• Title/Summary/Keyword: Primary astrocytes

Search Result 78, Processing Time 0.032 seconds

The Inhibitory Effect of Nicotine on TNF-α Expression in Human Fetal Astrocytes (담배 니코틴에 의한 사람 태아 성상세포에서 종양괴사인자(TNF-α)의 발현 억제작용)

  • Son, Il-Hong;Lee, Sung-Ik;Yang, Hyun-Duk;Han, Sun-Jung;Suk, Seung-Han;Lee, Jai-Kyoo;Kim, Jae-Hyun;Park, Joo-Young;Moon, Hyung-In;Lee, Sung-Soo
    • Journal of the Korean Chemical Society
    • /
    • v.51 no.3
    • /
    • pp.251-257
    • /
    • 2007
  • The Tumor necrosis factor-α, (TNF-α), is involved in the pathogenesis of multiple sclerosis and contributes to the degeneration of oligodendrocytes as well as neurons. Nicotine has been found to have immunosuppressive and inflammation-suppressing effects. Astrocytes, the major glial cells in the CNS, are capable of producing TNF-α at both the mRNA and protein levels in response to interleukin-1 (IL-1) or TNF-α. Nicotine has been shown to influence glial cell functions. To order to explore the role of astrocytes in the production of TNF-α, astrocytes were pretreated with nicotine and are stimulated with IL-1β to determine their effects on TNF-α production. The results are as follows. Cytotoxic effects of nicotine on human fetal astrocytes were noted above 10 μg/ml of nicotine. The effect of IL-1β on TNF-α mRNA expression in primary cultured human fetal astrocytes was maximal at 2 h after IL- 1β(100 pg/ml) treatment. Human fetal astrocytes were pretreated with 0.1, 1, and 10 μg/ml of nicotine and then stimulated with IL-1β (100 pg/ml) for 2 h. The inhibitory effect of nicotine on expressions of TNF-α mRNA in human fetal astrocytes with pretreated 0.1 μg/ml of nicotine is first noted at 8 hr, and the inhibitory effect is maximal at 12 h. The inhibitory effect at 1 μg/ml of nicotine is inhibited maximal at 24 h. Nicotine at 0.1, 1 and 10 μg/ml concentrations significantly inhibits IL-1β-induced NF-κB activation. Collectively, this study indicates that nicotine might inhibit the expression of TNF-α in activated human fetal astrocytes.

Melatonin Induces Akt Phosphorylation through Melatonin Receptor- and PI3K-Dependent Pathways in Primary Astrocytes

  • Kong, Pil-Jae;Byun, Jong-Seon;Lim, So-Young;Lee, Jae-Jun;Hong, Sung-Jun;Kwon, Kwang-Jun;Kim, Sung-Soo
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.12 no.2
    • /
    • pp.37-41
    • /
    • 2008
  • Melatonin has been reported to protect neurons from a variety of neurotoxicity. However, the underlying mechanism by which melatonin exerts its neuroprotective property has not yet been clearly understood. We previously demonstrated that melatonin protected kainic acid-induced neuronal cell death in mouse hippocampus, accompanied by sustained activation of Akt, a critical mediator of neuronal survival. To further elucidate the neuroprotective action of melatonin, we examined in the present study the causal mechanism how Akt signaling pathway is regulated by melatonin in a rat primary astrocyte culture model. Melatonin resulted in increased astrocytic Akt phosphorylation, which was significantly decreased with wortmannin, a specific inhibitor of PI3K, suggesting that activation of Akt by melatonin is mediated through the PI3K-Akt signaling pathway. Furthermore, increased Akt activation was also significantly decreased with luzindole, a non-selective melatonin receptor antagonist. As downstream signaling pathway of Akt activation, increased levels of CREB phoshorylation and GDNF expression were observed, which were also attenuated with wortmannin and luzindole. These results strongly suggest that melatonin exerts its neuroprotective property in astrocytes through the activation of plasma membrane receptors and then PI3K-Akt signaling pathway.

Neurotoxicity Assessment of Methamphetamine and Cadmium Using Cultured Neuronal Cells of Long-Evans Rats (신경세포 배양법을 이용한 methamphetamine과 cadmium의 신경독성 평가)

  • Cho, Dae-Hyun;Kim, Jun-Gyon;Jeong, Yong;Lee, Bong-Hun;Kim, Eun-Youb;Kim, Jeong-Goo;Cho, Tai-Soon;Kim, Jin-Suk;Moon, Hwa-Hwey
    • Toxicological Research
    • /
    • v.12 no.1
    • /
    • pp.69-79
    • /
    • 1996
  • Primary culture of cerebellar neuronal cells derived from 8-day old Long-Evans rats was used. Pure granule cells, astrocytes or mixed cells culture systems were prepared. These cells were differentiated and developed synaptic connections. And the astrocytes were identified by immunostaining with glial fibrillary acidic protein (GFAP). Methamphetamine (MAP), which acts on dopaminergic system and cadmium (Cd), a toxic heavy metal, were applied and biochemical assays and electrophysiological studies were performed. $LC_50$ values estimated by MTT assay of MAP and Cd were 3 mM and 2$\mu M$ respectively. Cells were treated with 1 mM or 2 mM MAP and 1$\mu M$ $CdCl_2$ for 48 hour, and the incubation media were analyzed for the content of released LDH. MAP (2 mM) and Cd significantly increased the LDH release. Cell viability was decreased in both groups and some cytopathological changes like cell swelling or vacuolization were seen. The cerebellar granule cells were used for measuring membrane currents using whole-cell clamp technique. Sodium and potassium currents were not affected by MAP neither Cd, but calcium current was significantly reduced by Cd but not affected by MAP. Therefore, in vitro neurotoxicity test system using neuronaI cells and astrocytes cultures were established and can be used in screening of potential neurotoxic chemicals.

  • PDF

Expression of microRNA-218 and its Clinicopathological and Prognostic Significance in Human Glioma Cases

  • Cheng, Mao-Wei;Wang, Ling-Ling;Hu, Gu-Yu
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.5
    • /
    • pp.1839-1843
    • /
    • 2015
  • Background: MicroRNAs are a class of noncoding RNAs which regulate multiple cellular processes during tumor development. The purpose of this report is to investigate the clinicopathological and prognostic significance of miR-218 in human gliomas. Materials and Methods: Quantitative RT-PCR (qRT-PCR) was conducted to detect the expression of miR-218 in primary normal human astrocytes, three glioma cell lines and 98 paired glioma and adjacent normal brain tissues.Associations of miR-218 with clinicopathological variables of glioma patients were statistically analyzed. Finally, a survival analysis was performed using the Kaplan-Meier method and Cox's proportional hazards model. Results: The expression level of miR-218 in primary normal human astrocytes was significantly higher than that in glioma cell lines (p<0.01). Also, the expression level of miR-218 in glioma tissues was significantly downregulated in comparison with that in the adjacent normal brain tissues (p<0.001). Statistical analyses demonstrated that low miR-218 expression was closely associated with advanced WHO grade (p=0.002) and low Karnofsky performance score (p=0.010) of glioma patients. Kaplan-Meier analysis with the log-rank test showed that patients with low-miR-218 expression had poorer disease-free survival and overall survival (p=0.0045 and 0.0124, respectively). Multivariate analysis revealed that miR-218 expression was independently associated with the disease-free survival (p=0.009) and overall survival (p=0.004) of glioma patients. Conclusions: Our results indicate that miR-218 is downregulated in gliomas and that its status might be a potential valuable biomarker for glioma patients.

Glial Fibrillary Acidic Protein Splice Variants in Hepatic Stellate Cells - Expression and Regulation

  • Lim, Michelle Chin Chia;Maubach, Gunter;Zhuo, Lang
    • Molecules and Cells
    • /
    • v.25 no.3
    • /
    • pp.376-384
    • /
    • 2008
  • The glial fibrillary acidic protein (GFAP) is traditionally used as a marker for astrocytes of the brain, and more recently for the hepatic stellate cells (HSCs) of the liver. Several GFAP splice variants have been previously reported in the astrocytes of the CNS and in the non-myelinating Schwann cells of the PNS. In this study, we investigate whether GFAP splice variants are present in the HSCs and their expression as a function of HSCs activation. Furthermore, the regulation of these transcripts upon treatment with interferon gamma ($IFN-{\gamma}$) will be explored. Using semi-quan-titative RT-PCR and real-time PCR, we examine the expression and regulation of GFAP splice variants in HSCs as well as their respective half-life. We discover that most of the GFAP splice variants ($GFAP{\alpha}$, ${\beta}$, ${\delta}$, ${\varepsilon}$ and $\kappa$) found in the neural system are also expressed in quiescent and culture-activated primary HSCs. Interestingly, $GFAP{\alpha}$ is the predominant form in quiescent and culture-activated primary HSCs, while $GFAP{\beta}$, predominates in the SV40-immortalized activated HSC-T6. $GFAP{\delta}$, ${\varepsilon}$ and ${\kappa}$ have similar half-lives of 10 hours, while $GFAP{\beta}$ has a half-life of 17 hours. Treatment of HSC-T6 with $IFN-{\gamma}$ results in a significant 1.29-fold up-regulation of $GFAP{\alpha}$ whereas the level of the other transcripts remains unchanged. In summary, $GFAP{\alpha}$, ${\beta}$, ${\delta}$, ${\varepsilon}$ and $\kappa$ are present in HSCs. They are differentially regulated on the transcription level, implying a role of the 5' and 3' untranslated regions.

White Ginseng Saponin Upregulated the Production of -TNFTNF-α, IL-1β and NO in Primary Cultures of Mixed Glial Cells (고려인삼에 의한 신경면역 및 염증반응 조절: 백삼사포닌에 의한 교세포에서의 TNF-α, IL-1β 및 NO 생성 증가)

  • 성정훈;최동희;김동훈;전보권;최상현
    • Journal of Ginseng Research
    • /
    • v.28 no.2
    • /
    • pp.120-126
    • /
    • 2004
  • Glial cells such as astrocytes and microglial cells are the main source of proinflammatory cytokines and nitric oxide(NO) in the central nervous system, which exert neuroimmune and inflammatory functions and other various neurobiologic effects. Though Panax ginseng C.A. Meyer has been known to strengthen the body's defence mechanisms and also to maintain the homeostasis in the central nervous system, the effects of Panax ginseng on the production of immune and inflammatory mediators have not been studied well in the brain. Therefore, this study was designed to study the effects of ginseng saponins on the production of proinflammatory cytokines and NO in the primary cultures of mixed glial cells. White ginseng saponin, 200-500 $\mu$g/ml, showed significant cytotoxicity after 72 hrs and increased TNF-$\alpha$, IL-$\beta$, and NO production. Lower doses of 50-100 $\mu\textrm{g}$/ml showed little cytotoxicity until 72 hrs and also increased the production of TNF-$\alpha$, IL-1$\beta$, and NO. Triple immune staining showed that white ginseng saponin, 200$\mu\textrm{g}$/ml for 72 ks, induced stellation of astrocytes and iNOS expression exclusively in microglial cells. Taken together, the white ginseng saponin increased the production of proinflammatory cytokines such as TNF-$\alpha$ and IL-1$\beta$, and induced iNOS expression and NO production in mixed glial cell cultures, which may be ascribed to the enhancement of central immune responses and the regulation of inflammatory reactions by Panax ginseng.

Inhibitory Effects of Phylligenin on the Proliferation of Cultured Rat Neural Progenitor Cells

  • Lee, Sung-Hoon;Go, Hyo-Sang;Choi, Chang-Soon;Cheong, Jae-Hoon;Han, Sun-Young;Bae, Ki-Hwan;Ko, Kwang-Ho;Park, Seung-Hwa
    • Biomolecules & Therapeutics
    • /
    • v.18 no.1
    • /
    • pp.48-55
    • /
    • 2010
  • Neural progenitor cells (NPCs) differentiate into astrocytes, neurons and oligodendrocytes, which is controlled by various factors in brain. Recent evidences suggest that small molecules modulating the proliferation and differentiation of NPCs may have therapeutic value as well as the potential use as chemical probes. Phylligenin is a lignan with anti-inflammatory activity that is isolated from the fruits of Forsythia koreana. We investigated effects of phylligenin on proliferation and differentiation of NPCs. Treatment of phylligenin decreased the number of proliferating NPCs in culture without effects on the differentiation and survival of neural cells such as neurons and astrocytes. To examine the mechanism of the decreased NPCs number, we performed cell cycle analysis. Proliferation of NPCs was decreased via G1-S transition block by phylligenin treatment, and it was mediated by the increase of p21 level. However, phylligenin did not induce apoptosis of NPCs as determined by TUNEL assay and PARP cleavage. We also found that viability of glioma cell lines such as C6 and U87MG glioma cells, but not that of primary neuron and astrocyte, was inhibited by phylligenin. These results suggest that phylligenin selectively inhibits proliferation of rapidly growing cells such as neural stem cells and glioma cells. Given that the possible role of brain tumor stem cells in the pathology of brain cancers, the inhibitory effects of phylligenin might be useful in the development of new therapeutic agents against brain cancers.

The complement system: a potential target for the comorbidity of chronic pain and depression

  • Shanshan Tang;Wen Hu;Helin Zou;Qingyang Luo;Wenwen Deng;Song Cao
    • The Korean Journal of Pain
    • /
    • v.37 no.2
    • /
    • pp.91-106
    • /
    • 2024
  • The mechanisms of the chronic pain and depression comorbidity have gained significant attention in recent years. The complement system, widely involved in central nervous system diseases and mediating non-specific immune mechanisms in the body, remains incompletely understood in its involvement in the comorbidity mechanisms of chronic pain and depression. This review aims to consolidate the findings from recent studies on the complement system in chronic pain and depression, proposing that it may serve as a promising shared therapeutic target for both conditions. Complement proteins C1q, C3, C5, as well as their cleavage products C3a and C5a, along with the associated receptors C3aR, CR3, and C5aR, are believed to have significant implications in the comorbid mechanism. The primary potential mechanisms encompass the involvement of the complement cascade C1q/C3-CR3 in the activation of microglia and synaptic pruning in the amygdala and hippocampus, the role of complement cascade C3/C3a-C3aR in the interaction between astrocytes and microglia, leading to synaptic pruning, and the C3a-C3aR axis and C5a-C5aR axis to trigger inflammation within the central nervous system. We focus on studies on the role of the complement system in the comorbid mechanisms of chronic pain and depression.

Primary Culture of Endothelial Cells from Murine Brain Microvessels

  • Lee Sun-Ryung
    • Biomedical Science Letters
    • /
    • v.12 no.2
    • /
    • pp.127-130
    • /
    • 2006
  • It is important to coordinated interaction among neurons, astrocytes and endothelial cells to maintain the function of brain. To study their regulatory mechanisms in vitro system, the co-culture system among the isolated cells from brain may be needed. However, the method for purifying brain microvascular endothelial cells (BMEC) far culture have not established yet. In this study, the proper culture methods of mice cells using two different strains, CD1 and C57BL6, to obtain the pure and plentiful endothelial cells were described. The flatted-round forms of CD1 endothelial cells grew on the collagen-IV coating plates, while the purified cells from C57 mice preferred type collagen-I dishes for their growth. Both cells displayed anti-PECAM-1 (CD31) and von Willebrand Factor immune-reactivity. These results indicated that different coating materials not only improve attachment of isolated cells but also promoting growth of cells, suggesting that this method of purifying murine Brain microvascular endothelial cells (BMEC) provides a suitable model to investigate blood-brain-barrier (BBB) properties within neurovascular unit in vitro.

  • PDF