• Title/Summary/Keyword: Neuronal activation

검색결과 289건 처리시간 0.028초

Clostridium difficile Toxin A Induces Reactive Oxygen Species Production and p38 MAPK Activation to Exert Cellular Toxicity in Neuronal Cells

  • Zhang, Peng;Hong, Ji;Yoon, I Na;Kang, Jin Ku;Hwang, Jae Sam;Kim, Ho
    • Journal of Microbiology and Biotechnology
    • /
    • 제27권6호
    • /
    • pp.1163-1170
    • /
    • 2017
  • Clostridium difficile releases two exotoxins, toxin A and toxin B, which disrupt the epithelial cell barrier in the gut to increase mucosal permeability and trigger inflammation with severe diarrhea. Many studies have suggested that enteric nerves are also directly involved in the progression of this toxin-mediated inflammation and diarrhea. C. difficile toxin A is known to enhance neurotransmitter secretion, increase gut motility, and suppress sympathetic neurotransmission in the guinea pig colitis model. Although previous studies have examined the pathophysiological role of enteric nerves in gut inflammation, the direct effect of toxins on neuronal cells and the molecular mechanisms underlying toxin-induced neuronal stress remained to be unveiled. Here, we examined the toxicity of C. difficile toxin A against neuronal cells (SH-SY5Y). We found that toxin A treatment time- and dose-dependently decreased cell viability and triggered apoptosis accompanied by caspase-3 activation in this cell line. These effects were found to depend on the up-regulation of reactive oxygen species (ROS) and the subsequent activation of p38 MAPK and induction of $p21^{Cip1/Waf1}$. Moreover, the N-acetyl-$\text\tiny L$-cysteine (NAC)-induced down-regulation of ROS could recover the viability loss and apoptosis of toxin A-treated neuronal cells. These results collectively suggest that C. difficile toxin A is toxic for neuronal cells, and that this is associated with rapid ROS generation and subsequent p38 MAPK activation and $p21^{Cip1/Waf1}$ up-regulation. Moreover, our data suggest that NAC could inhibit the toxicity of C. difficile toxin A toward enteric neurons.

Kainic Acid-induced Neuronal Death is Attenuated by Aminoguanidine but Aggravated by L-NAME in Mouse Hippocampus

  • Byun, Jong-Seon;Lee, Sang-Hyun;Jeon, Seong-Ho;Kwon, Yong-Soo;Lee, Hee-Jae;Kim, Sung-Soo;Kim, Young-Myeong;Kim, Myong-Jo;Chun, Wan-Joo
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제13권4호
    • /
    • pp.265-271
    • /
    • 2009
  • Nitric oxide (NO) has both neuroprotective and neurotoxic effects depending on its concentration and the experimental model. We tested the effects of NG-nitro-L-arginine methyl ester (L-NAME), a nonselective nitric oxide synthase (NOS) inhibitor, and aminoguanidine, a selective inducible NOS (iNOS) inhibitor, on kainic acid (KA)-induced seizures and hippocampal CA3 neuronal death. L-NAME (50 mg/kg, i.p.) and/or aminoguanidine (200 mg/kg, i.p.) were administered 1 h prior to the intracerebroventricular (i.c.v.) injection of KA. Pretreatment with L-NAME significantly increased KA-induced CA3 neuronal death, iNOS expression, and activation of microglia. However, pretreatment with aminoguanidine significantly suppressed both the KA-induced and L-NAME-aggravated hippocampal CA3 neuronal death with concomitant decreases in iNOS expression and microglial activation. The protective effect of aminoguanidine was maintained for up to 2 weeks. Furthermore, iNOS knockout mice ($iNOS^{-1-}$) were resistant to KA-induced neuronal death. The present study demonstrates that aminoguanidine attenuates KA-induced neuronal death, whereas L-NAME aggravates neuronal death, in the CA3 region of the hippocampus, suggesting that NOS isoforms play different roles in KA-induced excitotoxicity.

Effects of Glutamate Receptor Antagonists and Protein Synthesis Inhibitor on Delayed Neuronal Death Induced by Transient Global Ischemia in Rat Brain

  • Ko, Jun-Seog;Bae, Choon-Sang;Kim, Jong-Keun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제2권3호
    • /
    • pp.279-286
    • /
    • 1998
  • It has been well documented that transient forebrain global ischemia causes selective neuronal degeneration in hippocampal CA1 pyramidal neurons with a delay of a few days. The mechanism of this delayed hippocampal CA1 pyramidal neuronal death (DND) is still controversial. To delineate the mechanisms of the DND, the effects of treatment with MK-801, an NMDA receptor antagonist, kynurenic acid, a NMDA/non-NMDA receptor antagonist, and/or cycloheximide, a protein synthesis inhibitor, on the DND were investigated in male Wistar rats. To examine the participation of apoptotic neuronal death in the DND, TUNEL staining was performed in ischemic brain section. Global ischemia was induced by 4-vessel occlusion for 20 min. All animals in this study showed the DND 3 and 7 days after the ischemic insult. The DND that occured 3 days and 7 days after the ischemia were not affected by pretreatment with MK-801 (1 mg/kg), but markedly attenuated by the pretreatment with kynurenic acid (500 mg/kg). Treatment with cycloheximide (1 mg/kg) also markedly inhibited the DND. The magnitudes of attenuation by the two drugs were similar. The magnitude of attenuation by co-treatments with kynurenic acid and cycloheximide was not greater than that with any single treatment. TUNEL staining was negative in the sections obtained 1 or 2 days after the ischemic insults, but it was positive at hippocampal CA1 pyramidal cells in sections collected 3 days after the ischemia. These results suggested that the DND should be mediated by the activation of non-NMDA receptor, not by the activation of NMDA receptor and that the activation of AMPA receptor should induce the apoptotic process in the DND.

  • PDF

Cellular and Molecular Pathways of Ischemic Neuronal Death

  • Won, Seok-Joon;Kim, Doo-Yeon;Gwag, Byoung-Joo
    • BMB Reports
    • /
    • 제35권1호
    • /
    • pp.67-86
    • /
    • 2002
  • Three routes have been identified triggering neuronal death under physiological and pathological conditions. Excess activation of ionotropic glutamate receptors cause influx and accumulation of $Ca^{2+}$ and $Na^+$ that result in rapid swelling and subsequent neuronal death within a few hours. The second route is caused by oxidative stress due to accumulation of reactive oxygen and nitrogen species. Apoptosis or programmed cell death that often occurs during developmental process has been coined as additional route to pathological neuronal death in the mature nervous system. Evidence is being accumulated that excitotoxicity, oxidative stress, and apoptosis propagate through distinctive and mutually exclusive signal transduction pathway and contribute to neuronal loss following hypoxic-ischemic brain injury. Thus, the therapeutic intervention of hypoxic-ischemic neuronal injury should be aimed to prevent excitotoxicity, oxidative stress, and apoptosis in a concerted way.

Odorant Stimulation Promotes Survival of Rodent Olfactory Receptor Neurons via PI3K/Akt Activation and Bcl-2 Expression

  • Kim, So Yeun;Yoo, Seung-Jun;Ronnett, Gabriele V;Kim, Eun-Kyoung;Moon, Cheil
    • Molecules and Cells
    • /
    • 제38권6호
    • /
    • pp.535-539
    • /
    • 2015
  • Olfactory stimulation activates multiple signaling cascades in order to mediate activity-driven changes in gene expression that promote neuronal survival. To date, the mechanisms involved in activity-dependent olfactory neuronal survival have yet to be fully elucidated. In the current study, we observed that olfactory sensory stimulation, which caused neuronal activation, promoted activation of the phosphatidylinositol 3'-kinase (PI3K)/Akt pathway and the expression of Bcl-2, which were responsible for olfactory receptor neuron (ORN) survival. We demonstrated that Bcl-2 expression increased after odorant stimulation both in vivo and in vitro. We also showed that odorant stimulation activated Akt, and that Akt activation was completely blocked by incubation with both a PI3K inhibitor (LY294002) and Akt1 small interfering RNA. Moreover, blocking the PI3K/Akt pathway diminished the odorantinduced Bcl-2 expression, as well as the effects on odorant-induced ORN survival. A temporal difference was noted between the activation of Akt1 and the expression of Bcl-2 following odorant stimulation. Blocking the PI3K/Akt pathway did not affect ORN survival in the time range prior to the increase in Bcl-2 expression, implying that these two events, activation of the PI3K pathway and Bcl-2 induction, were tightly connected to promote post-translational ORN survival. Collectively, our results indicated that olfactory activity activated PI3K/Akt, induced Bcl-2, and promoted long term ORN survival as a result.

Sustained Intracellular Acidosis Triggers the Na+/H+ Exchager-1 Activation in Glutamate Excitotoxicity

  • Lee, Bo Kyung;Jung, Yi-Sook
    • Biomolecules & Therapeutics
    • /
    • 제25권6호
    • /
    • pp.593-598
    • /
    • 2017
  • The $Na^+/H^+$ exchanger-1 (NHE-1) is a ubiquitously expressed pH-regulatory membrane protein that functions in the brain, heart, and other organs. It is increased by intracellular acidosis through the interaction of intracellular $H^+$ with an allosteric modifier site in the transport domain. In the previous study, we reported that glutamate-induced NHE-1 phosphorylation mediated by activation of protein kinase C-${\beta}$ (PKC-${\beta}$) in cultured neuron cells via extracellular signal-regulated kinases (ERK)/p90 ribosomal s6 kinases (p90RSK) pathway results in NHE-1 activation. However, whether glutamate stimulates NHE-1 activity solely by the allosteric mechanism remains elusive. Cultured primary cortical neuronal cells were subjected to intracellular acidosis by exposure to $100{\mu}M$ glutamate or 20 mM $NH_4Cl$. After the desired duration of intracellular acidosis, the phosphorylation and activation of PKC-${\beta}$, ERK1/2 and p90RSK were determined by Western blotting. We investigated whether the duration of intracellular acidosis is controlled by glutamate exposure time. The NHE-1 activation increased while intracellular acidosis sustained for >3 min. To determine if sustained intracellular acidosis induced NHE-1 phosphorylation, we examined phosphorylation of NHE-1 induced by intracellular acidosis by transient exposure to $NH_4Cl$. Sustained intracellular acidosis led to activation and phosphorylation of NHE-1. In addition, sustained intracellular acidosis also activated the PKC-${\beta}$, ERK1/2, and p90RSK in neuronal cells. We conclude that glutamate stimulates NHE-1 activity through sustained intracellular acidosis, which mediates NHE-1 phosphorylation regulated by PKC-${\beta}$/ERK1/2/p90RSK pathway in neuronal cells.

Glial Mechanisms of Neuropathic Pain and Emerging Interventions

  • Jo, Daehyun;Chapman, C. Richard;Light, Alan R.
    • The Korean Journal of Pain
    • /
    • 제22권1호
    • /
    • pp.1-15
    • /
    • 2009
  • Neuropathic pain is often refractory to intervention because of the complex etiology and an incomplete understanding of the mechanisms behind this type of pain. Glial cells, specifically microglia and astrocytes, are powerful modulators of pain and new targets of drug development for neuropathic pain. Glial activation could be the driving force behind chronic pain, maintaining the noxious signal transmission even after the original injury has healed. Glia express chemokine, purinergic, toll-like, glutaminergic and other receptors that enable them to respond to neural signals, and they can modulate neuronal synaptic function and neuronal excitability. Nerve injury upregulates multiple receptors in spinal microglia and astrocytes. Microglia influence neuronal communication by producing inflammatory products at the synapse, as do astrocytes because they completely encapsulate synapses and are in close contact with neuronal somas through gap junctions. Glia are the main source of inflammatory mediators in the central nervous system. New therapeutic strategies for neuropathic pain are emerging such as targeting the glial cells, novel pharmacologic approaches and gene therapy. Drugs targeting microglia and astrocytes, cytokine production, and neural structures including dorsal root ganglion are now under study, as is gene therapy. Isoform-specific inhibition will minimize the side effects produced by blocking all glia with a general inhibitor. Enhancing the anti-inflammatory cytokines could prove more beneficial than administering proinflammatory cytokine antagonists that block glial activation systemically. Research on therapeutic gene transfer to the central nervous system is underway, although obstacles prevent immediate clinical application.

배양 대뇌피질 신경세포에서 glutamate에 의한 $Ca^{2+}$/calmodulin-dependent protein kinase IV의 활성변화 (Glutamate-induced Modulation of $Ca^{2+}$/Calmodulin-dependent Protein Kinase IV in Cultured Rat Cortical Neurons)

  • 조정숙
    • 약학회지
    • /
    • 제45권4호
    • /
    • pp.419-425
    • /
    • 2001
  • The neuronal cell death induced by excess glutamate (Glu) has been implicated in many acute and chronic neurodegenerative diseases including cerebral ischemia. Glu-induced elevation of intra-cellular $Ca^{2+}$ plays a critical role in the excitotoxicity, partly through the activation of a variety of $Ca^{2+}$ dependent enzymes. In the present study, we investigated the Glu-induced modulation of $Ca^{2+}$/calmodulin-dependent protein kinase IV (CaMK IV), a multifunctional enzyme abundantly present in the nuclei of neurons. The exposure of cultured rat cortical neurons to $100{\mu}$M Glu for 3 min dramatically increased CaMK IV activity up to 4.5-fold of the control-treated enzyme activity. The activation was very rapid, reaching peak at 3 min and then declined gradually. Under the same experimental conditions, time-dependent acute and delayed neuronal cell death was observed. Immunoblot analyses using specific antibodies showed that the expressions of CaMK IV and $CaMKK_{\alpha}$ were time-dependently modulated by Glu. Taken together, these results imply that the modulation of CaMK IV activity by Glu may be involved in the cascade of events resulting in neuronal cell death in cortical cultures.

  • PDF

신경세포 사멸과 미세아교세포활성화 억제 동시 가능 천연물질 탐색 연구 (A Screen for Dual-protection Molecules from a Natural Product Library against Neuronal Cell Death and Microglial Cell Activation)

  • 민주식;이동석
    • 생명과학회지
    • /
    • 제25권6호
    • /
    • pp.656-662
    • /
    • 2015
  • 천연물을 기반으로 한 신약 개발은 일반적으로 오랜 기간 동안의 원료 약물로써 사용해 온 경험에 의한 다양한 임상적 결과의 축적과 이로 인한 안정성(stability)과 안전성(safety)의 확보 및 신약 개발 시간의 단축과 같은 이점을 가지고 있어, 천연물 유래 약물 연구는 꼭 필요한 실정이다. 다양한 신경질환에서 신경세포의 사멸과 미세아교세포의 과도한 활성화 즉 뇌염증이 관찰되며 이를 억제할 수 있는 물질에 대한 연구는 활발히 진행 중이지만, 현재까지 신경세포 사멸과 뇌염증을 동시에 억제하는 물질 개발 시도는 거의 없었다. 따라서, 본 연구에서는 천연물에서 추출한 물질로 총 240개로 구성된 라이브러리로부터 신경전달물질 중의 하나인 glutamate 과잉처리에 의한 산화적 스트레스 유도 신경세포(HT22) 사멸과 LPS에 의한 미세아교세포(BV2)의 과도한 활성화 즉 뇌염증의 표지 인자 중 하나인 NO의 생산량의 감소 효과가 동시에 나타나는 물질을 검출한 결과, 대황에서 추출한 Chrysophanol이 검출되었으며 더욱이 Chrysophanol이 신경세포와 미세아교세포 모두에서 glutamate와 LPS에 의해 각각 유도된 세포내 활성산소(ROS) 발생을 억제하는 것을 확인하였다. 앞으로 Chrysophanol에 대한 보다 깊은 연구를 통하여 산화적 스트레스에 의한 신경세포 사멸과 미세아교세포의 과잉 활성화에 따른 뇌염증의 발생을 동시에 억제하는 신경질환의 치료 및 예방 신약개발 후보 물질 가능성을 제시 하고자 한다.

Signaling Pathways Controlling Microglia Chemotaxis

  • Fan, Yang;Xie, Lirui;Chung, Chang Y.
    • Molecules and Cells
    • /
    • 제40권3호
    • /
    • pp.163-168
    • /
    • 2017
  • Microglia are the primary resident immune cells of the central nervous system (CNS). They are the first line of defense of the brain's innate immune response against infection, injury, and diseases. Microglia respond to extracellular signals and engulf unwanted neuronal debris by phagocytosis, thereby maintaining normal cellular homeostasis in the CNS. Pathological stimuli such as neuronal injury induce transformation and activation of resting microglia with ramified morphology into a motile amoeboid form and activated microglia chemotax toward lesion site. This review outlines the current research on microglial activation and chemotaxis.