• 제목/요약/키워드: Neuronal activation

검색결과 290건 처리시간 0.026초

Neuronal Nitric Oxide-mediated Cytotoxicity in Trophoblast Cells Induced by Increase of Intracellular Calcium

  • Shin, Mi-Kyung;Kwon, Yong-Hyun;Shin, Jong-Chul;Yang, Dong-Eun;Lee, Sung-Keun;Kang, Ju-Hee;Park, Chang-Shin
    • Molecular & Cellular Toxicology
    • /
    • 제4권1호
    • /
    • pp.16-21
    • /
    • 2008
  • Cell death of trophoblast, particularly by abnormal release of physiological nitric oxide (NO) has been known to be a causative factor of pre-eclampsia. In the present study, effects of intracellular calcium increase enhancing the activity of NO synthases (neuronal NO synthase, nNOS in this trophoblast cells) on the cell death were examined in a human placental full-term cell line (HT-1). Furthermore, we analyzed the possible mechanisms underlying the augmentation of $Ca^{++}$-mediated NOS activity mediated by protein kinases like PKC, PKA, or CaM-KII. In experiments for cell toxicity, a calcium ionophore (ionomycin $10{\mu}M$) enhanced cell death confirmed by MTT assay, and increased significantly nNOS activity determined with a hemoglobin oxidation assay. This cell death was partially protected by pre-treatment of 7-nitroindazole (7-NI, $10{\mu}M$ and $100{\mu}M$), a nNOS-specific inhibitor. Additionally, $Ca^{++}$-ionophore -induced increase of nNOS activity also was partially normalized by pre-treatment of specific inhibitors of protein kinases, PKC, PKA or CaM-KII. Therefore, we suggest that an increase of calcium influx, leading to the activation of nNOS activity, which in turn may result in the death of trophoblast cells by involvement of signaling mechanisms of protein kinases.

Coupling Efficiencies of m1, m3 and m5 Muscarinic Receptors to the Stimulation of Neuronal Nitric Oxide Synthase

  • Park, Sun-Hye;Lee, Seok-Yong;Cho, Tai-Soon
    • 한국응용약물학회:학술대회논문집
    • /
    • 한국응용약물학회 1996년도 춘계학술대회
    • /
    • pp.207-207
    • /
    • 1996
  • Through molecular cloning, five muscarinic receptors have been identified. The muscarinic receptors can be generally grouped according to their coupling to either stimulation of phospholipase C (m1, m3, and m5) or the inhibition of adenylate cyclase (m2 and m4). Each m1, m3, and m5 receptors has the additional potential to couple to the activation of phospholipase A$_2$, C, and D, tyrosine kinase, and the mobilization of Ca$\^$2+/. However, the differences in coupling efficiencies to different second messenger systems between these receptors have not been studied well. Ectopic expression of each of these receptors in mammalian cells has provided the opportunity to evaluate the signal transduction of each in some detail. In this work we compared the coupling efficiencies of the m1, m3 and m5 muscarinic receptors expressed in chinese hamster ovary (CHO) cells to the Ca$\^$2+/ mobilization and the stimulation of neuronal nitric oxide synthase (nNOS). Because G protein/PLC/PI turnover/[(Ca$\^$2+/])i/NOS pathway was supposed as a main pathway for the production of nitric oxide via muscarinic receptors, we studied on ml, m3 and m5 receptors. Stimulation of guanylate cyclase activity in detector neuroblastoma cells was used as an index of generation nitric oxide (NO) in CHO cells. The agonist carbachol increased the cGMP formation and the intracellular [Ca$\^$2+/] in concentration dependent manner in three types of receptors and the increased cGMP formation was significantly attenuated by scavenger of NO or inhibitor of NOS. m5 receptors was most efficiently coupled to stimulation of nNOS, And, the coupling efficiencies to the stimulation of neuronal nitric oxide synthase in three types of receptors were parallel with them to the Ca$\^$2+/ mobilization.

  • PDF

The Effect of Morphine on REST Expression in Human Neuroblastoma NMB Cells

  • Kim, Do-Kyung;Kim, Chun-Sung;Kim, Heung-Joong;Kook, Joong-Ki;Kim, Seung-Hee;Lee, Baek-Hee;Lee, Yun-Ho;Mo, Shin-Yeob;Loh, Horace H.
    • International Journal of Oral Biology
    • /
    • 제35권2호
    • /
    • pp.69-74
    • /
    • 2010
  • The mu opioid receptor (MOR) has been regarded as the main site of interaction with analgesics in major clinical use, particularly morphine. The repressor element-1 silencing transcription factor (REST) functions as a transcriptional repressor of neuronal genes in non-neuronal cells. However, it is expressed in certain mature neurons, suggesting that it may have complex and novel roles. In addition, the interactions between MOR and REST and their functions remain unclear. In this study, we examined the effects of morphine on the expression of REST mRNA and protein in human neuroblastoma NMB cells to investigate the roles of REST induced by MOR activation in neuronal cells. To determine the effects of morphine on REST expression, we performed RT-PCR, real-time quantitative RT-PCR, western blot analysis and radioligand binding assays in NMB cells. By RTPCR and real-time quantitative RT-PCR, the expression of REST was found to be unchanged by either the MOR agonist morphine or the MOR specific antagonist CTOP. By western blot, morphine was shown to significantly inhibit the expression of REST, but this suppression was completely blocked by treatment with CTOP. In the radioligand binding assay, the overexpression of REST led to an increased opioid ligand binding activity of endogenous MOR in the NMB cells. These results together suggest that morphine inhibits the expression of REST in human neuroblastoma cells through a post-transcriptional regulatory mechanism mediated through MOR.

뇌허혈로 인한 흰쥐에서의 기억력 및 학습효과 저해에 대한 황금의 보호효과 (Protective effects of Scutellariae Radix on impairments in learning and memory induced by brain ischemia in rats)

  • 김영옥;이세나;김명규;부영민;김선여;김호철;임강현
    • 대한본초학회지
    • /
    • 제21권4호
    • /
    • pp.189-195
    • /
    • 2006
  • Objectives : It has been reported previously that the roots of Scutellaria baicalensis (known as Huang-Gum in Korean, henceforth referred to as S. baicalensis) could prevent neuronal cell death after global cerebral ischemia. In Genuine Korean medicine, S. baicalensis is known to relieve fever in upper body, and it was thus thought to be able to alleviate deteriorations in brain function. Methods : The protective effects of S. baicalensis against post-stroke memory retardation using 4-vessel occlusion model were examined in the present study. Results : S. baicalensis was shown to significantly alleviate the deficits in learning and memory by increasing the fraction of time spent in the quadrant in which the platform was initially placed ($34.9\;{\pm}\;3.2%$, p < 0.05) compared to that of the ischemia group ($28.0\;{\pm}\;2.5%$). The cytoprotective effect of S. baicalensis on CA1 hippocampal neurons was evaluated by measuring the neuronal cell density. Neuronal cell density in S. baicalensis extracts-treated ischemia group ($138.0\;{\pm}\;13.6\;cells/mm^2$) was significantly increased compared to saline-treated ischemia group ($22.1 \;{\pm}\;9.3\;cells/mm^2$, p < 0.05). In the study of OX-42 immunohistochemistry, S. baicalensis could decrease the micrgial activation in hippocampus after brain ischemia. Conclusion : These results may provide experimental support for the use of S. baicalensis in treating post-stroke memory impairment.

  • PDF

Bark Constituents from Mushroom-detoxified $Rhus$ $verniciflua$ Suppress Kainic Acid-induced Neuronal Cell Death in Mouse Hippocampus

  • Byun, Jong-Seon;Han, Yoon-Hee;Hong, Sung-Jun;Hwang, Sung-Mi;Kwon, Yong-Soo;Lee, Hee-Jae;Kim, Sung-Soo;Kim, Myong-Jo;Chun, Wan-Joo
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제14권5호
    • /
    • pp.279-283
    • /
    • 2010
  • Urushinol, a plant allergen, has significantly restricted the medical application of $Rhus$ $verniciflua$, although it has been reported to possess a wide variety of biological activities such as anti-inflammatory, antioxidant, and anti-cancer actions. To reduce the urushinol content while maintaining the beneficial biological activities, mushroom-mediated fermentation of $Rhus$ $verniciflua$ was carried out and this method resulted in significantly attenuated allergenicity [1]. In the present study, to examine the neuroprotective properties of mushroom-fermented stem bark of $Rhus$ $verniciflua$, two constituents were isolated from mushroom-fermented bark and their neuroprotective properties were examined in a mouse model of kainic acid (KA)-induced excitotoxicity. KA resulted in significant apoptotic neuronal cell death in the CA3 region of mouse hippocampus. However, seven daily administrations of RVH-1 or RVH-2 prior to KA injection significantly attenuated KA-induced pyramidal neuronal cell death in the CA3 region. Furthermore, pretreatment with RVH-1 and RVH-2 also suppressed KA-induced microglial activation in the mouse hippocampus. The present study demonstrates that RVH-1 and RVH-2 isolated from $Rhus$ $verniciflua$ and detoxified using mushroom species possess neuroprotective properties against KA-induced excitotoxicity. This leads to the possibility that detoxified $Rhus$ $verniciflua$ can be a valuable asset in herbal medicine.

Genistein attenuates isoflurane-induced neurotoxicity and improves impaired spatial learning and memory by regulating cAMP/CREB and BDNF-TrkB-PI3K/Akt signaling

  • Jiang, Tao;Wang, Xiu-qin;Ding, Chuan;Du, Xue-lian
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제21권6호
    • /
    • pp.579-589
    • /
    • 2017
  • Anesthetics are used extensively in surgeries and related procedures to prevent pain. However, there is some concern regarding neuronal degeneration and cognitive deficits arising from regular anesthetic exposure. Recent studies have indicated that brain-derived neurotrophic factor (BDNF) and cyclic AMP response element-binding protein (CREB) are involved in learning and memory processes. Genistein, a plant-derived isoflavone, has been shown to exhibit neuroprotective effects. The present study was performed to examine the protective effect of genistein against isoflurane-induced neurotoxicity in rats. Neonatal rats were exposed to isoflurane (0.75%, 6 hours) on postnatal day 7 (P7). Separate groups of rat pups were orally administered genistein at doses of 20, 40, or 80 mg/kg body weight from P3 to P15 and then exposed to isoflurane anesthesia on P7. Neuronal apoptosis was detected by TUNEL assay and FluoroJade B staining following isoflurane exposure. Genistein significantly reduced apoptosis in the hippocampus, reduced the expression of proapoptotic factors (Bad, Bax, and cleaved caspase-3), and increased the expression of Bcl-2 and Bcl-xL. RT-PCR analysis revealed enhanced BDNF and TrkB mRNA levels. Genistein effectively upregulated cAMP levels and phosphorylation of CREB and TrkB, leading to activation of cAMP/CREB-BDNF-TrkB signaling. PI3K/Akt signaling was also significantly activated. Genistein administration improved general behavior and enhanced learning and memory in the rats. These observations suggest that genistein exerts neuroprotective effects by suppressing isoflurane-induced neuronal apoptosis and by activating cAMP/CREB-BDNF-TrkB-PI3/Akt signaling.

7,8-Dihydroxyflavone Protects High Glucose-Damaged Neuronal Cells against Oxidative Stress

  • Cho, Suk Ju;Kang, Kyoung Ah;Piao, Mei Jing;Ryu, Yea Seong;Fernando, Pincha Devage Sameera Madushan;Zhen, Ao Xuan;Hyun, Yu Jae;Ahn, Mee Jung;Kang, Hee Kyoung;Hyun, Jin Won
    • Biomolecules & Therapeutics
    • /
    • 제27권1호
    • /
    • pp.85-91
    • /
    • 2019
  • Oxidative stress is considered a major contributor in the pathogenesis of diabetic neuropathy and in diabetes complications, such as nephropathy and cardiovascular diseases. Diabetic neuropathy, which is the most frequent complications of diabetes, affect sensory, motor, and autonomic nerves. This study aimed to investigate whether 7,8-dihydroxyflavone (7,8-DHF) protects SH-SY5Y neuronal cells against high glucose-induced toxicity. In the current study, we found that diabetic patients exhibited higher lipid peroxidation caused by oxidative stress than healthy subjects. 7,8-DHF exhibits superoxide anion and hydroxyl radical scavenging activities. High glucose-induced toxicity severely damaged SH-SY5Y neuronal cells, causing mitochondrial depolarization; however, 7,8-DHF recovered mitochondrial polarization. Furthermore, 7,8-DHF effectively modulated the expression of pro-apoptotic protein (Bax) and anti-apoptotic protein (Bcl-2) under high glucose, thus inhibiting the activation of caspase signaling pathways. These results indicate that 7,8-DHF has antioxidant effects and protects cells from apoptotic cell death induced by high glucose. Thus, 7,8-DHF may be developed into a promising candidate for the treatment of diabetic neuropathy.

전뇌 허혈성 흰쥐 모델에서 mBHT의 신경보호효과 연구 (Neuroprotective effect of modify Bo-Yang-Hwan-O-Tang on global ischemia in rat)

  • 오태우;박용기
    • 대한본초학회지
    • /
    • 제27권6호
    • /
    • pp.83-90
    • /
    • 2012
  • Objectives : Modified Bo-Yang-Hwan-O-Tang (mBHT) is a polyherbal medicine of twelve herbs traditionally used in the treatment of cerebral and cardiac stroke and vascular dementia. The purpose of this study was to evaluate the neuroprotective effect, pyramidal neuronal cell, inflammation and apoptosis of mBHT against global ischemia in rats. Methods : Global ischemia was produced by two-vessel occlusion(2-VO) in SD male rats. mBHT at dose of 500 mg/kg was orally administrated for 2 weeks or 6 weeks after global ischemia. The histopathological changes of ischemic brain were observed by staining of hematoxylin and eosin (H&E) and Nissl and immunohistochemisty with anti-GFAP (glial fibrillary acidic protein) antibody as a astrocyte marker. The expression of inducible nitric oxide synthase (iNOS) and apoptotic proteins such as Bax, Bcl-2 and caspase-3 was determined by western blot. Results : mBHT treatment significantly inhibited the pyramidal neuronal loss in CA1 of hippocampus of global ischemic rats by 2-VO. mBHT also suppressed the activation of astrocytes in the CA1 at 6 weeks after ischemia. In addition, mBHT significantly increased the expression of anti-apoptotic protein, Bcl-2 on iscemic brain, and significantly attenuated the expression of apoptotic proteins, Bax and caspase-3. Conclusions : These results indicate that mBHT inhibits neuronal cell damage induced in global ischemia by 2-VO, suggesting that mBHT may be a potential candidate for the treatment of vascular dementia.

Ginsenoside compound K reduces ischemia/reperfusion-induced neuronal apoptosis by inhibiting PTP1B-mediated IRS1 tyrosine dephosphorylation

  • Jing, Fu;Liang, Yu;Qian, Yu;Nengwei, Yu;Fei, Xu;Suping, Li
    • Journal of Ginseng Research
    • /
    • 제47권2호
    • /
    • pp.274-282
    • /
    • 2023
  • Background: Ginsenoside compound K (CK) stimulated activation of the PI3K-Akt signaling is one of the major mechanisms in promoting cell survival after stroke. However, the underlying mediators remain poorly understood. This study aimed to explore the docking protein of ginsenoside CK mediating the neuroprotective effects. Materials and methods: Molecular docking, surface plasmon resonance, and cellular thermal shift assay were performed to explore ginsenoside CK interacting proteins. Neuroscreen-1 cells and middle cerebral artery occlusion (MCAO) model in rats were utilized as in-vitro and in-vivo models. Results: Ginsenoside CK interacted with recombinant human PTP1B protein and impaired its tyrosine phosphatase activity. Pathway and process enrichment analysis confirmed the involvement of PTP1B and its interacting proteins in PI3K-Akt signaling pathway. PTP1B overexpression reduced the tyrosine phosphorylation of insulin receptor substrate 1 (IRS1) after oxygen-glucose deprivation/reoxygenation (OGD/R) in neuroscreen-1 cells. These regulations were confirmed in the ipsilateral ischemic hemisphere of the rat brains after MCAO/R. Ginsenoside CK treatment reversed these alterations and attenuated neuronal apoptosis. Conclusion: Ginsenoside CK binds to PTP1B with a high affinity and inhibits PTP1B-mediated IRS1 tyrosine dephosphorylation. This novel mechanism helps explain the role of ginsenoside CK in activating the neuronal protective PI3K-Akt signaling pathway after ischemia-reperfusion injury.

Ginsenoside Rg1 alleviates Aβ deposition by inhibiting NADPH oxidase 2 activation in APP/PS1 mice

  • Zhang, Han;Su, Yong;Sun, Zhenghao;Chen, Ming;Han, Yuli;Li, Yan;Dong, Xianan;Ding, Shixin;Fang, Zhirui;Li, Weiping;Li, Weizu
    • Journal of Ginseng Research
    • /
    • 제45권6호
    • /
    • pp.665-675
    • /
    • 2021
  • Background: Ginsenoside Rg1 (Rg1), an active ingredient in ginseng, may be a potential agent for the treatment of Alzheimer's disease (AD). However, the protective effect of Rg1 on neurodegeneration in AD and its mechanism of action are still incompletely understood. Methods: Wild type (WT) and APP/PS1 AD mice, from 6 to 9 months old, were used in the experiment. The open field test (OFT) and Morris water maze (MWM) were used to detect behavioral changes. Neuronal damage was assessed by hematoxylin and eosin (H&E) and Nissl staining. Immunofluorescence, western blotting, and quantitative real-time polymerase chain reaction (q-PCR) were used to examine postsynaptic density 95 (PSD95) expression, amyloid beta (Aβ) deposition, Tau and phosphorylated Tau (p-Tau) expression, reactive oxygen species (ROS) production, and NAPDH oxidase 2 (NOX2) expression. Results: Rg1 treatment for 12 weeks significantly ameliorated cognitive impairments and neuronal damage and decreased the p-Tau level, amyloid precursor protein (APP) expression, and Aβ generation in APP/PS1 mice. Meanwhile, Rg1 treatment significantly decreased the ROS level and NOX2 expression in the hippocampus and cortex of APP/PS1 mice. Conclusions: Rg1 alleviates cognitive impairments, neuronal damage, and reduce Aβ deposition by inhibiting NOX2 activation in APP/PS1 mice.