1.Sodium valprovate suppresses autophagy in SH-SY5Y cells activating miR-34c-5p/ATG4B signaling pathway.
Xufang DAI ; Xiaojing YAN ; Peng XIE ; Jiqin LIAN
Journal of Southern Medical University 2018;38(12):1415-1420
OBJECTIVE:
To investigate the effect of sodium valproate (VPA) on activation of miR-34c-5p/ATG4B signaling pathway and autophagy in SH-SY5Y cells.
METHODS:
Routinely cultured SH-SY5Y cells were treated with VPA at different doses for 24 h, and the changes in the mRNA levels of ATG4B and miR-34c-5p and the protein expression of ATG4B were assessed using qRTPCR and immunoblotting, respectively. The effect of transfection with a plasmid containing ATG4B promoter on the promoter activity of ATG4B in VPA-treated SH-SY5Y cells was assessed using the reporter gene assay. The stability of ATG4B mRNA was analyzed with qPCR in SH-SY5Y cells treated with VPA alone or with VPA combined with the transcription inhibitor actinomycin D. The expression level of miR-34c-5p was detected using qPCR in SH-SY5Y cells treated with VPA alone or with VPA combined with miR-34c-5p mimics or antagonist, and the role of miR-34c-5p in VPA-induced ATG4B down-regulation was evaluated. The changes in the level of autophagy were evaluated by detecting LC3-Ⅱ expression in the cells after treatment with VPA or VPA combined with miR-34c-5p antagonist.
RESULTS:
VPA dose-dependently down-regulated the expression of ATG4B at both the mRNA and protein levels in SH-SY5Y cells. VPA treatment did not significantly affect the promoter activity of ATG4B, but obviously lowered the mRNA stability of ATG4B in SH-SY5Y cells. VPA treatment up-regulated the expression of miR-34c-5p, and the miR-34c-5p antagonist reversed VPA-induced down-regulation of ATG4B in SH-SY5Y cells. VPA also down-regulated the expression level of LC3-Ⅱ in SH-SY5Y cells.
CONCLUSIONS
VPA suppresses autophagy in SH-SY5Y cells possibly via activating miR-34c-5p/ATG4B signaling pathway.
Autophagy
;
drug effects
;
Autophagy-Related Proteins
;
genetics
;
metabolism
;
Cell Line
;
Cysteine Endopeptidases
;
genetics
;
metabolism
;
Dactinomycin
;
pharmacology
;
Down-Regulation
;
Genes, Reporter
;
Humans
;
MicroRNAs
;
antagonists & inhibitors
;
metabolism
;
Microtubule-Associated Proteins
;
metabolism
;
RNA, Messenger
;
metabolism
;
Signal Transduction
;
drug effects
;
Transfection
;
Valproic Acid
;
administration & dosage
;
antagonists & inhibitors
;
pharmacology
2.Autophagy induction by SIRT6 is involved in oxidative stress-induced neuronal damage.
Jiaxiang SHAO ; Xiao YANG ; Tengyuan LIU ; Tingting ZHANG ; Qian Reuben XIE ; Weiliang XIA
Protein & Cell 2016;7(4):281-290
SIRT6 is a NAD(+)-dependent histone deacetylase and has been implicated in the regulation of genomic stability, DNA repair, metabolic homeostasis and several diseases. The effect of SIRT6 in cerebral ischemia and oxygen/glucose deprivation (OGD) has been reported, however the role of SIRT6 in oxidative stress damage remains unclear. Here we used SH-SY5Y neuronal cells and found that overexpression of SIRT6 led to decreased cell viability and increased necrotic cell death and reactive oxygen species (ROS) production under oxidative stress. Mechanistic study revealed that SIRT6 induced autophagy via attenuation of AKT signaling and treatment with autophagy inhibitor 3-MA or knockdown of autophagy-related protein Atg5 rescued H2O2-induced neuronal injury. Conversely, SIRT6 inhibition suppressed autophagy and reduced oxidative stress-induced neuronal damage. These results suggest that SIRT6 might be a potential therapeutic target for neuroprotection.
Adenine
;
analogs & derivatives
;
toxicity
;
Autophagy
;
drug effects
;
Autophagy-Related Protein 5
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Blotting, Western
;
Cell Line, Tumor
;
Humans
;
Hydrogen Peroxide
;
toxicity
;
Microtubule-Associated Proteins
;
metabolism
;
Oxidative Stress
;
drug effects
;
Proto-Oncogene Proteins c-akt
;
metabolism
;
RNA Interference
;
RNA, Messenger
;
metabolism
;
RNA, Small Interfering
;
metabolism
;
Reactive Oxygen Species
;
metabolism
;
Real-Time Polymerase Chain Reaction
;
Signal Transduction
;
drug effects
;
Sirtuins
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Transfection
3.Molecular mechanism of rhein on inhibiting autophagic protein expression in renal tubular epithelial cells via regulating mTOR signaling pathway activation.
Yue TU ; Wei SUN ; Liu-bao GU ; Yi-Gang WAN ; Hao HU ; Hong LIU
China Journal of Chinese Materia Medica 2014;39(21):4090-4095
OBJECTIVETo explore the effects and molecular mechanisms of rhein on reducing starvation-induced autophagic protein expression in renal tubular epithelial ( NRK-52E) cells.
METHODHank's balanced salt solution (HBSS) was used to induce NRK-52E cells to be in the state of starvation. After the intervention of HBSS for 0, 0.5,1, 2 and 6 hours, firstly, the protein expression of microtubule-associated protein 1 light chain 3(LC3 I/II), which is a key protein in autophagy, was detected. Secondly, the protein expressions of mammalian target of rapamycin (mTOR) and phosphorylated-mTOR Ser2448 (p-mTOR S2448) were examined. And then, after the co-treatment of rhein (5 mg x L(-1)) and HBSS (1 mL) without or with mTOR inhibitor, rapamycin (100 nmol x L(-1)), the protein expressions of LC3 I/II, mTOR and p-mTOR S2448 were tested, respectively.
RESULTHBSS could induce the up-regulation of LC3 II and the down-regulation of p-mTOR S2448 at protein expression level in NRK-52E cells. The co-treatment of rhein and HBSS could reversely regulate the protein expressions of LC3 II and p-mTOR S2448 in NRK-52E cells significantly. The co-treatment of rapamycin, rhein and HBSS could recover the level of LC3 II protein expression in HBSS-intervened NRK-52E cells.
CONCLUSIONHBSS induces autophagy in renal tubular epithelial cells by inhibiting mTOR signaling pathway activation. Rhein reduces the autophagic protein expression in renal tubular epithelial cells through regulating mTOR signaling pathway activation, which is the possible effects and molecular mechanisms.
Animals ; Anthraquinones ; pharmacology ; Autophagy ; drug effects ; Cells, Cultured ; Epithelial Cells ; drug effects ; metabolism ; Isotonic Solutions ; pharmacology ; Kidney Tubules ; drug effects ; metabolism ; Microtubule-Associated Proteins ; genetics ; Rats ; Signal Transduction ; drug effects ; TOR Serine-Threonine Kinases ; antagonists & inhibitors ; genetics ; physiology
4.End-binding protein 1 stimulates paclitaxel sensitivity in breast cancer by promoting its actions toward microtubule assembly and stability.
Youguang LUO ; Dengwen LI ; Jie RAN ; Bing YAN ; Jie CHEN ; Xin DONG ; Zhu LIU ; Ruming LIU ; Jun ZHOU ; Min LIU
Protein & Cell 2014;5(6):469-479
Paclitaxel is a microtubule-targeting agent widely used for the treatment of many solid tumors. However, patients show variable sensitivity to this drug, and effective diagnostic tests predicting drug sensitivity remain to be investigated. Herein, we show that the expression of end-binding protein 1 (EB1), a regulator of microtubule dynamics involved in multiple cellular activities, in breast tumor tissues correlates with the pathological response of tumors to paclitaxel-based chemotherapy. In vitro cell proliferation assays reveal that EB1 stimulates paclitaxel sensitivity in breast cancer cell lines. Our data further demonstrate that EB1 increases the activity of paclitaxel to cause mitotic arrest and apoptosis in cancer cells. In addition, microtubule binding affinity analysis and polymerization/depolymerization assays show that EB1 enhances paclitaxel binding to microtubules and stimulates the ability of paclitaxel to promote microtubule assembly and stabilization. These findings thus reveal EB1 as a critical regulator of paclitaxel sensitivity and have important implications in breast cancer chemotherapy.
Antineoplastic Agents, Phytogenic
;
pharmacology
;
therapeutic use
;
Apoptosis
;
drug effects
;
Breast Neoplasms
;
drug therapy
;
metabolism
;
pathology
;
Cell Cycle Checkpoints
;
drug effects
;
Cell Line, Tumor
;
Female
;
Humans
;
MCF-7 Cells
;
Microtubule-Associated Proteins
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Microtubules
;
chemistry
;
metabolism
;
Paclitaxel
;
pharmacology
;
therapeutic use
;
RNA Interference
;
RNA, Small Interfering
;
metabolism
5.Coronavirus membrane-associated papain-like proteases induce autophagy through interacting with Beclin1 to negatively regulate antiviral innate immunity.
Xiaojuan CHEN ; Kai WANG ; Yaling XING ; Jian TU ; Xingxing YANG ; Qian ZHAO ; Kui LI ; Zhongbin CHEN
Protein & Cell 2014;5(12):912-927
Autophagy plays important roles in modulating viral replication and antiviral immune response. Coronavirus infection is associated with the autophagic process, however, little is known about the mechanisms of autophagy induction and its contribution to coronavirus regulation of host innate responses. Here, we show that the membrane-associated papain-like protease PLP2 (PLP2-TM) of coronaviruses acts as a novel autophagy-inducing protein. Intriguingly, PLP2-TM induces incomplete autophagy process by increasing the accumulation of autophagosomes but blocking the fusion of autophagosomes with lysosomes. Furthermore, PLP2-TM interacts with the key autophagy regulators, LC3 and Beclin1, and promotes Beclin1 interaction with STING, the key regulator for antiviral IFN signaling. Finally, knockdown of Beclin1 partially reverses PLP2-TM's inhibitory effect on innate immunity which resulting in decreased coronavirus replication. These results suggested that coronavirus papain-like protease induces incomplete autophagy by interacting with Beclin1, which in turn modulates coronavirus replication and antiviral innate immunity.
Apoptosis Regulatory Proteins
;
antagonists & inhibitors
;
genetics
;
immunology
;
Autophagy
;
Beclin-1
;
Coronavirus NL63, Human
;
genetics
;
immunology
;
Gene Expression Regulation
;
HEK293 Cells
;
HeLa Cells
;
Host-Pathogen Interactions
;
immunology
;
Humans
;
Immune Evasion
;
Immunity, Innate
;
Interferon-gamma
;
genetics
;
immunology
;
Lysosomes
;
metabolism
;
virology
;
MCF-7 Cells
;
Membrane Fusion
;
Membrane Proteins
;
antagonists & inhibitors
;
genetics
;
immunology
;
Microtubule-Associated Proteins
;
genetics
;
immunology
;
Papain
;
genetics
;
immunology
;
Phagosomes
;
metabolism
;
virology
;
RNA, Small Interfering
;
genetics
;
immunology
;
Signal Transduction
;
Virus Replication
6.Salidroside via ERK1/2 and PI3K/AKT/mTOR signal pathway induces mouse bone marrow mesenchymal stem cells differentiation into neural cells.
Ya-Nan CHEN ; Hui LIU ; Hong-Bin ZHAO ; Yang LIU ; Jie BAI ; Xiao-Juan ZHU ; Yu WANG
Acta Pharmaceutica Sinica 2013;48(8):1247-1252
To investigate the role of the extracellular signal-regulated kinase (ERK1/2) and PI3K/AKT/ mTOR signal pathway inducing bone marrow mesenchymal stem cells (BMSCs) differentiation into neural cells, mouse bone marrow-derived mesenchymal stem cell lines D1 cells were used as research object. And they were divided into control groups and salidroside (SD) groups. Different concentrations (5, 25, 50, 100 and 200 microg x mL(-1) of SD were used and SD (100 microg x mL(-1)) was used to induce at different time (0.5, 1, 3, 6, 9, 12, 24, 48 and 72 h). The immunofluorescence staining chemical technology, real-time PCR and Western blotting were used to detect the positive rates of NSE, MAP2, beta-Tubulin III, NES, GFAP and the expression levels of beta-Tubulin III, NSE, ERK1/2, AKT. The expression of ERK1/2 and NSE was detected when the ERK1/2 and PI3K/AKT/ mTOR signal pathway was blocked by PD98059 and LY294002. It indicated that the positive rates of NSE, MAP2, beta-Tubulin III, NES and GFAP were gradually enhanced with time increased. The expression level of NSE and beta-Tubulin III protein were significantly higher than those in control groups (P < 0.01). The expression of ERK1/2, AKT mRNA and protein were higher with concentration and time increased. When the ERK1/2 and PI3K/AKT/mTOR signal pathway were blocked, the expression levels of NSE, NES and beta-Tubulin III mRNA and NSE protein were inhibited significantly. It points out that SD can stimulate the ERK1/2 and PI3K/AKT/mTOR signal pathway to promote BMSCs differentiation into neural cells.
Animals
;
Bone Marrow Cells
;
cytology
;
Cell Differentiation
;
drug effects
;
Cells, Cultured
;
Chromones
;
pharmacology
;
Enzyme Inhibitors
;
pharmacology
;
Flavonoids
;
pharmacology
;
Glial Fibrillary Acidic Protein
;
metabolism
;
Glucosides
;
antagonists & inhibitors
;
isolation & purification
;
pharmacology
;
MAP Kinase Signaling System
;
drug effects
;
Mesenchymal Stromal Cells
;
cytology
;
Mice
;
Microtubule-Associated Proteins
;
metabolism
;
Mitogen-Activated Protein Kinase 1
;
genetics
;
metabolism
;
Mitogen-Activated Protein Kinase 3
;
genetics
;
metabolism
;
Morpholines
;
pharmacology
;
Nestin
;
metabolism
;
Neurons
;
cytology
;
metabolism
;
Phenols
;
antagonists & inhibitors
;
isolation & purification
;
pharmacology
;
Phosphatidylinositol 3-Kinases
;
metabolism
;
Phosphopyruvate Hydratase
;
genetics
;
metabolism
;
Plants, Medicinal
;
chemistry
;
Protein Kinase Inhibitors
;
pharmacology
;
Proto-Oncogene Proteins c-akt
;
genetics
;
metabolism
;
RNA, Messenger
;
metabolism
;
Rhodiola
;
chemistry
;
Signal Transduction
;
drug effects
;
TOR Serine-Threonine Kinases
;
metabolism
;
Tubulin
;
metabolism
7.ULK1 and JNK are involved in mitophagy incurred by LRRK2 G2019S expression.
Yuangang ZHU ; Chunyan WANG ; Mei YU ; Jie CUI ; Liang LIU ; Zhiheng XU
Protein & Cell 2013;4(9):711-721
Mutations in LR RK2 (Leucine rich repeat kinase 2) are a major cause of Parkinson's disease (PD). We and others reported recently that expression of the pathogenic gainof-function mutant form of LRRK2, LRRK2 G2019S, induces mitochondrial fission in neurons through DLP1. Here we provide evidence that expression of LRRK2 G2019S stimulates mitochondria loss or mitophagy. We have characterized several LRRK2 interacting proteins and found that LRRK2 interacts with ULK1 which plays an essential role in autophagy. Knockdown of either ULK1 or DLP1 expression with shRNAs suppresses LRRK2 G2019S expression-induced mitochondrial clearance, suggesting that LRRK2 G2019S expression induces mitochondrial fission through DLP1 followed by mitophagy via an ULK1 dependent pathway. In addition to ULK1, we found that LRRK2 interacts with the endogenous MKK4/7, JIP3 and coordinates with them in the activation of JNK signaling. Interestingly, LRRK2 G2019S-induced loss of mitochondria can also be suppressed by 3 different JNK inhibitors, implying the involvement of the JNK pathway in the pathogenic mechanism of mutated LRRK2. Thus our findings may provide an insight into the complicated pathogenesis of PD as well as some clues to the development of novel therapeutic strategies.
Amino Acid Substitution
;
Autophagosomes
;
metabolism
;
pathology
;
Autophagy-Related Protein-1 Homolog
;
chemistry
;
genetics
;
metabolism
;
GTP Phosphohydrolases
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Gene Knockdown Techniques
;
HeLa Cells
;
Humans
;
Intracellular Signaling Peptides and Proteins
;
chemistry
;
genetics
;
metabolism
;
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2
;
chemistry
;
genetics
;
metabolism
;
MAP Kinase Signaling System
;
Microtubule-Associated Proteins
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Mitochondrial Degradation
;
genetics
;
physiology
;
Mitochondrial Proteins
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Mutant Proteins
;
chemistry
;
genetics
;
metabolism
;
Mutation
;
Parkinson Disease
;
genetics
;
metabolism
;
pathology
;
Protein Interaction Domains and Motifs
;
Recombinant Proteins
;
chemistry
;
genetics
;
metabolism
8.Regulation of tumor angiogenesis by the microtubule-binding protein CLIP-170.
Xiaodong SUN ; Fang LI ; Bin DONG ; Shaojun SUO ; Min LIU ; Dengwen LI ; Jun ZHOU
Protein & Cell 2013;4(4):266-276
Angiogenesis, the expansion of preexisting blood vessels, is a complex process required for tumor growth and metastasis. Although current antiangiogenic strategies have shown promising results in several cancer types, identification of additional antiangiogenic targets is required to improve the therapeutic response. Herein, we show that the microtubule-binding protein CLIP-170 (cytoplasmic linker protein of 170 kDa) is highly expressed in breast tumor samples and correlates positively with blood vessel density. Depletion of CLIP-170 significantly impaired vascular endothelial tube formation and sprouting in vitro and inhibited breast tumor growth in mice by decreasing tumor vascularization. Our data further show that CLIP-170 is important for the migration but not the proliferation of vascular endothelial cells. In addition, CLIP-170 promotes the polarization of endothelial cells in response to the angiogenic stimulus. These findings thus demonstrate a critical role for CLIP-170 in tumor angiogenesis and suggest its potential as a novel antiangiogenic target.
Animals
;
Breast Neoplasms
;
blood supply
;
metabolism
;
pathology
;
Cell Line, Tumor
;
Cell Movement
;
Cell Polarity
;
Female
;
Human Umbilical Vein Endothelial Cells
;
Humans
;
MCF-7 Cells
;
Mice
;
Mice, Nude
;
Microtubule-Associated Proteins
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Microtubules
;
metabolism
;
Neoplasm Proteins
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Neovascularization, Pathologic
;
RNA Interference
;
RNA, Small Interfering
;
metabolism
;
Transplantation, Heterologous
9.Inhibition of survivin expression and mechanisms of reversing drug-resistance of human lung adenocarcinoma cells by siRNA.
Jing-lei LIU ; Yan WANG ; Ji JIANG ; Rui KONG ; Yan-mei YANG ; Hong-fei JI ; Yu-zhi SHI
Chinese Medical Journal 2010;123(20):2901-2907
BACKGROUNDSurvivin, a member of the inhibitor of apoptosis protein (IAP) family, overexpresses in tumor cells and not expresses in terminally differentiated adult tissues. This study aimed to investigate the effects of survivin-specific siRNA on cell proliferation, apoptosis and chemosensitivity to cisplatin in vitro and in vivo and explore the mechanisms about decreasing expression of survivin in reversing cancer cells resistance to chemotherapeutic drug.
METHODSSurvivin-specific siRNA was transfected into A549/DDP cells. The expression of survivin and lung resistance-related protein (LRP) mRNA levels were determined by RT-PCR, chemosensitivity of A549/DDP (cisplatin) cells to cisplatin was determined by MTT assay, and apoptosis and cell cycle were determined by flow cytometry (FCM). The protein expression levels of survivin, LRP, cyclin-D(1), caspase-3 and bcl-2 were determined by Western blotting analyses. The effect of survivin siRNA inhibition on tumor growth was studied in athymic nude mice in vivo.
RESULTSSurvivin-specific siRNA efficiently down-regulated survivin expression. The cell cycle was arrested at G2/M phase, and apoptosis was obviously found. Inhibition of survivin expression could make the IC50 and drug-resistant index of cisplatin decrease, and enhance the cancer cells sensitivity to cisplatin. After transfection by survivin-specific siRNA, expression of LRP and cyclin-D1 were downregulated, caspase-3 expression was upregulated, bcl-2 expression had no obvious change. The animal experiment confirmed knockdown of survivin could inhibit the tumor growth.
CONCLUSIONSSurvivin-specific siRNA can efficiently suppress the expression of survivin, increase apoptosis, inhibit cells proliferation and enhance the chemosensitivity to cisplatin in vitro and in vivo. Suppression of survivin expression helping to reverse drug-resistance may have relationship with downregulation of LRP and upregulation of caspase-3. Anti-tumor strategies based on the inhibition of survivin may be useful in targeting lung adenocarcinomas.
Adenocarcinoma ; drug therapy ; pathology ; Animals ; Apoptosis ; Caspase 3 ; analysis ; Cell Line, Tumor ; Cisplatin ; pharmacology ; Cyclin D1 ; analysis ; Drug Resistance, Neoplasm ; Female ; Humans ; Inhibitor of Apoptosis Proteins ; Lung Neoplasms ; drug therapy ; pathology ; Male ; Mice ; Mice, Inbred BALB C ; Microtubule-Associated Proteins ; antagonists & inhibitors ; genetics ; Proto-Oncogene Proteins c-bcl-2 ; analysis ; RNA, Messenger ; analysis ; RNA, Small Interfering ; genetics ; Vault Ribonucleoprotein Particles ; genetics
10.Tunicamycin enhances TRAIL-induced apoptosis by inhibition of cyclin D1 and the subsequent downregulation of survivin.
Hai Yan ZHANG ; Zhen Xian DU ; Bao Qin LIU ; Yan Yan GAO ; Xin MENG ; Yifu GUAN ; Wei Wei DENG ; Hua Qin WANG
Experimental & Molecular Medicine 2009;41(5):362-369
TNF-related apoptosis-inducing ligand (TRAIL) has been proposed as a promising cancer therapy that preferentially induces apoptosis in cancer cells, but not most normal tissues. However, many cancers are resistant to TRAIL by mechanisms that are poorly understood. In this study, we showed that tunicamycin, a naturally occurring antibiotic, was a potent enhancer of TRAIL-induced apoptosis through downregulation of survivin. The tunicamycin-mediated sensitization to TRAIL was efficiently reduced by forced expression of survivin, suggesting that the sensitization was mediated at least in part through inhibition of survivin expression. Tunicamycin also repressed expression of cyclin D1, a cell cycle regulator commonly overexpressed in thyroid carcinoma. Furthermore, silencing cyclin D1 by RNA interference reduced survivin expression and sensitized thyroid cancer cells to TRAIL; in contrast, forced expression of cyclin D1 attenuated tunicamycin-potentiated TRAIL-induced apoptosis via over-riding downregulation of survivin. Collectively, our results demonstrated that tunicamycin promoted TRAIL-induced apoptosis, at least in part, by inhibiting the expression of cyclin D1 and subsequent survivin. Of note, tunicamycin did not sensitize the differentiated thyroid epithelial cells to TRAIL-induced apoptosis. Thus, combined treatment with tunicamycin and TRAIL may offer an attractive strategy for safely treating resistant thyroid cancers.
Anti-Bacterial Agents/*pharmacology
;
*Apoptosis
;
Cell Line, Tumor
;
Cyclin D1/*antagonists & inhibitors/metabolism
;
*Down-Regulation
;
Humans
;
Microtubule-Associated Proteins/*genetics/metabolism
;
TNF-Related Apoptosis-Inducing Ligand/*metabolism
;
Tunicamycin/*pharmacology

Result Analysis
Print
Save
E-mail