• Title/Summary/Keyword: alzheimer's disease β-amyloid interleukin-6

Search Result 6, Processing Time 0.02 seconds

The Effect of Jeongshin-tang on Interleukin-1 $\beta$ and $\beta$-Amyloid-Induced Cytokine Production in Human Brain Astrocytes

  • Kim Bo Kyung;Shin Soon Shik;Kang Seon Tae
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.18 no.1
    • /
    • pp.254-259
    • /
    • 2004
  • Jeongshin-tang (JST) is a Korean herbal prescription, which has been successfully applied for the various neuronal diseases. However, it's effect remains unknown in experimental models. To investigate the biological effect of JST in Alzheimer's disease (AD) in vitro model, we analized the production of interleukin (IL)-6 and IL-8, and expression of cyclooxygenase (COX)-2 in IL-1β plus β-amyloid [25-35] fragment (A)-stimulated human astrocytoma cell line U373MG. JST alone had no effect on the cell viability. The production of IL-6 and IL-8 was significantly inhibited by pretreatment with JST (1mg/㎖) on IL-1β plus A-stimulated U373MG cells. Maximal inhibition rate of IL-6 and IL-8 production by JST was about 41.22% (P<0.01) and 34.45% (P<0.05), respectively. The expression level of COX-2 protein was up-regulated by IL-1β plus A but the increased level of COX-2 was inhibited by pretreatment with JST (1 mg/㎖). These data indicate that JST has a regulatory effect on cytokine production and COX-2 expression, which might explain it's beneficial effect in the treatment of AD.

Attenuation of β-amyloid-induced neuroinflammation by KHG21834 in vivo

  • Kim, Eun-A;Hahn, Hoh-Gyu;Kim, Tae-Ue;Choi, Soo-Young;Cho, Sung-Woo
    • BMB Reports
    • /
    • v.43 no.6
    • /
    • pp.413-418
    • /
    • 2010
  • Beta-Amyloid ($A{\beta}$)-induced neuroinflammation is one of the key events in the development of neurodegenerative disease. We previously reported that KHG21834, a benzothiazole derivative, attenuates $A{\beta}$-induced degeneration of cortical and mesencephalic neurons in vitro. In the present work, we show that KHG21834 reduces $A{\beta}$-mediated neuroinflammation in brain. In vivo intracerebroventricular infusion of KHG21834 leads to decreases in the numbers of activated astrocytes and microglia and level of proinflammatory cytokines such as interleukin-$1{\beta}$ and tumor necrosis factor-$\alpha$ induced by $A{\beta}$ in the hippocampus. This suppression of neuroinflammation is associated with decreased neuron loss, restoration of synaptic dysfunction biomarkers in the hippocampus to control level, and diminished amyloid deposition. These results may suggest the potential therapeutic efficacy of KHG21834 for the treatment of $A{\beta}$-mediated neuroinflammation.

The effect of scopoletin on Aβ-induced neuroinflammatory response in microglial BV-2 cells

  • Mun, Hui-Jin;Cho, Hyun-Jeong
    • Journal of the Korea Society of Computer and Information
    • /
    • v.25 no.6
    • /
    • pp.165-170
    • /
    • 2020
  • In this paper, it was confirmed that scopoletin inhibits neuroinflammation induced by amyloid beta oligomer (Aβ1-42) in microglial BV-2. The mechanisms of inflammatory cytokines and inflammatory mediators by scopoletin were identified. Alzheimer's disease is the most common neurodegenerative disease, but it is a disease whose specific etiology is unknown, and many studies are trying to solve it. We first measured the cell viability with the CCK-8 assay method to confirm that scopoletin and Aβ1-42 are toxic to BV-2 cells. Expression levels of interleukin 1 beta (IL-1β), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and nuclear factor-κB (NF-κB) in inflammatory reactions induced by Aβ1-42 with western blot were analyzed. The ANOVA assay was used to compare protein expression differences between BV-2 cells treated with Aβ1-42 alone and BV-2 cells pretreated with Aβ1-42 and scopoletin. Therefore, this study suggested that scopoletin is worth developing as a neuroinflammatory protection agent for Alzheimer's disease in the future.

Protective effects of kaempferol, quercetin, and its glycosides on amyloid beta-induced neurotoxicity in C6 glial cell (Kaempferol, quercetin 및 그 배당체의 amyloid beta 유도 신경독성에 대한 C6 신경교세포 보호 효과)

  • Kim, Ji Hyun;Kim, Hyun Young;Cho, Eun Ju
    • Journal of Applied Biological Chemistry
    • /
    • v.62 no.4
    • /
    • pp.327-332
    • /
    • 2019
  • Alzheimer's disease (AD) is a common neurodegenerative disease. Oxidative stress by amyloid beta peptide (Aβ) of neuronal cell is the most cause of AD. In the present study, protective effects of several flavonoids such as kaempferol (K), kaempferol-3-O-glucoside (KG), quercetin (Q) and quercetin-3-β-ᴅ-glucoside (QG) from Aβ25-35 were investigated using C6 glial cell. Treatment of Aβ25-35 to C6 glial cell showed decrease of cell viability, while treatment of flavonoids such as Q and QG increased cell viability. In addition, treatment of flavonoids declined reactive oxygen species (ROS) production compared with Aβ25-35-induced control. The ROS production was increased by treatment of Aβ25-35 to 133.39%, while KG and QG at concentration of 1 μM decreased ROS production to 107.44 and 113.10%, respectively. To study mechanisms of protective effect of these flavonoids against Aβ25-35, the protein expression related to inflammation under Aβ25-35-induced C6 glial cell was investigated. The results showed that C6 glial cell under Aβ25-35-induced oxidative stress up-regulated inflammation-related protein expressions. However, treatment of flavonoids led to reduction of protein expression such as inducible nitric oxide synthase, cyclooxygenase-2 and interleukin-1β. Especially, treatment of KG and QG decreased more effectively inflammation-related protein expression than its aglycones, K and Q. Therefore, the present results indicated that K, Q and its glycosides attenuated Aβ25-35-induced neuronal oxidative stress and inflammation.

Protective effects of N,4,5-trimethylthiazol-2-amine hydrochloride on hypoxia-induced β-amyloid production in SH-SY5Y cells

  • Han, A Reum;Yang, Ji Woong;Na, Jung-Min;Choi, Soo Young;Cho, Sung-Woo
    • BMB Reports
    • /
    • v.52 no.7
    • /
    • pp.439-444
    • /
    • 2019
  • Although hypoxic/ischemic injury is thought to contribute to the incidence of Alzheimer's disease (AD), the molecular mechanism that determines the relationship between hypoxia-induced ${\beta}$-amyloid ($A{\beta}$) generation and development of AD is not yet known. We have now investigated the protective effects of N,4,5-trimethylthiazol-2-amine hydrochloride (KHG26702), a novel thiazole derivative, on oxygen-glucose deprivation (OGD)-reoxygenation (OGD-R)-induced $A{\beta}$ production in SH-SY5Y human neuroblastoma cells. Pretreatment of these cells with KHG26702 significantly attenuated OGD-R-induced production of reactive oxygen species and elevation of levels of malondialdehyde, prostaglandin $E_2$, interleukin 6 and glutathione, as well as superoxide dismutase activity. KHG26702 also reduced OGD-R-induced expression of the apoptotic protein caspase-3, the apoptosis regulator Bcl-2, and the autophagy protein becn-1. Finally, KHG26702 reduced OGD-R-induced $A{\beta}$ production and cleavage of amyloid precursor protein, by inhibiting secretase activity and suppressing the autophagic pathway. Although supporting data from in vivo studies are required, our results indicate that KHG26702 may prevent neuronal cell damage from OGD-R-induced toxicity.

Paeoniflorin ameliorates Aβ-stimulated neuroinflammation via regulation of NF-κB signaling pathway and Aβ degradation in C6 glial cells

  • Cho, Eun Ju;Kim, Hyun Young;Lee, Ah Young
    • Nutrition Research and Practice
    • /
    • v.14 no.6
    • /
    • pp.593-605
    • /
    • 2020
  • BACKGROUND/OBJECTIVES: Alzheimer's disease is common age-related neurodegenerative condition characterized by amyloid beta (Aβ) accumulation that leads cognitive impairment. In the present study, we investigated the protective effect of paeoniflorin (PF) against Aβ-induced neuroinflammation and the underlying mechanism in C6 glial cells. MATERIALS/METHODS: C6 glial cells were treated with PF and Aβ25-35, and cell viability, nitric oxide (NO) production, and pro-inflammatory cytokine release were measured. Furthermore, the mechanism underlying the effect of PF on inflammatory responses and Aβ degradation was determined by Western blot. RESULTS: Aβ25-35 significantly reduced cell viability, but this reduction was prevented by the pretreatment with PF. In addition, PF significantly inhibited Aβ25-35-induced NO production in C6 glial cells. The secretion of interleukin (IL)-6, IL-1β, and tumor necrosis factor-alpha was also significantly reduced by PF. Further mechanistic studies indicated that PF suppressed the production of these pro-inflammatory cytokines by regulating the nuclear factor-kappa B (NF-κB) pathway. The protein levels of inducible NO synthase and cyclooxygenase-2 were downregulated and phosphorylation of NF-κB was blocked by PF. However, PF elevated the protein expression of inhibitor kappa B-alpha and those of Aβ degrading enzymes, insulin degrading enzyme and neprilysin. CONCLUSIONS: These findings indicate that PF exerts protective effects against Aβ-mediated neuroinflammation by inhibiting NF-κB signaling, and these effects were associated with the enhanced activity of Aβ degradation enzymes.