Injury/prevention & control , STAT1 Transcription Factor/metabolism , Animals , Brain/enzymology , Brain/pathology , Brain/physiopathology , Dementia, Vascular/enzymology , Dementia, Vascular/etiology , Dementia, Vascular/psychology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Gardenia , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/physiopathology , Inflammation Mediators/metabolism , Male , Memory Disorders/enzymology , Memory Disorders/etiology , Memory Disorders/psychology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/etiology , Reperfusion Injury/physiopathology , Signal Transduction , Synapses/drug effects , Synapses/metabolism , Synapses/pathology
5.
J Stroke Cerebrovasc Dis ; 30(3): 105165, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33360522

ABSTRACT

BACKGROUND: Cerebral infarction is one of the most common causes of disability and death worldwide. It is reported that electric acupuncture was able to improve the prognosis of cerebral infarction by promoting angiogenesis. However, the corresponding signal pathways of angiogenesis promotes by electric acupuncture treatment needs to be further explored. METHODS: MCAO rat was employed as the animal model, and clopidogrel hydrogen sulfate treatment was set as the positive control. Behaviors of rats, H&E staining, and TTC-staining was used to evaluate the recovery of infarcted brain tissue and nervous function. After that, immunocytochemical and immunofluorescence staining was used to quantify the angiogenesis and compensatory circulation, which including the analysis of microvessel density, field/ microvessel area ratio, and microvessel diameter. Western blot and RT-PCR for the detection of the related signal molecule, PI3K, Src, and EphB4/ephrinB2. RESULTS: The neurologic impairment scores were decreased, and the brain tissue damage that showed with H&E and TTC-staining was relieved by the treatment of electric acupuncture in MCAO rat. The quantification of microvessel density and field/ microvessel area ratio was improved obviously, and the microvessel diameter was decreased which represent the angiogenesis of capillary in day 3 and 7 by the electric acupuncture treatment. We also found that the level of Src and PI3K was increased markedly followed by the up-regulation of EphB4 and EphrinB2 mRNA during the electric acupuncture treatment, and the pre-treatment of Src and/or PI3K inhibitor was able to disturb the angiogenesis of capillary. CONCLUSIONS: We proved that electric acupuncture was able to accelerate the recovery of infarcted brain tissue and nervous function in MCAO rat by the promotion of angiogenesis, which was regulated by EphB4/EphrinB2 mediated Src/PI3K signal pathway. Our study provides a potential therapy and theoretical basis for the clinical treatment of cerebral infarction by the use of electric acupuncture.


Subject(s)
Brain/blood supply , Brain/enzymology , Electroacupuncture , Ephrin-B2/metabolism , Infarction, Middle Cerebral Artery/therapy , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinase/metabolism , Receptor, EphB4/metabolism , src-Family Kinases/metabolism , Animals , Disease Models, Animal , Ephrin-B2/genetics , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/physiopathology , Microvascular Density , Rats, Sprague-Dawley , Receptor, EphB4/genetics , Recovery of Function , Signal Transduction , Time Factors
6.
Biomed Pharmacother ; 133: 111021, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33227709

ABSTRACT

OBJECTIVE: To observe the brain protective effect of Leonuri Herba Total Alkali (LHA) on cerebral ischemia reperfusion injury in rats, so as to provide basis for clinical research. METHODS: Adult male SD rats were randomly assigned into sham group, middle cerebral artery occlusion/reperfusion (MCAO/R) group, and LHA + MCAO/R group (25 mg/kg, 50 mg/kg, and 100 mg/kg). Fourteen days before MCAO/R surgery, the rats in treatment groups were orally administered with LHA in ultrapure water once daily for 14 days, while rats in the sham and MCAO groups were given the same amount of saline in advance. After 1 h of administration on the 14th day, MCAO surgery was subjected. The neurological deficits, brain infarct volume, histopathology, immunofluorescence, inflammation indicators and the gene/protein expressions of BDNF-TrKB-PI3K/Akt signaling pathway in the rat brain tissue were evaluated 24 h after the MCAO/R-injury. RESULTS: It was found that rats in LHA pre-administration group showed significantly reduced neurological deficit scores, infarction volume, the serum levels of NSE and S100ß. Meanwhile, the content of Evans Blue (EB) in brain tissue from LHA group was decreased, as well as the levels of inflammatory cytokines and their gene levels. Moreover, LHA pre-administration inhibited the expression of CD44, GFAP, FOXO1 and promoted the expression of BDNF and NeuN. In addition, LHA pre-administration could up-regulate the protein expression of TrkB, p-PI3K, p-Akt, Bcl-2, and down-regulate the protein expression of Bax, and increase the level of Bcl-2/Bax. CONCLUSIONS: The study demonstrated that LHA pre-administration could regulate the PI3K/Akt pathway by increasing BDNF levels, and play a neuroprotective role in cerebral ischemia-reperfusion injury.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/drug effects , Drugs, Chinese Herbal/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Leonurus/chemistry , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkB/metabolism , Reperfusion Injury/prevention & control , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/enzymology , Brain/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphorylation , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction
7.
Aging (Albany NY) ; 12(23): 23872-23888, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33263567

ABSTRACT

The current study was designed to seek the role of the glycogen synthase kinase-3ß (GSK-ß)-regulated NF-E2-related factor 2 (Nrf2) pathway in the antioxidant effect induced by Apigenin-7-O-ß-D-(-6"-p-coumaroyl)-glucopyranoside (APG). Rat primary cultured cortical neurons were challenged by oxygen and glucose deprivation/reoxygenation (OGD/R) and then treated with APG. Cell viability, phosphorylation of GSK-ß at Ser9 and nuclear expression of Nrf2 were measured. Male Sprague Dawley rats challenged by 2-h middle cerebral artery occlusion were treated with 50 mg/kg APG, and the neurological score, infarct volume, phosphorylation of GSK-3ß and nuclear expression of Nrf2 were analyzed. The neuroprotective effect of APG and the expression levels of antioxidant enzymes and oxidative products were also examined in the presence and absence of Nrf2-siRNA and PI3K inhibitors. APG reduced the apoptotic proportion, attenuated LDH release and increased cell viability, and in vivo, APG improved neurological scores and reduced infarct volume. APG increased GSK-3ß phosphorylation and Nrf2 nuclear translocation, while these effects were prevented by PI3K inhibitors or Nrf2-siRNA treatment in both OGD/R cell cultures and ischemic/reperfusion rats. These findings reveal that GSK-3ß phosphorylation-mediated Nrf2 activation is involved in the neuroprotective effect of APG.


Subject(s)
Antioxidants/pharmacology , Apigenin/pharmacology , Cerebral Cortex/drug effects , Glucosides/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Infarction, Middle Cerebral Artery/drug therapy , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , NF-E2-Related Factor 2/genetics , Neurons/enzymology , Neurons/pathology , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction
8.
Basic Res Cardiol ; 115(6): 64, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057972

ABSTRACT

By cleaving sphingomyelin into ceramide, which is an essential component of plasma membrane microdomains, acid sphingomyelinase (Asm) pivotally controls cell signaling. To define how the activation of the Asm/ceramide pathway, which occurs within seconds to minutes upon stress stimuli, influences brain ischemia/reperfusion (I/R) injury, we exposed male and female wildtype mice carrying both alleles of Asm's gene sphingomyelinase phosphodiesterase-1 (Smpd1+/+), heterozygously Asm-deficient mice (Smpd1+/-) and homozygously Asm-deficient mice (Smpd1-/-) of different age (8, 12 or 16 weeks) to 30, 60 or 90 min intraluminal middle cerebral artery occlusion (MCAO). For studying the contribution of brain-invading polymorphonuclear neutrophils (PMN) to I/R injury, PMNs were depleted by delivery of a PMN-specific Ly6G antibody. In male and female mice exposed to 30 min, but not 60 or 90 min MCAO, homozygous Smpd1-/- consistently increased I/R injury, blood-brain barrier permeability and brain leukocyte and PMN infiltration, whereas heterozygous Smpd1+/- reduced I/R injury. Increased abundance of the intercellular leukocyte adhesion molecule ICAM-1 was noted on cerebral microvessels of Smpd1-/- mice. PMN depletion by anti-Ly6G delivery prevented the exacerbation of I/R injury in Smpd1-/- compared with wildtype mice and reduced brain leukocyte infiltrates. Our results show that Asm tempers leukocyte entry into the reperfused ischemic brain, thereby attenuating I/R injury.


Subject(s)
Brain/blood supply , Infarction, Middle Cerebral Artery/enzymology , Microvessels/enzymology , Neutrophil Infiltration , Neutrophils/enzymology , Reperfusion Injury/enzymology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Disease Models, Animal , Female , Heterozygote , Homozygote , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Sphingomyelin Phosphodiesterase/genetics , Time Factors
9.
J Stroke Cerebrovasc Dis ; 29(10): 105029, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32912542

ABSTRACT

BACKGROUND: We investigated whether exogenous lysophosphatidic acid (LPA), a phospholipid extracellular signaling molecule, would increase infarct size and blood-brain barrier (BBB) disruption during the early stage of cerebral ischemia-reperfusion, and whether it works through Akt-mTOR-S6K1 intracellular signaling. MATERIAL AND METHODS: Rats were given either vehicle or LPA 1 mg/kg iv three times during reperfusion after one hour of middle cerebral artery (MCA) occlusion. In another group, prior to administration of LPA, 30 mg/kg of PF-4708671, an S6K1 inhibitor, was injected. After one hour of MCA occlusion and two hours of reperfusion the transfer coefficient (Ki) of 14C-α-aminoisobutyric acid and the volume of 3H-dextran distribution were determined to measure the degree of BBB disruption. At the same time, the size of infarct was determined and western blot analysis was performed to determine the levels of phosphorylated Akt (p-Akt) and phosphorylated S6 (pS6). RESULTS: LPA increased the Ki in the ischemic-reperfused cortex (+43%) when compared with Control rats and PF-4708671 pretreatment prevented the increase of Ki by LPA. LPA increased the percentage of cortical infarct out of total cortical area (+36%) and PF-4708671 pretreatment prevented the increase of the infarct size. Exogenous LPA did not significantly change the levels of p-Akt as well as pS6 in the ischemic-reperfused cortex. CONCLUSION: Our data demonstrate that the increase in BBB disruption could be one of the reasons of the increased infarct size by LPA. S6K1 may not be the major target of LPA. A decrease of LPA during early cerebral ischemia-reperfusion might be beneficial for neuronal survival.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Cerebral Cortex/drug effects , Infarction, Middle Cerebral Artery/therapy , Lysophospholipids/toxicity , Reperfusion Injury/chemically induced , Reperfusion , Animals , Blood-Brain Barrier/physiopathology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Inbred F344 , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Ribosomal Protein S6 Kinases/metabolism
10.
Eur J Pharmacol ; 883: 173294, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32681941

ABSTRACT

Neuroinflammation is critical for the pathogenesis of ischemia brain damage. Over-activated microglia-mediated inflammation plays a very important role in ischemia cerebral injuries. 6-Gingerol, obtained from edible ginger (Zingiber Officinale) exhibits protective effects against inflammation. In this study, we found that 6-Gingerol could reduce the size of infarction (P = 0.0184) and improve neurological functions (P = 0.04) at the third day after ischemic brain injury in vivo. Since 6-Gingerol has the anti-inflammatory effects, we further investigated its impacts on neuroinflammation mediated by microglia both in vivo and in vitro. We found that the levels of pro-inflammatory cytokines Interleukin-1 beta (IL-1ß, P = 0.0213), Interleukin-6 (IL-6, P = 0.0316), and inducible NO synthase (iNOS, P = 0.0229) in the infarct penumbra were lower in 6-Gingerol treated groups. Furthermore, microglia induced pro-inflammatory cytokines, such as IL-6, IL-1ß, incremental intercellular nitric oxide (NO), as well as iNOS were blocked by the treatment of 6-Gingerol in lipopolysaccharide (LPS) stimulated microglia. In terms of mechanism, 6-Gingerol potently suppressed phosphorylation of serine-threonine protein kinase (Akt) - mammalian target of rapamycin (mTOR) - signal transducer and activator of transcription 3 (STAT3) in LPS-treated microglia. Taken together, the present study suggested that 6-Gingerol improved cerebral ischemia injury by suppressing microglia-mediated neuroinflammation by down-regulating Akt-mTOR-STAT3 pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Catechols/pharmacology , Fatty Alcohols/pharmacology , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Stroke/prevention & control , Microglia/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Brain/enzymology , Brain/pathology , Brain/physiopathology , Cells, Cultured , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Inflammation Mediators/metabolism , Ischemic Stroke/enzymology , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Male , Mice, Inbred C57BL , Microglia/enzymology , Microglia/pathology , Phosphorylation , Signal Transduction
11.
Indian J Pharmacol ; 52(2): 108-116, 2020.
Article in English | MEDLINE | ID: mdl-32565598

ABSTRACT

We investigate the protective effect of Carthamus tinctorius L. (CTL, also known as Honghua in China or Safflower) on cerebral ischemia-reperfusion and explored the possible mechanisms on regulating apoptosis and matrix metalloproteinases (MMPs). High-performance liquid chromatography method with diode array detection analysis was established to analyze the components of CTL. Middle cerebral artery occlusion rats model was established to evaluate Neurological Function Score and hematoxylin-eosin staining, as well as triphenyltetrazolium was used to examine the infarction area ratio. Transferase-mediated dUTP nick-end labeling was performed for the apoptosis. Apoptosis-related factors, including B-cell lymphoma-2 (Bcl-2), Bax and Caspase3, and MMPs-related MMP2, MMP9, tissue inhibitor of metalloproteinases 1 (TIMP1) in ischemic brain, were assayed by Western blot, reverse transcription polymerase chain reaction, and immunohistochemistry. The data showed that CTL (2, 4 g crude drug/kg/d) treatment could significantly reduce the ischemic damage in brain tissue and improve a significant neurological function score. In addition, CTL could also attenuate apoptosis degree of brain tissues and regulate Bcl-2, Bax, and Caspase 3 and also have a significant decrease on MMP-9 expression, followed by a significant increase of TIMP1 protein expression. These findings indicated that regulation of CTL on apoptosis and MMPs contributed to its protective effect on ischemia/reperfusion injury.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Carthamus tinctorius , Infarction, Middle Cerebral Artery/drug therapy , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Reperfusion Injury/prevention & control , Animals , Apoptosis Regulatory Proteins/metabolism , Brain/enzymology , Brain/pathology , Carthamus tinctorius/chemistry , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , Neuroprotective Agents/isolation & purification , Plant Extracts/isolation & purification , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/metabolism
12.
Biomed Pharmacother ; 128: 110045, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32460187

ABSTRACT

Ischemic stroke is a major cause of death and disability worldwide. Necroptosis is known as a form of cell death, playing an essential role in regulating ischemia-induced brain injury. Triad3A is a ubiquitin ligase of the RING-in-between-RING family, and regulates necroptotic cell death under different pathological conditions, including neurodegenerative disorders. In the present study, the effects of Triad3A on experimental stroke were explored on a mouse model with middle cerebral artery occlusion (MCAO). The results indicated that Triad3A expression was markedly induced in the ischemic brain after MCAO operation. The neurons and microglia cells were the major cellular sources for Triad3A induction. Triad3A knockdown enhanced the infarction area, cell death, microglia activity, and the expression levels of pro-inflammatory markers including tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, inducible nitric oxide synthase (iNOS), CD32 and CD68 in MCAO mice. Triad3A and necroptosis were triggered in mouse microglia cells treated with oxygen and glucose deprivation (OGD), and in TNFα-incubated mouse hippocampal neuronal cells treated with Z-VAD-fmk, known as a pan-caspase inhibitor. Moreover, Triad3A knockdown accelerated cell death in microglial cells and neurons under these stresses. Furthermore, pre-treatment with necroptosis inhibitor markedly inhibited the cell death promoted by Triad3A silence in brain of mice with MCAO operation, demonstrating that Triad3A could regulate necroptosis to meditate the progression of cerebral I/R injury. Collectively, these finding illustrated that Triad3A could be served as a potential target for stroke therapy.


Subject(s)
Brain/enzymology , Infarction, Middle Cerebral Artery/enzymology , Microglia/enzymology , Necroptosis , Neurons/enzymology , Reperfusion Injury/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Brain/pathology , Cell Line , Disease Models, Animal , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Microglia/pathology , Neurons/pathology , Protein Kinases/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Signal Transduction , Ubiquitin-Protein Ligases/genetics
13.
J Cardiovasc Pharmacol ; 76(2): 227-236, 2020 08.
Article in English | MEDLINE | ID: mdl-32453073

ABSTRACT

Cerebral ischemia-reperfusion (I/R) is a kind of neurovascular disease that causes serious cerebral damage. MicroRNAs (miRNAs) have been widely reported to participate in multiple diseases, including cerebral I/R injury. However, the exact mechanisms of miR-7-5p in cerebral I/R injury was not fully elucidated. In this study, we explored the biological role and regulatory mechanism of miR-7-5p in cerebral I/R injury. We established an in vivo model of cerebral I/R by middle cerebral artery occlusion and an in vitro cellular model of cerebral I/R injury through treating neurons (SH-SY5Y cells) with oxygen-glucose deprivation (OGD). In addition, miR-7-5p expression was confirmed to be upregulated in the cerebral I/R rat model and OGD/R-treated SH-SY5Y cells. Moreover, miR-7-5p inhibition overtly suppressed cerebral injury, cerebral inflammation, and SH-SY5Y cells apoptosis. Sirtuin 1 (sirt1) is previously reported to alleviate I/R, and in this study, it was identified to be a target of miR-7-5p based on luciferase reporter assay. Reverse transcription-quantitative polymerase chain reaction revealed sirt1 expression was downregulated in the cerebral I/R rat model and OGD/R-treated SH-SY5Y cells. Besides, miR-7-5p negatively regulated sirt1. Finally, rescue assays delineated sirt1 overexpression recovered the miR-7-5p upregulation-induced promotion on cerebral I/R injury. In conclusion, miR-7-5p enhanced cerebral I/R injury by degrading sirt1, providing a new paradigm to investigate cerebral I/R injury.


Subject(s)
Brain/enzymology , Infarction, Middle Cerebral Artery/enzymology , MicroRNAs/metabolism , Neurons/enzymology , RNA Stability , RNA, Messenger/metabolism , Reperfusion Injury/enzymology , Sirtuin 1/metabolism , Animals , Apoptosis , Brain/pathology , Cell Hypoxia , Cell Line, Tumor , Female , Gene Expression Regulation , Glucose/deficiency , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , MicroRNAs/genetics , NF-kappa B/metabolism , Neurons/pathology , RNA, Messenger/genetics , Rats, Wistar , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Sirtuin 1/genetics
14.
J Am Heart Assoc ; 9(9): e014920, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32349637

ABSTRACT

Background Although multiple signaling cascades and molecules contributing to the pathophysiological process have been studied, the treatments for stroke against present targets have not acquired significant clinical progress. Although CARD3 (caspase activation and recruitment domain 3) protein is an important factor involved in regulating immunity, inflammation, lipid metabolism, and apoptosis, its role in cerebral stroke is currently unknown. Methods and Results Using a mouse model of ischemia-reperfusion (I-R) injury based on transient blockage of the middle cerebral artery, we have found that CARD3 expression is upregulated in a time-dependent manner during I-R injury. Further animal study revealed that, relative to control mice, CARD3-knockout mice exhibited decreased inflammatory response and neuronal apoptosis, with reduced infarct volume and lower neuropathological scores. In contrast, neuron-specific CARD3-overexpressing transgenic (CARD3-TG) mice exhibited increased I-R induced injury compared with controls. Mechanistically, we also found that the activation of TAK1 (transforming growth factor-ß-activated kinase 1) was enhanced in CARD3-TG mice. Furthermore, the increased inflammation and apoptosis seen in injured CARD3-TG brains were reversed by intravenous administration of the TAK1 inhibitor 5Z-7-oxozeaenol. Conclusions These results indicate that CARD3 promotes I-R injury via activation of TAK1, which not only reveals a novel regulatory axis of I-R induced brain injury but also provides a new potential therapeutic approach for I-R injury.


Subject(s)
Brain/enzymology , Infarction, Middle Cerebral Artery/enzymology , MAP Kinase Kinase Kinases/metabolism , Neurons/enzymology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Reperfusion Injury/enzymology , Animals , Apoptosis , Brain/pathology , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Phosphorylation , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinase 2/deficiency , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
15.
J Stroke Cerebrovasc Dis ; 29(6): 104801, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32249206

ABSTRACT

BACKGROUND: Ischemic stroke is the leading cause of disability and death globally. Micro-RNAs (miRNAs) have been reported to play important roles in the development and pathogenesis of the nervous system. However, the exact function and mechanism of miRNAs have not been fully elucidated about brain damage caused by cerebral ischemia/reperfusion (I/R). METHODS: In this study, we explored the neuroprotective effects of miR-219a-5p on brain using an in vitro ischemia model (mouse neuroblastoma N2a cells treated with oxyglucose deprivation and reperfusion), and in vivo cerebral I/R model in mice. Western blot assay and Reverse Transcription-Polymerase Chain Reaction were used to check the expression of molecules involved. Flow cytometry and cholecystokinin were used to examine cell apoptosis, respectively. RESULTS: Our research shows that miR-219a-5p gradually decreases in cerebral I/R models in vivo and in vitro. In vitro I/R, we find that miR-219a-5p mimics provided evidently protection for cerebral I/R damage, as shown by increased cell viability and decreased the release of LDH and cell apoptosis. Mechanically, our findings indicate that miR-219a-5p binds to cAMP specific 3', 5'-cyclic phosphodiesterase 4D (PDE4D) mRNA in the 3'-UTR region, which subsequently leads to a decrease in Pde4d expression in I/R N2a cells. CONCLUSIONS: Our results provide new ideas for the study of the mechanism of cerebral ischemia/reperfusion injury, and lay the foundation for further research on the treatment of brain I/R injury. Upregulation of miR-219a-5p decreases cerebral ischemia/reperfusion injury by targeting Pde4d in vitro.


Subject(s)
Apoptosis , Brain/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Infarction, Middle Cerebral Artery/enzymology , MicroRNAs/metabolism , Neurons/enzymology , Reperfusion Injury/enzymology , 3' Untranslated Regions , Animals , Binding Sites , Brain/pathology , Cell Line, Tumor , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Disease Models, Animal , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Neurons/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Signal Transduction
16.
J Stroke Cerebrovasc Dis ; 29(5): 104743, 2020 May.
Article in English | MEDLINE | ID: mdl-32127256

ABSTRACT

BACKGROUND: Bone marrow stromal cell (BMSC) transplantation is a promising therapeutic approach for cerebral ischemia, as it elicits multiple neuroprotective effects. However, it remains unclear how BMSC transplantation modulates the ubiquitin-proteasome system (UPS) and autophagy under cerebral ischemia. METHODS: In the present study, an intermediate level of cerebral ischemia (30 minutes) was chosen to examine the effect of BMSC transplantation on the molecular switch regulating UPS and autophagy. BMSC or vehicle was stereotactically injected into the penumbra 15 minutes after sham operation or transient middle cerebral artery occlusion (tMCAO). RESULTS: Thirty minutes of tMCAO artery occlusion significantly increased TUNEL-, ubiquitin-, and p62-positive cells (which peaked at 72 hours, 2 hours, and 2 hours after reperfusion, respectively) and ratios of both BAG3/BAG1 and LC3-II/LC3-I at 24 hours after reperfusion. However, intracerebral injection of BMSCs significantly reduced infarct volume and numbers of TUNEL- and p62-positive cells, and improved BAG3/BAG1 and LC3-II/LC3-I ratios. In addition, observed increases in ubiquitin-positive cells 2 hours after reperfusion were slightly suppressed by BMSC transplantation. CONCLUSIONS: These data suggest a protective role of BMSC transplantation, which drove the molecular switch from autophagy to UPS in a murine model of ischemic stroke.


Subject(s)
Autophagy , Brain/enzymology , Infarction, Middle Cerebral Artery/surgery , Mesenchymal Stem Cell Transplantation , Proteasome Endopeptidase Complex/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Brain/pathology , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction , Time Factors , Transcription Factors/metabolism , Ubiquitination
17.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1753-1760, 2020 09.
Article in English | MEDLINE | ID: mdl-31900521

ABSTRACT

Isorhapontigenin (ISO) is one of the main bioactive components of Gnetum cleistostachyum and was shown to possess antioxidant and antitumor functions. Herein, we hope to examine the neuroprotection impacts of ISO in rats subjected to transient middle cerebral artery occlusion/reperfusion (MCAO/R, 2/24 h) injuries. ISO was injected intraperitoneally into the rats immediately after cerebral ischemia. After 24 h of the reperfusion, infarct volume, brain water contents, neurological deficit, and cerebral blood flow were assessed. Hippocampus histopathology change was detected by H&E and TUNEL staining. The expressions of cleaved caspase-3, Bax and Bcl-2, and phospho-Akt (p-Akt) were investigated by real-time RT-PCR or western blot analysis. We found that ISO significantly suppressed the infarct volumes, brain water contents, and neurological deficit, increased CBF, and relieved histopathologic change in a dose-dependent manner. Reduced malondialdehyde (MDA) and elevated activities of superoxide dismutase (SOD) and GSH and glutathione peroxidase (GSH-PX) were observed in ISO group. ISO remarkably decreased caspase-3 and Bax and increased levels of Bcl-2. Additionally, ISO upregulated p-Akt expression. Blocking of PI3K activities by wortmannin can abolish the ISO-caused decrease in infarct volumes and neurologic deficit scores and abrogate the promotion of p-Akt. The data indicated that ISO played neuroprotective impacts against focal I/R injuries, possibly related to the activating of PI3K/Akt signaling.


Subject(s)
Hippocampus/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/prevention & control , Stilbenes/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Disease Models, Animal , Hippocampus/enzymology , Hippocampus/pathology , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , Oxidative Stress/drug effects , Phosphorylation , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction
18.
J Stroke Cerebrovasc Dis ; 28(12): 104470, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31680031

ABSTRACT

BACKGROUND: The nitric oxide (NO)-producing activity of endothelial nitric oxide synthase (eNOS) plays a significant role in maintaining endothelial function and protecting against the stroke injury. However, the activity of the eNOS enzyme and the metabolism of major NO metabolite S-nitrosoglutathione (GSNO) are dysregulated after stroke, causing endothelial dysfunction. We investigated whether an administration of exogenous of GSNO or enhancing the level of endogenous GSNO protects against neurovascular injury in wild-type (WT) and eNOS-null (endothelial dysfunction) mouse models of cerebral ischemia-reperfusion (IR). METHODS: Transient cerebral ischemic injury was induced by middle cerebral artery occlusion (MCAO) for 60 minutes in male adult WT and eNOS null mice. GSNO (0.1 mg/kg body weight, intravenously) or N6022 (GSNO reductase inhibitor, 5.0 mg/kg body weight, intravenously) was administered 30 minutes before MCAO in preinjury and at the reperfusion in postinjury studies. Brain infarctions, edema, and neurobehavioral functions were evaluated at 24 hours after the reperfusion. RESULTS: eNOS-null mice had a higher degree (P< .05) of injury than WT. Pre- or postinjury treatment with either GSNO or N6022 significantly reduced infarct volume, improved neurological and sensorimotor function in both WT and eNOS-null mice. CONCLUSION: Reduced brain infarctions and edema, and improved neurobehavioral functions by pre- or postinjury GSNO treatment of eNOS knock out mice indicate that GSNO can attenuate IR injury, likely by mimicking the eNOS-derived NO-dependent anti-ischemic and anti-inflammatory functions. Neurovascular protection by GSNO/N6022 in both pre- and postischemic injury groups support GSNO as a promising drug candidate for the prevention and treatment of stroke injury.


Subject(s)
Alcohol Dehydrogenase/antagonists & inhibitors , Benzamides/pharmacology , Brain/drug effects , Enzyme Inhibitors/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Pyrroles/pharmacology , S-Nitrosoglutathione/pharmacology , Alcohol Dehydrogenase/metabolism , Animals , Behavior, Animal/drug effects , Brain/embryology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Brain Edema/prevention & control , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics
19.
J Stroke Cerebrovasc Dis ; 28(10): 104276, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31350168

ABSTRACT

BACKGROUND: We tested the hypothesis that inhibition of p70 ribosomal S6 kinase (S6K1) would decrease infarct size and improve microregional O2 supply/consumption balance after cerebral ischemia-reperfusion. METHODS: This was tested in isoflurane-anesthetized rats with middle cerebral artery blockade for 1 hour and reperfusion for 2 hours with or without PF-4708671 (S6K1 inhibitor, 75 mg/kg, 15 minutes after blockade). Regional cerebral blood flow was determined using a C14-iodoantipyrine autoradiographic technique. Regional small vessel (20-60 µm diameter) arterial and venous oxygen saturations were determined microspectrophotometrically. RESULTS: There were no significant hemodynamic or arterial blood gas differences between groups. The control ischemic-reperfused cortex had a similar O2 consumption to the contralateral cortex. However, microregional O2 supply/consumption balance was significantly reduced in the ischemic-reperfused cortex with many areas of low O2 saturation (23 of 80 veins with O2 saturation below 45%). PF-4708671 did not significantly alter cerebral blood flow or O2 consumption. However, it significantly reduced the number of small veins with low O2 saturations in the reperfused region (6 of 80 veins with O2 saturation below 45%). This was associated with a significantly reduced cortical infarct size after S6K1 inhibition (12.9 ± .8% control versus 6.6 ± .3% PF-4708671). CONCLUSION: This suggests that S6K1 inhibition is important for cell survival and that it reduces the number of small microregions with reduced local oxygen balance after cerebral ischemia-reperfusion.


Subject(s)
Brain/blood supply , Brain/drug effects , Cerebrovascular Circulation/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Microcirculation/drug effects , Neuroprotective Agents/pharmacology , Oxygen Consumption/drug effects , Oxygen/blood , Piperazines/pharmacology , Reperfusion Injury/prevention & control , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Animals , Brain/enzymology , Brain/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Rats, Inbred F344 , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Ribosomal Protein S6 Kinases/metabolism
20.
J Stroke Cerebrovasc Dis ; 28(7): 1832-1840, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31078389

ABSTRACT

GOAL: The present study aimed to examine whether Am80 (tamibarotene) protects the hippocampus against cerebral ischemia-reperfusion (I/R) injury and whether phosphoinositide-3-kinase/Akt (PI3K/Akt) pathway mediates this effect. MATERIALS AND METHODS: Rats were subjected to 90 minutes of middle cerebral artery occlusion followed by 24 hours of reperfusion. The animals were randomly divided into 7 groups: sham-operated group; I/R group; groups pretreated with 2 mg/kg, 6 mg/kg, and 10 mg/kg of Am80; Am80 (6 mg/kg) combined with the selective PI3K inhibitor wortmannin (0.6 mg/kg), and wortmannin (0.6 mg/kg) only group. After 24 hours of reperfusion, neurological deficits and infarct volume were measured. Pathological changes in hippocampal neurons were analyzed by transmission electron microscopy. Neuronal survival was examined by TUNEL staining. The expression of Bcl-2, Bax, and Akt, and Akt phosphorylation (p-Akt) were measured by Western blotting and quantitative real-time polymerase chain reaction. FINDINGS: The pretreatment with Am80 improved the neurologic deficit score, reduced infarct volume, and decreased the number of TUNEL-positive cells in the hippocampus. Moreover, Am80 pretreatment downregulated the expression of Bax, upregulated the expression of Bcl-2, and increased the level of p-Akt. Wortmannin abolished in part the increase in p-Act and the neuroprotective effect exerted on the ischemic by Am80 pretreatment. CONCLUSIONS: Our results documented that Am80 pretreatment protects ischemic hippocampus after cerebral I/R by regulating the expression of apoptosis-related proteins through the activation of the PI3K/Akt signaling pathway.


Subject(s)
Benzoates/pharmacology , Hippocampus/drug effects , Infarction, Middle Cerebral Artery/prevention & control , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/prevention & control , Tetrahydronaphthalenes/pharmacology , Animals , Apoptosis/drug effects , Disease Models, Animal , Hippocampus/enzymology , Hippocampus/ultrastructure , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , Neurons/enzymology , Neurons/ultrastructure , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL