• Title/Summary/Keyword: Hippocampal Cell

Search Result 238, Processing Time 0.029 seconds

Acupuncture Treatment at HT8 Protects Hippocampal Cells in Dentate Gyrus on Kainic Acid-Induced Epilepsy Mice Model (소부혈(少府穴) 자침(刺鍼)이 Kainic Acid로 유도(誘導)된 간질(癎疾) 동물(動物) 모델의 해마(海馬) 치상회(齒狀回)에 미치는 영향(影響))

  • Kim, Seung-Tae;Chung, Joo-Ho;Jeong, Wu-Byung;Kim, Jang-Hyun;Kang, Min-Jung;Hong, Mee-Sook;Park, Hae-Jeong;Kim, Yeon-Jung;Park, Hi-Joon;Lee, Hye-Jeong
    • Korean Journal of Acupuncture
    • /
    • v.24 no.4
    • /
    • pp.99-110
    • /
    • 2007
  • Objectives : Epilepsy is one of the most common serious brain disorders that affect people of all ages, and it is characterized by recurrent unprovoked seizures. We examined whether acupuncture can reduce both the incidence of seizures and hippocampal cell death in dentate gyrus (DG) using a mouse model of kainic acid (KA)-induced epilepsy. Methods : ICR mice ($20{\sim}25$ g) were given acupuncture once a day at acupoint HT8 (sobu) bilaterally during 2 days before KA injection. After an intracerebroventricular injection of 0.1${\mu}g$ of KA, acupuncture treatment was subsequently administered once more (total 3 times), and the degree of seizure was observed for 20 min. Three hours after injection, we confirmed the neural cell death using cresyl violet staining and silver impregnation staining, and determined the expressions of c-Fos and glutamate decarboxylase (GAD)-67 using immunohistochemistry techniques in the DG. Results : KA induced epileptic seizure, neural cell death, increased c-Fos expression and decreased GAD-67 expression in the DG. Acupuncture treatment at HT8 reduced the severity of the epileptic seizure and inhibited neural cell death from KA. In addition, acupuncture normalized the expressions of c-Fos and GAD-67 in the same areas. Conclusions : These results demonstrated that acupuncture treatment at HT8 may reduce the KA-induced epileptic seizure and neural cell death in the DG possibly by normalizing c-Fos expressions and the gamma-aminobutyric acid neurons.

  • PDF

Tat-indoleamine 2,3-dioxygenase 1 elicits neuroprotective effects on ischemic injury

  • Park, Jung Hwan;Kim, Dae Won;Shin, Min Jea;Park, Jinseu;Han, Kyu Hyung;Lee, Keun Wook;Park, Jong Kook;Choi, Yeon Joo;Yeo, Hyeon Ji;Yeo, Eun Ji;Sohn, Eun Jeong;Kim, Hyoung-Chun;Shin, Eun-Joo;Cho, Sung-Woo;Kim, Duk-Soo;Cho, Yong-Jun;Eum, Won Sik;Choi, Soo Young
    • BMB Reports
    • /
    • v.53 no.11
    • /
    • pp.582-587
    • /
    • 2020
  • It is well known that oxidative stress participates in neuronal cell death caused production of reactive oxygen species (ROS). The increased ROS is a major contributor to the development of ischemic injury. Indoleamine 2,3-dioxygenase 1 (IDO-1) is involved in the kynurenine pathway in tryptophan metabolism and plays a role as an anti-oxidant. However, whether IDO-1 would inhibit hippocampal cell death is poorly known. Therefore, we explored the effects of cell permeable Tat-IDO-1 protein against oxidative stress-induced HT-22 cells and in a cerebral ischemia/reperfusion injury model. Transduced Tat-IDO-1 reduced cell death, ROS production, and DNA fragmentation and inhibited mitogen-activated protein kinases (MAPKs) activation in H2O2 exposed HT-22 cells. In the cerebral ischemia/reperfusion injury model, Tat-IDO-1 transduced into the brain and passing by means of the blood-brain barrier (BBB) significantly prevented hippocampal neuronal cell death. These results suggest that Tat-IDO-1 may present an alternative strategy to improve from the ischemic injury.

Neuroprotective Effects of Scrophulariae Radix on Cerebral Ischemia in Mongolian Gerbils (Mongolian gerbil의 뇌허혈에 대한 현삼의 신경보호효과)

  • Lee, Jun-Hwan;Song, Mi-Yeon;Lee, Jong-Soo;Kim, Sung-Su;Shin, Hyun-Dae;Chung, Seok-Hee
    • Journal of Korean Medicine Rehabilitation
    • /
    • v.18 no.4
    • /
    • pp.1-11
    • /
    • 2008
  • Objectives : Cerebral ischemia resulting from transient or permanent occlusion of cerebral arteries leads to neuronal cell death and eventually causes neurological impairments. Scrophulariae radix is the roots of Scrophularia buergeria. In the present study, we investigated the effects of the aqueous extract of Scrophulariae radix on apoptotic cell death in the hippocampal dentate gyrus following transient global ischemia in gerbils. Methods : For this study, step-down avoidance task, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay, and immunohistochemistry for caspase-3 were performed. Results : The present results showed that apoptotic cell death in the hippocampal dentate gyrus was significantly increased following transient global ischemia in gerbils. Treatment with the aqueous extract of Scrophulariae radix suppressed the ischemia-induced apoptosis in the dentate gyrus and thus facilitated the recovery of short-term memory impairment induced by ischemic cerebral injury. Conclusions : Here in this study, we have shown that Scrophulariae radix has a positive effect on-and possesses protective qualities against ischemia-induced apoptotic neuronal cell death, and it can be used for the treatment of ischemic brain diseases.

Tat-Thioredoxin-like protein 1 attenuates ischemic brain injury by regulation of MAPKs and apoptosis signaling

  • Hyun Ju Cha;Won Sik Eum;Gi Soo Youn;Jung Hwan Park;Hyeon Ji Yeo;Eun Ji Yeo;Hyun Jung Kwon;Lee Re Lee;Na Yeon Kim;Su Yeon Kwon;Yong-Jun Cho;Sung-Woo Cho;Oh-Shin Kwon;Eun Jeong Sohn;Dae Won Kim;Duk-Soo Kim;Yu Ran Lee;Min Jea Shin;Soo Young Choi
    • BMB Reports
    • /
    • v.56 no.4
    • /
    • pp.234-239
    • /
    • 2023
  • Thioredoxin-like protein 1 (TXNL1), one of the thioredoxin superfamily known as redox-regulator, plays an essential in maintaining cell survival via various antioxidant and anti-apoptotic mechanisms. It is well known that relationship between ischemia and oxidative stress, however, the role of TXNL1 protein in ischemic damage has not been fully investigated. In the present study, we aimed to determine the protective role of TXNL1 against on ischemic injury in vitro and in vivo using cell permeable Tat-TXNL1 fusion protein. Transduced Tat-TXNL1 inhibited ROS production and cell death in H2O2-exposed hippocampal neuronal (HT-22) cells and modulated MAPKs and Akt activation, and pro-apoptotic protein expression levels in the cells. In an ischemia animal model, Tat-TXNL1 markedly decreased hippocampal neuronal cell death and the activation of astrocytes and microglia. These findings indicate that cell permeable Tat-TXNL1 protects against oxidative stress in vitro and in vivo ischemic animal model. Therefore, we suggest Tat-TXNL1 can be a potential therapeutic protein for ischemic injury.

Pharmacological Properties of CDBT in Hypoxia-induced Neuronal Cell Injury and Their Underlying Mechanisms

  • Park, Sang-kyu;Jung, Eun-sun;Cha, Ji-yoon;Cho, Hyun-kyoung;Yoo, Ho-ryong;Kim, Yoon-sik;Seol, In-chan
    • The Journal of Internal Korean Medicine
    • /
    • v.40 no.3
    • /
    • pp.425-442
    • /
    • 2019
  • Objectives: This study aimed to reveal the pharmacological properties of the newly prescribed herbal mixture, Chenmadansamgamibokhap-tang(CDBT), against hypoxia-induced neuronal cell injury (especially mouse hippocampal neuronal cell line, HT-22 cells) and their corresponding mechanisms. Methods: A cell-based in vitro experiment, in which a hypoxia condition induced neuronal cell death, was performed. Various concentrations of the CDBT were pre-treated to the HT-22 cells for 4 h before 18 h in the hypoxia chamber. The glial cell BV-2 cells were stimulated with $IFN{\gamma}$ and LSP to produce inflammatory cytokines and reactive oxygen species. When the neuronal HT-22 cells were treated with this culture solution, the drug efficacy against neuronal cell death was examined. Results: CDBT showed cytotoxicity in the normal condition of HT-22 cells at a dose of $125{\mu}g/mL$ and showed a protective effect against hypoxia-induced neuronal cell death at a dose of $31.3{\mu}g/mL$. CDBT prevented hypoxia-induced neuronal cell death in a dose-dependent manner in the HT-22 cells by regulating $HIF1{\alpha}$ and cell death signaling. CDBT prevented neuronal cell death signals and DNA fragmentation due to the hypoxia condition. CDBT significantly reduced cellular oxidation, cell death signals, and caspase-3 activities due to microglial cell activations. Moreover, CDBT significantly ameliorated LPS-induced BV-2 cell activation and evoked cellular oxidation through the recovery of redox homeostasis. Conclusions: CDBT cam be considered as a vital therapeutic agent against neuronal cell deaths. Further studies are required to reveal the other functions of CDBT in vivo or in the clinical field.

Pre-ischemic Treatment with Ampicillin Reduces Neuronal Damage in the Mouse Hippocampus and Neostriatum after Transient Forebrain Ischemia

  • Lee, Kyung-Eon;Kim, Seul-Ki;Cho, Kyung-Ok;Kim, Seong-Yun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.12 no.6
    • /
    • pp.287-291
    • /
    • 2008
  • Ampicillin, a $\beta$-lactam antibiotic, has been reported to induce astrocytic glutamate transporter-l which plays a crucial role in protecting neurons against glutamate excitotoxicity. We investigated the effect of ampicillin on neuronal damage in the mouse hippocampus and neostriatum following transient global forebrain ischemia. Male C57BL/6 mice were anesthetized with halothane and subjected to bilateral occlusion of the common carotid artery for 40 min. Ampicillin was administered post-ischemically (for 3 days) and/or pre-ischemically (for $3{\sim}5$ days until one day before the onset of ischemia). Pre- and post-ischemic treatment with ampicillin (50 mg/kg/day or 200 mg/kg/day) prevented ischemic neuronal death in the medial CAI area of the hippocampus as well as the neostriatum in a dose-dependent manner. In addition, ischemic neuronal damage was reduced by pre-ischemic treatment with ampicillin (200 mg/kg/day). In summary, our results suggest that ampicillin plays a functional role as a chemical preconditioning agent that protects hippocampal neurons from ischemic insult.

Neuroprotective Activity of Spirulina maxima Hot Ethanol Extract (스피루리나 에탄올 추출물의 신경세포 보호활성)

  • Ryu, Gahee;Ma, Choong Je
    • Korean Journal of Pharmacognosy
    • /
    • v.52 no.3
    • /
    • pp.149-156
    • /
    • 2021
  • Excessive glutamate can cause oxidative stress in neuronal cells and this can be the reason for neurodegenerative disease. In this study, we investigated the protective effect of Spirulina maxima hot ethanol extract on mouse hippocampal HT22 cell of which glutamate receptor has no function. HT22 cells were pre-treated with S. maxima sample at a dose dependent manner (1, 10 and 100 ㎍/ml). After an hour, glutamate was treated. Cell viability, reactive oxygen species (ROS) accumulation, Ca2+ influx, decrease of mitochondrial membrane potential level and glutathione related assays were followed by then. S. maxima ethanol extract improved the cell viability by suppressing the ROS and Ca2+ formation, retaining the mitochondrial membrane potential level and protecting the activity of the antioxidant enzymes compared with group of vehicle-treated controls. These suggest that S. maxima may decelerate the neurodegeneration by attenuating neuronal damage and oxidative stress.

Neuroanatomical Localization of Cells Containing Gonadotropin Releasing Hormone mRNA in the Brain of Frog, Rana dvbowskii, by in situ Hybridization (In situ hybridization법에 의한 북방산개구리 뇌에서 GnRH mRNA를 함유한 세포의 분포 연구)

  • 최완성;김정우
    • The Korean Journal of Zoology
    • /
    • v.37 no.3
    • /
    • pp.304-310
    • /
    • 1994
  • Using in situ hybridization, we have mapped the anatomical localization of perikarya containing myNA that codes for sonadotropin releasing hormone (GnRH) in the brains of female frogs, R. dybowskii. DNA olisomers, with sequences complementary to the GnRH portion of pro-GnRH myNA sequence, were synthesized and hybridized to paraformaldehvde-fixed, sagittal sections of the whole brain stem. The distribution of the GnRH mRNA containing cell bodies was similar to that described for GnRH peptide by immunohistochemistrv. That is, cells containing GnRH mRNA were observed in the medial septal area, anterior preoptic area, ventromedial hvpothalamus and infundibular regions. However, another cell groups which contains GnRH mRNAs were also detected by in situ hybridization in the bed nucleus of hippocampal commissure, preoptic area, nucleus infundibularis dorsalis, mesencephalic nuclei and intermediolateral cell column of spinal cord areas. These results demonstrate the feasibility of using in situ hybridization as a strategy to study the distribution of GnRH neurons and the detection of GnRH gene expression in the vertebrates.

  • PDF

Protective Effects of Seok-Jeong on the Toxicity of Cadmium in Neuronal Cells (뇌신경세포에서 토양미생물 발효추출액인 석정의 카드뮴 독성에 대한 방어효과)

  • 홍순해;안성희;장봉기;박종안;이종화
    • YAKHAK HOEJI
    • /
    • v.47 no.2
    • /
    • pp.85-92
    • /
    • 2003
  • Seok-jeong (SJ) is a solution of various metal ions and numerous other organic substances produced through extraction and fermentation of herbs and soil using geo-microbes, and it has been shown to improve symptoms of senile dementia. In this study, we investigated the protective effects of SJ against neurotoxicity of cadmium in HT22 hippocampal neuron cell line. SJ significantly protected from the cadmium-induced decreased cell viability measured by MTT assay (p<0.01). The protective effects of SJ against cadmium toxicity were confirmed through observing morphological changes using inverted microscope. Additionally, SJ significantly repressed the formation of lipid peroxidation induced by high concentration of cadmium, and likewise, significantly repressed the reduction of glutathione by cadmium in HT22 cells. Vitamin C at the concentration found in SJ did not show any protective effect against cadmium toxicity in HT22 cells, indicating that vitamin C may not have a major role in the protective mechanism of SJ. Taken together, these results suggest that SJ may be a valuable agent for the protection of cadmium toxicity on the neuronal cells, and that the mechanism of the action of SJ may be due to reduced lipid peroxidation and increased glutathione level.

5-Hydroxytryptamine 6 Receptor (5-HT6R)-Mediated Morphological Changes via RhoA-Dependent Pathways

  • Rahman, Md. Ataur;Kim, Hanna;Lee, Kang Ho;Yun, Hyung-Mun;Hong, Jung-Hwa;Kim, Youngjae;Choo, Hyunah;Park, Mikyoung;Rhim, Hyewhon
    • Molecules and Cells
    • /
    • v.40 no.7
    • /
    • pp.495-502
    • /
    • 2017
  • The $5-HT_6R$ has been considered as an attractive therapeutic target in the brain due to its exclusive expression in the brain. However, the mechanistic linkage between $5-HT_6Rs$ and brain functions remains poorly understood. Here, we examined the effects of $5-HT_6R$-mediated cell morphological changes using immunocytochemistry, Western blot, and live-cell imaging assays. Our results showed that the activation of $5-HT_6Rs$ caused morphological changes and increased cell surface area in HEK293 cells expressing $5-HT_6Rs$. Treatment with 5-HT specifically increased RhoA-GTP activity without affecting other Rho family proteins, such as Rac1 and Cdc42. Furthermore, live-cell imaging in hippocampal neurons revealed that activation of $5-HT_6Rs$ using a selective agonist, ST1936, increased the density and size of dendritic protrusions along with the activation of RhoA-GTP activity and that both effects were blocked by pretreatment with a selective $5-HT_6R$ antagonist, SB258585. Taken together, our results show that $5-HT_6R$ plays an important role in the regulation of cell morphology via a RhoA-dependent pathway in mammalian cell lines and primary neurons.