• Title/Summary/Keyword: neuronal loss

Search Result 163, Processing Time 0.023 seconds

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
    • /
    • v.27 no.6
    • /
    • 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.

Ginsenoside Rg3 from Red Ginseng Prevents Damage of Neuronal Cells through the Phosphorylation of the Cell Survival Protein Akt

  • Joo, Seong-Soo;Won, Tae-Joon;Lee, Yong-Jin;Hwang, Kwang-Woo;Lee, Seon-Gu;Yoo, Yeong-Min;Lee, Do-Ik
    • Food Science and Biotechnology
    • /
    • v.15 no.2
    • /
    • pp.244-247
    • /
    • 2006
  • Neuronal cell death significantly contributes to neuronal loss in neurological injury and disease. Typically, neuronal loss or destruction upon exposure to neurotoxins, oxidative stress, or DNA damage causes neurodegenerative diseases such as Alzheimer's disease. In this study, we attempted to determine whether ginsenoside Rg3 from red ginseng has a neuroprotective effect via an anti-apoptotic role induced by S-nitroso-N-acetylpenicillamine (SNAP) at the molecular level. We also investigated the antioxidant effect of Rg3 using a metal-catalyzed reaction with $Cu^{2+}/H_2O_2$. Our results showed that Rg3 ($40-100\;{\mu}g/mL$) protected SK-N-MC neuroblastoma cells under cytotoxic conditions and effectively protected DNA from fragmentation. In the signal pathway, caspase-3, and poly (ADP-ribose) polymerase (PARP) were kept at an inactivated status when pretreated with Rg3 in all ranges. In particular, the important upstream p-Akt signal pathway was increased in a dose-dependent manner, which indicates that Rg3 may contribute to cell survival. We also found that oxidative stress can be mitigated by Rg3. Therefore, we have concluded that Rg3 plays a certain role in neurodegenerative pathogenesis via an anti apoptotic, antioxidative effect.

Neuroprotective Effects of the Extracts from the Aerial Parts of Carthamus tinctorius L. on Transient Cerebral Global Ischemia in Rats (홍화 지상부 추출물의 전뇌허혈에 대한 신경보호 효과)

  • Kim, Young Ock;Lee, Sang Won;Yang, Seung Ok;Na, Sae Won;Kim, Su Kang;Chung, Joo Ho
    • Korean Journal of Medicinal Crop Science
    • /
    • v.22 no.1
    • /
    • pp.46-52
    • /
    • 2014
  • In traditional Korean and Chinese medicine, safflower (Carthamus tinctorius L.) for the treatment of central nervous system-related symptoms such as tremor, seizure, stroke and epilepsy. We investigated the effects of safflower could influence cerebral ischemia-induced neuronal and cognitive impairments. Administration of safflower for 1 day (200 mg/kg body weight, p.o.) increased the survival of hippocampal CA1 pyramidal neurons after transient global brain ischemia. And neurological functions measured as short term memory. Post-treatment with safflower for 2 times decreased the induction/reduction - induced production of neuronal cell loss from global cerebral ischemia. Safflower markedly decreased neuronal cell death and also caused a decrease in the content of thiobarbituric acid-reacting substances (TBARS) ($55.2{\pm}9.4{\mu}mol\;mg^{-1}$) and significant improvement of activities of glutathione (GSH) ($27.2{\pm}5.0{\mu}mol\;mg^{-1}$) in hippocampus. We conclude that treatment with safflower attenuated learning and memory deficits, and neuronal cell loss induced by global cerebral ischemia. These results suggest that safflower may be a potential candidate for the treatment of vascular dementia.

Cognitive Dysfunction and Hippocampal Damage Induced by Hypoxic-Ischemic Brain Injury and Prolonged Febrile Convulsions in Immature Rats

  • Byeon, Jung Hye;Kim, Gun-Ha;Kim, Joo Yeon;Sun, Woong;Kim, Hyun;Eun, Baik-Lin
    • Journal of Korean Neurosurgical Society
    • /
    • v.58 no.1
    • /
    • pp.22-29
    • /
    • 2015
  • Objective : Perinatal hypoxic-ischemic encephalopathy (HIE) and prolonged febrile seizures (pFS) are common neurologic problems that occur during childhood. However, there is insufficient evidence from experimental studies to conclude that pFS directly induces hippocampal injury. We studied cognitive function and histological changes in a rat model and investigated which among pFS, HIE, or a dual pathologic effect is most detrimental to the health of children. Methods : A rat model of HIE at postnatal day (PD) 7 and a pFS model at PD10 were used. Behavioral and cognitive functions were investigated by means of weekly open field tests from postnatal week (PW) 3 to PW7, and by daily testing with the Morris water maze test at PW8. Pathological changes in the hippocampus were observed in the control, pFS, HIE, and HIE+pFS groups at PW9. Results : The HIE priming group showed a seizure-prone state. The Morris water maze test revealed a decline in cognitive function in the HIE and HIE+pFS groups compared with the pFS and control groups. Additionally, the HIE and HIE+pFS groups showed significant hippocampal neuronal damage, astrogliosis, and volume loss, after maturation. The pFS alone induced minimal hippocampal neuronal damage without astrogliosis or volume loss. Conclusion : Our findings suggest that pFS alone causes no considerable memory or behavioral impairment, or cellular change. In contrast, HIE results in lasting memory impairment and neuronal damage, gliosis, and tissue loss. These findings may contribute to the understanding of the developing brain concerning conditions caused by HIE or pFS.

Ginsenoside Rb1 ameliorates cisplatin-induced learning and memory impairments

  • Chen, Chen;Zhang, Haifeng;Xu, Hongliang;Zheng, Yake;Wu, Tianwen;Lian, Yajun
    • Journal of Ginseng Research
    • /
    • v.43 no.4
    • /
    • pp.499-507
    • /
    • 2019
  • Background: Ginsenoside Rb1 (Rb1), a dominant component from the extract of Panax ginseng root, exhibits neuroprotective functions in many neurological diseases. This study was intended to investigate whether Rb1 can attenuate cisplatin-induced memory impairments and explore the potential mechanisms. Methods: Cisplatin was injected intraperitoneally with a dose of 5 mg/kg/wk, and Rb1 was administered in drinking water at the dose of 2 mg/kg/d to rats for 5 consecutive wk. The novel objects recognition task and Morris water maze were used to detect the memory of rats. Nissl staining was used to examine the neuron numbers in the hippocampus. The activities of superoxide dismutase, glutathione peroxidase, cholineacetyltransferase, acetylcholinesterase, and the levels of malondialdehyde, reactive oxygen species, acetylcholine, tumor necrosis factor-${\alpha}$, interleukin-$1{\beta}$, and interleukin-10 were measured by ELISA to assay the oxidative stress, cholinergic function, and neuroinflammation in the hippocampus. Results: Rb1 administration effectively ameliorates the memory impairments caused by cisplatin in both novel objects recognition task and Morris water maze task. Rb1 also attenuates the neuronal loss induced by cisplatin in the different regions (CA1, CA3, and dentate gyrus) of the hippocampus. Meanwhile, Rb1 is able to rescue the cholinergic neuron function, inhibit the oxidative stress and neuroinflammation in cisplatin-induced rat brain. Conclusion: Rb1 rescues the cisplatin-induced memory impairment via restoring the neuronal loss by reducing oxidative stress and neuroinflammation and recovering the cholinergic neuron functions.

Neuroprotective Effect of Wild Radish Extract on Scopolamine Induced Memory Impairment (갯무 추출물의 스코폴라민 유도 기억력 저하 모델에서의 뇌신경 보호 효과)

  • Hur, Jinyoung;Choi, Sang Yoon;Yeom, Mijung
    • Journal of the Korean Society of Food Culture
    • /
    • v.36 no.6
    • /
    • pp.633-639
    • /
    • 2021
  • Raphanus sativus var. hortensis f. raphanistroides Makino (Korean wild radish [WR]) are root vegetables belonging to the Brassicaceae family. These radish species mostly grow in sea areas in Asia, where they have been traditionally used as a medicinal food to treat various diseases. To investigate the effect of WR on neuronal cell death in SH-SY5Y cells, beta-amyloid was used to develop the cell death model. WR attenuated neuronal cell death in SH-SY5Y and regulated the mitogen-activated protein kinase (MAPK) signaling. WR extract also inhibited acetylcholinesterase inhibitor activity. Additionally, the WR treatment group ameliorated the behavior of the memory-impaired mice in a scopolamine-induced mouse model. In the behavior test, WR treated mice showed shorter escape latency and swimming distance and improved the platform-crossing number and the swimming time within the target quadrant. Furthermore, WR prevented histological loss of neurons in hippocampal CA1 regions induced by scopolamine. This study shows that WR can prevent memory impairment which may be a crucial way for the prevention and treatment of memory dysfunction and neuronal cell death.

Molecular Mechanism of Dietary Restriction in Neuroprevention and Neurogenesis: Involvement of Neurotrophic Factors

  • Park, Hee-Ra;Park, Mi-Kyung;Kim, Hyung-Sik;Lee, Jae-Won
    • Toxicological Research
    • /
    • v.24 no.4
    • /
    • pp.245-251
    • /
    • 2008
  • Dietary restriction (DR) is the most efficacious intervention for retarding the deleterious effects of aging. DR increases longevity, decreases the occurrence and severity of age-related diseases, and retards the physiological decline associated with aging. The beneficial effects of DR have been mostly studied in non-neuronal tissues. However, several studies have showed that DR attenuate neuronal loss after several different insults including exposure to kainate, ischemia, and MPTP. Moreover, administration of the non-metabolizable glucose analog 2-deoxy-D-glucose (2DG) could mimic the neuroprotective effect of DR in rodent, presumably by limiting glucose availability at the cellular level. Based on the studies of chemically induced DR, it has been proposed that the mechanism whereby DR and 2DG protect neurons is largely mediated by stress response proteins such as HSP70 and GRP78 which are increased in neurons of rats and mice fed a DR regimen. In addition, DR, as mild metabolic stress, could lead to the increased activity in neuronal circuits and thus induce expression of neurotrophic factors. Interestingly, such increased neuronal activities also enhance neurogenesis in the brains of adult rodents. In this review, we focus on what is known regarding molecular mechanisms of the protective role of DR in neurodegenerative diseases and aging process. Also, we propose that DR is a mild cellular stress that stimulates production of neurotrophic factors, which are major regulators of neuronal survival, as well as neurogenesis in adult brain.

Protective Effects of Celastrol, the Triterpenoid Component of Celastrus Orbiculatus, on Dopaminergic Neuronal Cells in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned Parkinson's Disease Rats (노박덩굴에 함유된 celastrol 성분의 파킨슨병을 유발시킨 쥐에서의 도파민 신경세포 보호효과)

  • Lee, Kap-Duk;Kim, Kwang-Jin;Park, Yong-Ki
    • The Journal of Korean Medicine
    • /
    • v.29 no.4
    • /
    • pp.94-103
    • /
    • 2008
  • Objectives: The aim of this study was to determine whether celastrol, the triterpenoid component of Celastrus orbiculatus, offers neuroprotection against Parkinson's disease (PD) in mice administered 1-methyl-4-phenyl-1,2,3,6- tetrahydropyridine(MPTP). Methods: We examined how celastrol affected MPTP-induced neuronal loss of tyrosine hydroxylase (TH)-positive dopaminergic neurons in substantia nigra pars compacta (SNpc) in the midbrain of mice. C57BL/6J mice were divided into four groups: (1) saline-saline, (2) saline-celastrol, (3) MPTP-saline, and (4) MPTP-celastrol. The mice were injected intraperitoneally (i.p.) with four administrations of MPTP (18mg/kg) at 2 h intervals and then i.p. administered celastrol (3mg/kg) two times at 12 h after last celastrol administration. Expression of TH on the SNpc of brain tissues were analyzed at 7 days after the treatments by immunohistochemistry and Western blot. Results: Immunohistochemical analysis using TH antibody showed that celastrol provided significantly protective effects against MPTP-induced loss of TH-positive dopaminergic neurons in the SNpc region of the midbrain of mice. Our Western blot study also showed that celastrol significantly inhibits the MPTP-induced neuronal damage via the up-regulation of TH protein levels in MPTP mice. Conclusions: The present results suggest that it may be possible to use celastrol for the prevention of nigral degenerative disorders including PD, caused by exposure to toxic substances.

  • PDF

Alzheimer's Disease and Apoptosis

  • Kim, Young-Hoon;Kim, Hye-Sun;Park, Cheol-Hyoung;Jeong, Sung-Jin;Kim, Young-Kyung;Kim, Sun-Hee;Lee, Sang-Kyeng;Suh, Yoo-Hun;Kim, Sung Su
    • Korean Journal of Biological Psychiatry
    • /
    • v.5 no.1
    • /
    • pp.66-70
    • /
    • 1998
  • Apoptosis is a form of cell death in which the cells shrink and exhibit nuclear chromatin condensation and DNA fragmentation, and yet maintain membrane integrity. Many lines of evidence have shown that brain neurons are vulnerable to degeneration by apoptosis. Also it has been suggested that apoptosis is one of the mechanism contributing neuronal loss in Alzheimer's disease(AD), since the conditions in the disease($A{\beta}$ peptide, oxidative stress, low energy metabolism) are the inducers that activate apoptosis. Indeed some neurons in vulnerable regions of the AD brain show DNA damage, chromatin condensation, and apoptic bodies. Consistently, mutations in AD causative genes(Amyloid precursor protein, Presenilin-1 and Presenilin- 2) increase $A{\beta}$ $peptide_{1-42}(A{\beta}_{1-42})$ and sensitize neuronal cell to apoposis. However, several lines of evidence have shown that the location of neuronal loss and $A{\beta}$ peptide deposition is not correlated in AD brain and transgenic mice brain over-expressing $A{\beta}_{1-42}$. Taken together, these data may indicated that $A{\beta}$ peptide(and other causative factors of AD) can interact with other cellular insults or risk factors to exacerbate pathological mechansim of AD through apoptosis. Thus, this review discusses possible role and mechanism of apoptosis in AD.

  • PDF

Effect of Chungganhaeju-hwan in Ethanol-induced Neuronal Cell Damage (청간해주환(淸肝解酒丸)의 알코올 유도 뇌신경세포 손상에 대한 보호 효과)

  • Ju, Mi-Sun;Kim, Hyo-Geun;Cho, Hae-Jeong;Sim, Jae-Jong;Jeon, Yong-Jun;Oh, Myung-Sook
    • The Korea Journal of Herbology
    • /
    • v.26 no.3
    • /
    • pp.75-82
    • /
    • 2011
  • Objectives : In this study, we evaluated the effect of Chungganhaeju-hwan(CGHJH) on hydrogen peroxide($H_2O_2$)-induced and ethanol(EtOH)-induced neuronal damage in vitro and in vivo, respectively. Methods:We carried out the anti-oxidant effects of CGHJH against hydrogen peroxide($H_2O_2$)-induced toxicity in HT22 and PC12 cells using thiazolyl blue tetrazolium bromide. Then, to investigate the protective effect on CGHJH against EtOH-induced memory impairment and hippocampal cell damage in male ICR mice, we performed novel object recognition test(NORT), and analysed the brain tissues after immunohistochemistry and western blotting. Results:CGHJH showed protective effect from $H_2O_2$-induced cell toxicity at doses of $1\sim100{\mu}g$/mL in both HT22 and PC12 cells. CGHJH had also recovery effect from EtOH-induced memory impairment in ICR mice from NORT and it protected hippocampal cells against EtOH toxicity in the result of cresyl violet and NeuN immunoreactivity. Conclusion : These results demonstrate that CGHJH has protective effect in neuronal cells against $H_2O_2$ and EtOH toxicities and this effect could be a main role of recovery effect on EtOH-induced memory loss.