• 제목/요약/키워드: Glycogen synthase kinase $3{\beta}$ (GSK3 ${\beta}$)

검색결과 52건 처리시간 0.024초

Regulatory B Subunits of Protein Phosphatase 2A Are Involved in Site-specific Regulation of Tau Protein Phosphorylation

  • Yu, Un Young;Yoo, Byong Chul;Ahn, Jung-Hyuck
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제18권2호
    • /
    • pp.155-161
    • /
    • 2014
  • Overexpression of amyloid precursor protein with the Swedish mutation causes abnormal hyperphosphorylation of the microtubule-associated protein tau. Hyperphosphorylated isoforms of tau are major components of neurofibrillary tangles, which are histopathological hallmarks of Alzheimer's disease. Protein phosphatase 2A (PP2A), a major tau protein phosphatase, consists of a structural A subunit, catalytic C subunit, and a variety of regulatory B subunits. The B subunits have been reported to modulate function of the PP2A holoenzyme by regulating substrate binding, enzyme activity, and subcellular localization. In the current study, we characterized regulatory B subunit-specific regulation of tau protein phosphorylation. We showed that the PP2A B subunit PPP2R2A mediated dephosphorylation of tau protein at Ser-199, Ser-202/Thr-205, Thr-231, Ser-262, and Ser-422. Down-regulation of PPP2R5D expression decreased tau phosphorylation at Ser-202/Thr-205, Thr-231, and Ser-422, which indicates activation of the tau kinase glycogen synthase kinase 3 beta ($GSK3{\beta}$) by PP2A with PPP2R5D subunit. The level of activating phosphorylation of the $GSK3{\beta}$ kinase Akt at Thr-308 and Ser-473 were both increased by PPP2R5D knockdown. We also characterized B subunit-specific phosphorylation sites in tau using mass spectrometric analysis. Liquid chromatography-mass spectrometry revealed that the phosphorylation status of the tau protein may be affected by PP2A, depending on the specific B subunits. These studies further our understanding of the function of various B subunits in mediating site-specific regulation of tau protein phosphorylation.

The Anti-Inflammatory Activity of Eucommia ulmoides Oliv. Bark. Involves NF-κB Suppression and Nrf2-Dependent HO-1 Induction in BV-2 Microglial Cells

  • Kwon, Seung-Hwan;Ma, Shi-Xun;Hwang, Ji-Young;Ko, Yong-Hyun;Seo, Ji-Yeon;Lee, Bo-Ram;Lee, Seok-Yong;Jang, Choon-Gon
    • Biomolecules & Therapeutics
    • /
    • 제24권3호
    • /
    • pp.268-282
    • /
    • 2016
  • In the present study, we investigated the anti-inflammatory properties of Eucommia ulmoides Oliv. Bark. (EUE) in lipopolysaccharide (LPS)-stimulated microglial BV-2 cells and found that EUE inhibited LPS-mediated up-regulation of pro-inflammatory response factors. In addition, EUE inhibited the elevated production of pro-inflammatory cytokines, mediators, and reactive oxygen species (ROS) in LPS-stimulated BV-2 microglial cells. Subsequent mechanistic studies revealed that EUE suppressed LPS-induced phosphorylation of mitogen-activated protein kinases (MAPKs), phosphoinositide-3-kinase (PI3K)/Akt, glycogen synthase $kinase-3{\beta}$ ($GSK-3{\beta}$), and their downstream transcription factor, nuclear factor-kappa B ($NF-{\kappa}B$). EUE also blocked the nuclear translocation of $NF-{\kappa}B$ and inhibited its binding to DNA. We next demonstrated that EUE induced the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and upregulated heme oxygenase-1 (HO-1) expression. We determined that the significant up-regulation of HO-1 expression by EUE was a consequence of Nrf2 nuclear translocation; furthermore, EUE increased the DNA binding of Nrf2. In contrast, zinc protoporphyrin (ZnPP), a specific HO-1 inhibitor, blocked the ability of EUE to inhibit NO and $PGE_2$ production, indicating the vital role of HO-1. Overall, our results indicate that EUE inhibits pro-inflammatory responses by modulating MAPKs, PI3K/Akt, and $GSK-3{\beta}$, consequently suppressing $NF-{\kappa}B$ activation and inducing Nrf2-dependent HO-1 activation.

Virtual Screening and Testing of GSK-3 Inhibitors Using Human SH-SY5Y Cells Expressing Tau Folding Reporter and Mouse Hippocampal Primary Culture under Tau Cytotoxicity

  • Chih-Hsin Lin;Yu-Shao Hsieh;Ying-Chieh Sun;Wun-Han Huang;Shu-Ling Chen;Zheng-Kui Weng;Te-Hsien Lin;Yih-Ru Wu;Kuo-Hsuan Chang;Hei-Jen Huang;Guan-Chiun Lee;Hsiu Mei Hsieh-Li;Guey-Jen Lee-Chen
    • Biomolecules & Therapeutics
    • /
    • 제31권1호
    • /
    • pp.127-138
    • /
    • 2023
  • Glycogen synthase kinase-3β (GSK-3β) is an important serine/threonine kinase that implicates in multiple cellular processes and links with the neurodegenerative diseases including Alzheimer's disease (AD). In this study, structure-based virtual screening was performed to search database for compounds targeting GSK-3β from Enamine's screening collection. Of the top-ranked compounds, 7 primary hits underwent a luminescent kinase assay and a cell assay using human neuroblastoma SH-SY5Y cells expressing Tau repeat domain (TauRD) with pro-aggregant mutation ΔK280. In the kinase assay for these 7 compounds, residual GSK-3β activities ranged from 36.1% to 90.0% were detected at the IC50 of SB-216763. In the cell assay, only compounds VB-030 and VB-037 reduced Tau aggregation in SH-SY5Y cells expressing ΔK280 TauRD-DsRed folding reporter. In SH-SY5Y cells expressing ΔK280 TauRD, neither VB-030 nor VB-037 increased expression of GSK-3α Ser21 or GSK-3β Ser9. Among extracellular signal-regulated kinase (ERK), AKT serine/threonine kinase 1 (AKT), mitogen-activated protein kinase 14 (P38) and mitogenactivated protein kinase 8 (JNK) which modulate Tau phosphorylation, VB-037 attenuated active phosphorylation of P38 Thr180/ Tyr182, whereas VB-030 had no effect on the phosphorylation status of ERK, AKT, P38 or JNK. However, both VB-030 and VB-037 reduced endogenous Tau phosphorylation at Ser202, Thr231, Ser396 and Ser404 in neuronally differentiated SH-SY5Y expressing ΔK280 TauRD. In addition, VB-030 and VB-037 further improved neuronal survival and/or neurite length and branch in mouse hippocampal primary culture under Tau cytotoxicity. Overall, through inhibiting GSK-3β kinase activity and/or p-P38 (Thr180/Tyr182), both compounds may serve as promising candidates to reduce Tau aggregation/cytotoxicity for AD treatment.

AGS 인체 위암 세포에서 Akt/mTOR/GSK-3β 신호경로 조절을 통한 개똥쑥 추출물의 Apoptosis 유도 효과 (Apoptosis-Induced Effects of Extract from Artemisia annua Linné by Modulating Akt/mTOR/GSK-3β Signal Pathway in AGS Human Gastric Carcinoma Cells)

  • 김은지;김근태;김보민;임은경;김상용;김영민
    • 한국식품영양과학회지
    • /
    • 제45권9호
    • /
    • pp.1257-1264
    • /
    • 2016
  • 개똥쑥은 예로부터 항암, 항바이러스 및 항균의 효능을 지니는 것으로 알려져 왔지만 작용 기작에 대한 내용이 많이 알려지지 않았다. 본 연구에서는 AGS 인체 위암 세포를 대상으로 개똥쑥 추출물(AAE)에 의한 apoptosis 효과와 신호경로 연구를 시행하였다. AAE의 암세포 성장에 미치는 영향을 확인하기 위하여 AGS cell에 AAE를 처리하고 MTT assay와 LDH assay를 수행한 결과 AAE 농도 의존적으로 나타난 세포 성장 억제가 세포 손상에 의한 것임을 확인하였다. 또한, AAE에 의한 암세포 증식 억제 효과가 apoptosis에 의한 것인지 확인하기 위하여 Hoechst 33342 staining과 Annexin V-PI staining을 수행한 결과, Hoechst 33342 staining에서 apoptotic body와 세포질 응축이 농도 의존적으로 증가하는 것을 확인하였고, Annexin V-PI staining에서 apoptotic cells의 변화가 농도 의존적으로 증가함을 확인하였다. Western blotting의 결과 AAE가 농도 의존적으로 세포 생장에 관여하는 신호 단백질인 p-Akt, p-TSC2, p-mTOR, p-GSK-$3{\beta}$의 발현이 감소함을 확인하였고, anti-apoptotic 단백질인 Bcl-2의 발현이 억제됨으로써 proapoptotic 단백질인 Bax, Bak의 발현이 증가하는 일련의 신호경로를 조절할 수 있다는 것을 확인하였다. 미토콘드리아 막 전위의 탈분극 유도를 확인하기 위한 JC-1 assay 수행 결과, AAE 농도 의존적으로 미토콘드리아 막 전위의 탈분극이 유도됨을 확인하였다. 탈분극에 의한 caspase 활성을 확인하기 위해 caspase-3/7 activity assay를 수행한 결과, AAE 농도 의존적으로 caspase activity 증가를 확인하였다. 또한, apoptosis가 일어나는 일련의 신호경로를 확인하기 위해 apoptosis 상위 단백질인 Akt, mTOR, GSK-$3{\beta}$의 활성을 억제하는 LY294002, Rapamycin, BIO를 각각 AGS cell에 처리하고 세포증식에 미치는 영향과 신호 단백질의 발현 양상을 알아보기 위해 MTT assay, LDH assay, western blotting을 수행하였다. 그 결과 AAE와 LY294002, Rapamycin 처리군에서 세포증식 억제와 LDH 방출량 증가뿐만 아니라 세포 생장 신호 단백질인 p-mTOR, p-TSC2, p-Akt, p-GSK-$3{\beta}$의 발현이 감소하는 것을 확인하였고, Bcl-2의 발현이 억제됨으로써 Bax와 Bak의 발현을 증가시키는 신호경로를 조절할 수 있다는 것을 확인하였다. 따라서 AGS cell에 개똥쑥 추출물을 처리하였을 때 유도되는 apoptosis 효과는 Akt/mTOR/GSK-$3{\beta}$ 경로 활성 억제를 통해 Bcl-2 발현이 감소함에 따라 Bax, Bak를 활성화해 세포질로의 cytochrome C 유리에 따른 caspase 활성으로 이루어진다는 것을 알 수 있었다.

Cytoprotective effect exerted by geraniin in HepG2 cells is through microRNA mediated regulation of BACH-1 and HO-1

  • Aayadi, Hoda;Mittal, Smriti P.K.;Deshpande, Anjali;Gore, Makarand;Ghaskadbi, Saroj S.
    • BMB Reports
    • /
    • 제50권11호
    • /
    • pp.560-565
    • /
    • 2017
  • Geraniin, a hydrolysable tannin, used in traditional medicine in Southeast Asia, is known to exhibit various biological activities. As an antioxidant it is known to up-regulate phase II enzyme Heme oxygenase-1 (HO-1). However its mechanism is not clearly understood. Nuclear factor erythroid-derived 2 related factor 2 (Nrf-2) is transcriptionally up-regulated by Extracellular signal-regulated kinase (ERK) 1/2 and retained in nucleus due to inactivated Glycogen synthase kinase 3 beta ($GSK-3{\beta}$). Geraniin additionally down-regulates expression of microRNA 217 and 377 (miR-217 and miR-377) which target HO-1 mRNA. Expression of BTB and CNC homolog 1 (BACH-1), another regulator of HO-1, is also down-regulated by up-regulating microRNA 98 (miR-98), a negative regulator of BACH-1. Thus, geraniin up-regulates HO-1 expression both through activating its positive regulator Nrf-2 and by down-regulating its negative regulator BACH-1. Up-regulation of HO-1 also confers protection to HepG2 cells from tertiary butyl hydroperoxide (TBH) induced cytotoxicity.

Niclosamide Inhibits Aortic Valve Interstitial Cell Calcification by Interfering with the GSK-3β/β-Catenin Signaling Pathway

  • Radhika Adhikari;Saugat Shiwakoti;Eunmin Kim;Ik Jun Choi;Sin-Hee Park;Ju-Young Ko;Kiyuk Chang;Min-Ho Oak
    • Biomolecules & Therapeutics
    • /
    • 제31권5호
    • /
    • pp.515-525
    • /
    • 2023
  • The most common heart valve disorder is calcific aortic valve stenosis (CAVS), which is characterized by a narrowing of the aortic valve. Treatment with the drug molecule, in addition to surgical and transcatheter valve replacement, is the primary focus of researchers in this field. The purpose of this study is to determine whether niclosamide can reduce calcification in aortic valve interstitial cells (VICs). To induce calcification, cells were treated with a pro-calcifying medium (PCM). Different concentrations of niclosamide were added to the PCM-treated cells, and the level of calcification, mRNA, and protein expression of calcification markers was measured. Niclosamide inhibited aortic valve calcification as observed from reduced alizarin red s staining in niclosamide treated VICs and also decreased the mRNA and protein expressions of calcification-specific markers: runt-related transcription factor 2 and osteopontin. Niclosamide also reduced the formation of reactive oxygen species, NADPH oxidase activity and the expression of Nox2 and p22phox. Furthermore, in calcified VICs, niclosamide inhibited the expression of β-catenin and phosphorylated glycogen synthase kinase (GSK-3β), as well as the phosphorylation of AKT and ERK. Taken together, our findings suggest that niclosamide may alleviate PCM-induced calcification, at least in part, by targeting oxidative stress mediated GSK-3β/β-catenin signaling pathway via inhibiting activation of AKT and ERK, and may be a potential treatment for CAVS.

Down-regulation of EZH2 by RNA Interference Inhibits Proliferation and Invasion of ACHN Cells via the Wnt/β-catenin Pathway

  • Yuan, Jun-Bin;Yang, Luo-Yan;Tang, Zheng-Yan;Zu, Xiong-Bing;Qi, Lin
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제13권12호
    • /
    • pp.6197-6201
    • /
    • 2012
  • Although enhancer of zeste homolog 2 (EZH2) has been reported as an independent prognostic factor in renal cell carcinoma (RCC), little is known about the exact mechanism of EZH2 in promoting the genesis of RCC. However, several studies have shown that dysregulation of the Wnt/${\beta}$-catenin signaling pathway plays a crucial role. Therefore, we determined whether EZH2 could affect ACHN human RCC cell proliferation and invasion via the Wnt/${\beta}$-catenin pathway. In the present study, we investigated the effects of short interfering RNA (siRNA)-mediated EZH2 gene silencing on Wnt/${\beta}$-catenin signaling in ACHN cells. EZH2-siRNA markedly inhibited the proliferation and invasion capabilities of ACHN, while also reducing the expression of EZH2, Wnt3a and ${\beta}$-catenin. In contrast, cellular expression of GSK-$3{\beta}$ (glycogen synthase kinase-$3{\beta}$), an inhibitor of the Wnt/${\beta}$-catenin pathway, was conspicuously higher after transfection of EZH2 siRNA. These preliminary findings suggest EZH2 may promote proliferation and invasion of ACHN cells via action on the Wnt/${\beta}$-catenin signaling pathway.

Swedish mutation within amyloid precursor protein modulates global gene expression towards the pathogenesis of Alzheimer's disease

  • Shin, Jong-Yeon;Yu, Saet-Byeol;Yu, Un-Young;Ahnjo, Sang-Mee;Ahn, Jung-Hyuck
    • BMB Reports
    • /
    • 제43권10호
    • /
    • pp.704-709
    • /
    • 2010
  • The Swedish mutation (K595N/M596L) of amyloid precursor protein (APP-swe) has been known to increase abnormal cleavage of cellular APP by Beta-secretase (BACE), which causes tau protein hyperphosphorylation and early-onset Alzheimer's disease (AD). Here, we analyzed the effect of APP-swe in global gene expression using deep transcriptome sequencing technique. We found 283 genes were down-regulated and 348 genes were up-regulated in APP-swe expressing H4-swe cells compared to H4 wild-type cells from a total of approximately 74 million reads of 38 base pairs from each transcriptome. Two independent mechanisms such as kinase and phosphatase signaling cascades leading hyperphosphorylation of tau protein were regulated by the expression of APP-swe. Expressions of catalytic subunit as well as several regulatory subunits of protein phosphatases 2A were decreased. In contrast, expressions of tau-phosphorylating glycogen synthase kinase $3\beta$(GSK-3$\beta$), cyclin dependent kinase 5 (CDK5), and cAMP-dependent protein kinase A (PKA) catalytic subunit were increased. Moreover, the expression of AD-related Aquaporin 1 and presenilin 2 expression was regulated by APP-swe. Taken together, we propose that the expression of APP-swe modulates global gene expression directed to AD pathogenesis.

20(S)-protopanaxadiol promotes the migration, proliferation, and differentiation of neural stem cells by targeting GSK-3β in the Wnt/GSK-3β/β-catenin pathway

  • Lin, Kaili;Liu, Bin;Lim, Sze-Lam;Fu, Xiuqiong;Sze, Stephen C.W.;Yung, Ken K.L.;Zhang, Shiqing
    • Journal of Ginseng Research
    • /
    • 제44권3호
    • /
    • pp.475-482
    • /
    • 2020
  • Background: Active natural ingredients, especially small molecules, have recently received wide attention as modifiers used to treat neurodegenerative disease by promoting neurogenic regeneration of neural stem cell (NSC) in situ. 20(S)-protopanaxadiol (PPD), one of the bioactive ingredients in ginseng, possesses neuroprotective properties. However, the effect of PPD on NSC proliferation and differentiation and its mechanism of action are incompletely understood. Methods: In this study, we investigated the impact of PPD on NSC proliferation and neuronal lineage differentiation through activation of the Wnt/glycogen synthase kinase (GSK)-3β/β-catenin pathway. NSC migration and proliferation were investigated by neurosphere assay, Cell Counting Kit-8 assay, and EdU assay. NSC differentiation was analyzed by Western blot and immunofluorescence staining. Involvement of the Wnt/GSK3β/β-catenin pathway was examined by molecular simulation and Western blot and verified using gene transfection. Results: PPD significantly promoted neural migration and induced a significant increase in NSC proliferation in a time- and dose-dependent manner. Furthermore, a remarkable increase in anti-microtubule-associated protein 2 expression and decrease in nestin protein expression were induced by PPD. During the differentiation process, PPD targeted and stimulated the phosphorylation of GSK-3β at Ser9 and the active forms of β-catenin, resulting in activation of the Wnt/GSK-3β/β-catenin pathway. Transfection of NSCs with a constitutively active GSK-3β mutant at S9A significantly hampered the proliferation and neural differentiation mediated by PPD. Conclusion: PPD promotes NSC proliferation and neural differentiation in vitro via activation of the Wnt/GSK-3β/β-catenin pathway by targeting GSK-3β, potentially having great significance for the treatment of neurodegenerative diseases.

Static magnetic fields promote osteoblastic/cementoblastic differentiation in osteoblasts, cementoblasts, and periodontal ligament cells

  • Kim, Eun-Cheol;Park, Jaesuh;Kwon, Il Keun;Lee, Suk-Won;Park, Su-Jung;Ahn, Su-Jin
    • Journal of Periodontal and Implant Science
    • /
    • 제47권5호
    • /
    • pp.273-291
    • /
    • 2017
  • Purpose: Although static magnetic fields (SMFs) have been used in dental prostheses and osseointegrated implants, their biological effects on osteoblastic and cementoblastic differentiation in cells involved in periodontal regeneration remain unknown. This study was undertaken to investigate the effects of SMFs (15 mT) on the osteoblastic and cementoblastic differentiation of human osteoblasts, periodontal ligament cells (PDLCs), and cementoblasts, and to explore the possible mechanisms underlying these effects. Methods: Differentiation was evaluated by measuring alkaline phosphatase (ALP) activity, mineralized nodule formation based on Alizarin red staining, calcium content, and the expression of marker mRNAs assessed by reverse transcription polymerase chain reaction (RT-PCR). Signaling pathways were analyzed by western blotting and immunocytochemistry. Results: The activities of the early marker ALP and the late markers matrix mineralization and calcium content, as well as osteoblast- and cementoblast-specific gene expression in osteoblasts, PDLCs, and cementoblasts were enhanced. SMFs upregulated the expression of Wnt proteins, and increased the phosphorylation of glycogen synthase $kinase-3{\beta}$ ($GSK-3{\beta}$) and total ${\beta}-catenin$ protein expression. Furthermore, p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK), and nuclear $factor-{\kappa}B$ ($NF-{\kappa}B$) pathways were activated. Conclusions: SMF treatment enhanced osteoblastic and/or cementoblastic differentiation in osteoblasts, cementoblasts, and PDLCs. These findings provide a molecular basis for the beneficial osteogenic and/or cementogenic effect of SMFs, which could have potential in stimulating bone or cementum formation during bone regeneration and in patients with periodontal disease.