• Title/Summary/Keyword: oxygen glucose deprivation

Search Result 58, Processing Time 0.023 seconds

Gene Expression Profile in Microglia following Ischemia-Reperfusion Injury

  • Oh, Ju-Hyeon;Han, Hyung-Soo;Park, Jae-Sik
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.10 no.4
    • /
    • pp.173-180
    • /
    • 2006
  • Microglial activation is thought to play a role in the pathogenesis of many brain disorders. Therefore, understanding the response of microglia to noxious stimuli may provide insights into their role in disorders such as stroke and neurodegeneration. Many genes involved in this response have been identified individually, but not systematically. In this regards, the microarray system permitted to screen a large number of genes in biological or pathological processes. Therefore, we used microarray technology to evaluate the effect of oxygen glucose deprivation (OGD) and reperfusion on gene expression in microglia under ischemia-like and activating conditions. Primary microglial cultures were prepared from postnatal mice brain. The cells were exposed to 4 hrs of OGD and 1 h of reperfusion at $37^{\circ}C$. Isolated mRNA were run on GeneChips. After OGD and reperfusion, >2-fold increases of 90 genes and >2-fold decrease of 41 genes were found. Among the genes differentially increased by OGD and reperfusion in microglia were inflammatory and immune related genes such as prostaglandin E synthase, $IL-1{\beta}$, and $TNF-{\alpha}$. Microarray analysis of gene expression may be useful for elucidating novel molecular mediators of microglial reaction to reperfusion injury and provide insights into the molecular basis of brain disorders.

Ginkgolide B Modulates BDNF Expression in Acute Ischemic Stroke

  • Wei, Hu;Sun, Tao;Tian, Yanghua;Wang, Kai
    • Journal of Korean Neurosurgical Society
    • /
    • v.60 no.4
    • /
    • pp.391-396
    • /
    • 2017
  • Objective : To investigate the neuroprotective effects of Ginkgolide B (GB) against ischemic stroke-induced injury in vivo and in vitro, and further explore the possible mechanisms concerned. Methods : Transient middle cerebral artery occlusion (tMCAO) mice and oxygen-glucose deprivation/reoxygenation (OGD/R)-treated N2a cells were used to explore the neuroprotective effects of GB. The expression of brain-derived neurotrophic factor (BDNF) was detected via Western blot and qRT-PCR. Results : GB treatment (4 mg/kg, i. p., bid) significantly reduced neurological deficits, water content, and cerebral infarct volume in tMCAO mice. GB also significantly increased Bcl-2/Bax ratio, reduced the expression of caspase-3, and protected against OGD/R-induced neuronal apoptosis. Meanwhile, GB caused the up-regulation of BDNF protein in vivo and in vitro. Conclusion : Our data suggest that GB might protect the brain against ischemic insult partly via modulating BDNF expression.

Bis is Induced by Oxidative Stress via Activation of HSF1

  • Yoo, Hyung Jae;Im, Chang-Nim;Youn, Dong-Ye;Yun, Hye Hyeon;Lee, Jeong-Hwa
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.18 no.5
    • /
    • pp.403-409
    • /
    • 2014
  • The Bis protein is known to be involved in a variety of cellular processes including apoptosis, migration, autophagy as well as protein quality control. Bis expression is induced in response to a number of types of stress, such as heat shock or a proteasome inhibitor via the activation of heat shock factor (HSF)1. We report herein that Bis expression is increased at the transcriptional level in HK-2 kidney tubular cells and A172 glioma cells by exposure to oxidative stress such as $H_2O_2$ treatment and oxygen-glucose deprivation, respectively. The pretreatment of HK-2 cells with N-acetyl cysteine, suppressed Bis induction. Furthermore, HSF1 silencing attenuated Bis expression that was induced by $H_2O_2$, accompanied by increase in reactive oxygen species (ROS) accumulation. Using a series of deletion constructs of the bis gene promoter, two putative heat shock elements located in the proximal region of the bis gene promoter were found to be essential for the constitutive expression is as well as the inducible expression of Bis. Taken together, our results indicate that oxidative stress induces Bis expression at the transcriptional levels via activation of HSF1, which might confer an expansion of antioxidant capacity against pro-oxidant milieu. However, the possible role of the other cis-element in the induction of Bis remains to be determined.

Protective effects of N,4,5-trimethylthiazol-2-amine hydrochloride on hypoxia-induced β-amyloid production in SH-SY5Y cells

  • Han, A Reum;Yang, Ji Woong;Na, Jung-Min;Choi, Soo Young;Cho, Sung-Woo
    • BMB Reports
    • /
    • v.52 no.7
    • /
    • pp.439-444
    • /
    • 2019
  • Although hypoxic/ischemic injury is thought to contribute to the incidence of Alzheimer's disease (AD), the molecular mechanism that determines the relationship between hypoxia-induced ${\beta}$-amyloid ($A{\beta}$) generation and development of AD is not yet known. We have now investigated the protective effects of N,4,5-trimethylthiazol-2-amine hydrochloride (KHG26702), a novel thiazole derivative, on oxygen-glucose deprivation (OGD)-reoxygenation (OGD-R)-induced $A{\beta}$ production in SH-SY5Y human neuroblastoma cells. Pretreatment of these cells with KHG26702 significantly attenuated OGD-R-induced production of reactive oxygen species and elevation of levels of malondialdehyde, prostaglandin $E_2$, interleukin 6 and glutathione, as well as superoxide dismutase activity. KHG26702 also reduced OGD-R-induced expression of the apoptotic protein caspase-3, the apoptosis regulator Bcl-2, and the autophagy protein becn-1. Finally, KHG26702 reduced OGD-R-induced $A{\beta}$ production and cleavage of amyloid precursor protein, by inhibiting secretase activity and suppressing the autophagic pathway. Although supporting data from in vivo studies are required, our results indicate that KHG26702 may prevent neuronal cell damage from OGD-R-induced toxicity.

LncRNA AC005332.7 Inhibited Ferroptosis to Alleviate Acute Myocardial Infarction Through Regulating miR-331-3p/CCND2 Axis

  • Rixin Dai;Xiheng Yang;Wujin He;Qiang Su;Xuexin Deng;Juanfen Li
    • Korean Circulation Journal
    • /
    • v.53 no.3
    • /
    • pp.151-167
    • /
    • 2023
  • Background and Objectives: Acute myocardial infarction (AMI) often occurs suddenly and leads to fatal consequences. Ferroptosis is closely related to the progression of AMI. However, the specific mechanism of ferroptosis in AMI remains unclear. Methods: We constructed a cell model of AMI using AC16 cells under oxygen and glucose deprivation (OGD) conditions and a mice model of AMI using the left anterior descending (LAD) ligation. The 3-(4, 5-dimethylthiazol-2-yl)-2, 5 diphenyltetrazolium bromide was employed to determine cell viability. The levels of lactate dehydrogenase, creatine kinase, reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and iron were measured using corresponding kits. Dual luciferase reporter gene assay, RNA-binding protein immunoprecipitation, and RNA pull-down were performed to validate the correlations among AC005332.7, miR-331-3p, and cyclin D2 (CCND2). Hematoxylin and eosin staining was employed to evaluate myocardial damage. Results: AC005332.7 and CCND2 were lowly expressed, while miR-331-3p was highly expressed in vivo and in vitro models of AMI. AC005332.7 sufficiency reduced ROS, MDA, iron, and ACSL4 while boosting the GSH and GPX4, indicating that AC005332.7 sufficiency impeded ferroptosis to improve cardiomyocyte injury in AMI. Mechanistically, AC005332.7 interacted with miR-331-3p, and miR-331-3p targeted CCND2. Additionally, miR-331-3p overexpression or CCND2 depletion abolished the suppressive impact of AC005332.7 on ferroptosis in OGD-induced AC16 cells. Moreover, AC005332.7 overexpression suppressed ferroptosis in mice models of AMI. Conclusions: AC005332.7 suppressed ferroptosis in OGD-induced AC16 cells and LAD ligation-operated mice through modulating miR-331-3p/CCND2 axis, thereby mitigating the cardiomyocyte injury in AMI, which proposed novel targets for AMI treatment.

Role of Nitric Oxide in Ischemia-evoked Release of Norepinephrine from Rat Cortex Slices (흰쥐 대뇌피질 절편에서 허혈에 의한 Norepinephrine 유리에 있어서 Nitric Oxide의 영향)

  • Eun, Young-Ah;Kim, Dong-Chan;Cho, Kyu-Park;Kim, Kee-Won
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.1 no.6
    • /
    • pp.673-679
    • /
    • 1997
  • It has been generally accepted that glutamate mediates the ischemic brain damage, excitotoxicity, and induces release of neurotransmitters, including norepinephrine(NE), in ischemic milieu. In the present study, the role of nitric oxide(NO) in the ischemia-induced $[^3H]norepinephrine([^3H]NE)$ release from cortex slices of the rat was examined. Ischemia, deprivation of oxygen and glucose from $Mg^{2+}-free$ artificial cerebrospinal fluid, induced significant release of $[^3H]NE$ from cortex slices. This ischemia-induced $[^3H]NE$ release was significantly attenuated by glutamatergic neurotransmission modifiers. $N^G-nitro-L-arginine$ methyl ester(L-NAME), $N^G-monomethyl-L-arginine$ (L-NMMA) or 7-nitroindazole, nitric oxide synthase inhibitors attenuated the ischemia-evoked $[^3H]NE$ release. Hemoglobin, a NO chelator, and 5, 5- dimethyl-L-pyrroline-N-oxide(DMPO), an electron spin trap, inhibited $[^3H]NE$ release dose-dependently. Ischemia-evoked $[^3H]NE$ release was inhibited by methylene blue, a soluble guanylate cyclase inhibitor, and potentiated by 8-bromo-cGMP, a cell permeable cGMP analog, zaprinast, a cGMP phosphodiesterase inhibitor, and S-nitroso-N-acetylpenicillamine (SNAP), a nitric oxide generator. These results suggest that the ischemia-evoked $[^3H]NE$ release is mediated by NMDA receptors, and activation of NO system is involved.

  • PDF

Cobalt Chloride-induced Hypoxia Ameliorates NLRP3-Mediated Caspase-1 Activation in Mixed Glial Cultures

  • Kim, Eun-Hee;Won, Ji-Hee;Hwang, Inhwa;Yu, Je-Wook
    • IMMUNE NETWORK
    • /
    • v.13 no.4
    • /
    • pp.141-147
    • /
    • 2013
  • Hypoxia has been shown to promote inflammation, including the release of proinflammatory cytokines, but it is poorly investigated how hypoxia directly affects inflammasome signaling pathways. To explore whether hypoxic stress modulates inflammasome activity, we examined the effect of cobalt chloride ($CoCl_2$)-induced hypoxia on caspase-1 activation in primary mixed glial cultures of the neonatal mouse brain. Unexpectedly, hypoxia induced by oxygen-glucose deprivation or $CoCl_2$ treatment failed to activate caspase-1 in microglial BV-2 cells and primary mixed glial cultures. Of particular interest, $CoCl_2$-induced hypoxic condition considerably inhibited NLRP3-dependent caspase-1 activation in mixed glial cells, but not in bone marrow-derived macrophages. $CoCl_2$-mediated inhibition of NLRP3 inflammasome activity was also observed in the isolated brain microglial cells, but $CoCl_2$ did not affect poly dA:dT-triggered AIM2 inflammasome activity in mixed glial cells. Our results collectively demonstrate that $CoCl_2$-induced hypoxia may negatively regulate NLRP3 inflammasome signaling in brain glial cells, but its physiological significance remains to be determined.

Effect of Chungpaesagan-tang on Ischemic Damage in Organotypic Hippocampal Slice Culture (청폐사간탕(淸肺瀉肝湯)이 뇌해마 조직배양의 신경세포 자연사에 미치는 영향)

  • Lee, Min-Young;Ku, Ja-Seung;Kim, Sung-Hoon;Kim, Yoon-Bum;Kim, Sun-Yeou;Choi, Hyeon;Sohn, Young-Joo;Jung, Hyuk-Sang;Sohn, Nak-Won
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.22 no.4
    • /
    • pp.771-777
    • /
    • 2008
  • Chungpaesagan-tang which is used for treating patients of brain in cerebrovascular disease frequently from clinical doctor has not reported about the effect of neuronal aptosis caused of brain ischemia. The aim of this study is to investigate effect of Chungpaesagan-tang protecting neuronal cells from being damaged by brain ischemia through using organotypic hippocampal slice cultures. We caused ischemic damage to organotypic hippocampal slice cultures by oxygen and glucose deprivation. And added Chungpaesagan-tang extract to cultures. thereafter we measured area percentage of propidium iodide (PI)-stained neuronal cell, lactate dehydrogenase (LDH) levels in culture media and Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells. Area percentage of PI-stained neuronal cells and count of TUNEL-positive cells in CA1 and DG area of organotypic hippocampal slice culture were significantly decreased in pertinent density level of Chungpaesagan-tang extract. LDH levels in culture media of organotypic hippocampal slice culture were significantly decreased in pertinent density level of Chungpaesagan-tang extract. Within pertinent density level, Chungpaesagan-tang has cell protection effect that prevents brain ischemia damaging neuronal cells and apoptosis increasing.

Eugenol Inhibits Excitotoxins-Induced Delayed Neurotoxicity, Oxidative Injury and Convulsion

  • Wie, Myung-Bok;Cheon, Byung-Hwa;Lee, Seon-Young;Son, Kun-Ho;Song, Dong-Keun;Shin, Tae-Kyun;Kim, Hyoung-Chun
    • Toxicological Research
    • /
    • v.22 no.3
    • /
    • pp.275-282
    • /
    • 2006
  • In previous our studies, we have reported that eugenol derived from Eugenia caryophyllata(Myrtaceace) exhibits acute N-methyl-D-aspartate(NMDA)- and oxygen/glucose deprivation-induced neurotoxicity in primary cortical cultures and protects hippocampal neurons from global ischemia. In this study, we investigated whether the extracts and fractions of E. caryophyllata or eugenol shows the neuroprotective effects against delayed neuronal injury evoked by NMDA or ${\alpha}$-amino-3-hydroxy-5-methylisoxazole propionate(AMPA), and oxidative damage induced by arachidonic acid-, hydrogen peroxide-, $FeCl_2$/ascorbic acid-, and buthionine sulfoximine(BSO) in primary cortical cultures. We examined the neurotoxicity of eugenol itself in cultures and inhibitory effect of eugenol on NMDA- or kainate(KA)-induced convulsion in BALB/c mice. Each water, methanol extract and methanol fraction of E. caryophyllata was significantly attenuated NMDA-induced delayed neurotoxicity, respectively. Eugenol exhibited a significant inhibitory action against the convulsion evoked by NMDA and KA, and reduced delayed or brief neurotoxicity induced by NMDA, AMPA, and various oxidative injuries. These results suggest that eugenol derived from E. caryophyllata may contribute the neuroprotection against delayed-type excitotoxicity and excitotoxins-mediated convulsion through the amelioration of oxidative stress.

A Study of Neuroproctective Effect of Bupleuri Radix on Hippocampal Neurons (시호(柴胡)의 뇌해마 신경세포 보호효능에 대한 연구)

  • Lee, Won-Chul;Shin, Kwang-Sik
    • The Journal of Internal Korean Medicine
    • /
    • v.25 no.4
    • /
    • pp.227-241
    • /
    • 2004
  • Objective : This study was performed to investigate neuroprotective effects of Bupleuri Radix against oxidative and ischemic damages. Method : To observe the neuroprotective effects against ischemic damage, ischemic insult was induced by oxygen/glucose deprivation (OGD) on organotypic hippocampal slice cultures (OHSC) from 1 week-old Sprague-Dawley rats. Propidium iodide (PI) fluorescence-stained neuronal dead-cell areas, area percentages and TUNEL-positive apoptotic cells in CA1 and dentate gyrus, and LDH levels in culture media of the OHSC were measured following Bupleuri Radix extract treatment. Result : The following results were obtained: (1) The $5\;{\mu}g/ml$ of Bupleuri Radix treatment demonstrated a significant decrease in PI fluorescence-stained neuronal dead-cell areas and area percentage in CA1 region of the OHSC from 18 hrs to 48 hrs following the OGD. The $50\;{\mu}g/ml$ of Bupleuri Radix treatment was also significant from 6 hrs to 48 hrs following the OGD and was more effective. (2) The 5 and $50\;{\mu}g/ml$ of Bupleuri Radix treatment demonstrated a significant decrease in PI fluorescence-stained neuronal dead-cell areas and area percentage in DG region of the OHSC from 6 hrs to 48 hrs following the OGD. The $50\;{\mu}g/ml$ treatment was more effective than the $5\;{\mu}g/ml$ treatment. (3) Bupleuri Radix treatment demonstrated a significant decrease in TUNEL-positive apoptotic cells in CA1 region (with 5 and $50\;{\mu}g/ml$) and in DG region (with $50\;{\mu}g/ml$) of the OHSC damaged by the OGD. (4) Bupleuri Radix treatment demonstrated a significant decrease in LDH concentrations in culture media of the OHSC damaged by the OGD. Conclusion : These results suggest that Bupleuri Radix has neuroprotective and control effects on inflammatory and immune responses where there has been ischemic damage to the central nervous system.

  • PDF