1.Regulation of HMGB1 release by inflammasomes.
Ben LU ; Haichao WANG ; Ulf ANDERSSON ; Kevin J TRACEY
Protein & Cell 2013;4(3):163-167
High mobility group box 1 (HMGB1) is an evolutionarily conserved non-histone chromatin-binding protein. During infection or injury, activated immune cells and damaged cells release HMGB1 into the extracellular space, where HMGB1 functions as a proinflammatory mediator and contributes importantly to the pathogenesis of inflammatory diseases. Recent studies reveal that inflammasomes, intracellular protein complexes, critically regulate HMGB1 release from activated immune cells in response to a variety of exogenous and endogenous danger signals. Double stranded RNA dependent kinase (PKR), an intracellular danger-sensing molecule, physically interacts with inflammasome components and is important for inflammasome activation and HMGB1 release. Together, these studies not only unravel novel mechanisms of HMGB1 release during inflammation, but also provide potential therapeutic targets to treat HMGB1-related inflammatory diseases.
HMGB1 Protein
;
chemistry
;
metabolism
;
Humans
;
Inflammasomes
;
metabolism
;
Macrophages
;
immunology
;
metabolism
;
eIF-2 Kinase
;
metabolism
2.Role of PERK/eIF2a signaling pathway in hepatocyte apoptosis of alcoholic liver injury rats.
Xiang-hui HAN ; Jian-yi WANG ; Lei WANG ; Pei-yong ZHENG ; Guang JI
Chinese Journal of Hepatology 2010;18(10):768-772
OBJECTIVETo investigate the role of PERK/eIF2alpha signaling pathway in hepatocyte apoptosis of alcoholic liver injury rats.
METHODSRat models with ethanol-induced liver injury were successfully developed by gastric gavage with ethanol-corn oil mixtures for 12 weeks. At different time points (4, 6, 10, 12 week), liver pathology was dynamically observed. The hepatocyte apoptosis was quantitatively analyzed by Annexin V-FITC/PI double-labeled flow cytometry, the serum total homocysteine (tHCY) level was detected by ELISA and the expressions of eIF2a, p-eIF2a, GRP78/Bip, GRP94, caspase-3 and caspase-12 in liver were examined using Real-time PCR and Western blot.
RESULTSTypical acute liver injury and chronic liver injury were observed at week 4 and week 12 respectively. The hepatocyte apoptosis rates in 6-week model rats significantly increased compared with normal rats (P value less than 0.05), and the degree of hepatocyte apoptosis continued to increase with the modeling time, and the percentages of early and total apoptosis reached 26% and 29% at week 12. From week 6 to week 12, the serum tHCY levels in model rats were obviously higher than in normal rats (P value less than 0.01). Since week 4, eIF2a protein phosphorylation in model rat livers remarkably elevated compared with that in normal rat livers (P value less than 0.01), and at week 12 the peIF2a protein expression in model rat livers increased by 2.81-fold. Since week 4 the expressions of GRP78/Bip, GRP94, caspase-12 and caspase-3 mRNA and protein in model rat livers showed a significant increase as compared to normal rat livers, and at week 12, these gene and protein levels increased 4.70, 12.95, 3.83, 4.05 fold and 3.93, 6.93, 9.88, 3.31 fold, respectively (P value less than 0.01).
CONCLUSIONActivation of PERK/eIF2a signaling pathway contributes to the occurrence and development of hepatocyte apoptosis in alcoholic liver injury rats and it might be as a potential target for therapeutic applications in alcoholic liver diseases.
Animals ; Apoptosis ; Eukaryotic Initiation Factor-2 ; metabolism ; Hepatocytes ; cytology ; pathology ; Liver Diseases, Alcoholic ; metabolism ; pathology ; Male ; Rats ; Rats, Sprague-Dawley ; Signal Transduction ; eIF-2 Kinase ; metabolism
3.Role of PERK/eIF2α/CHOP Endoplasmic Reticulum Stress Pathway in Oxidized Low-density Lipoprotein Mediated Induction of Endothelial Apoptosis.
Yong Kang TAO ; Pu Lin YU ; Yong Ping BAI ; Sheng Tao YAN ; Shui Ping ZHAO ; Guo Qiang ZHANG
Biomedical and Environmental Sciences 2016;29(12):868-876
OBJECTIVEPERK/eIF2α/CHOP is a major signaling pathway mediating endoplasmic reticulum (ER) stress related with atherosclerosis. Oxidized LDL (ox-LDL) also induces endothelial apoptosis and plays a vital role in the initiation and progression of atherosclerosis. The present study was conducted to explore the regulatory effect of ox-LDL on PERK/eIF2α/CHOP signaling pathway in vascular endothelial cells.
METHODSThe effects of ox-LDL on PERK and p-eIF2α protein expression of primary human umbilical vein endothelial cells (HUVECs) were investigated by Western blot analysis. PERK gene silencing and selective eIF2α phosphatase inhibitor, salubrinal were used to inhibit the process of ox-LDL induced endothelial cell apoptosis, caspase-3 activity, and CHOP mRNA level.
RESULTSOx-LDL treatment significantly increased the expression of PERK, PERK-mediated inactivation of eIF2α phosphorylation, and the expression of CHOP, as well as the caspase-3 activity and apoptosis. The effects of ox-LDL were markedly decreased by knocking down PERK with stable transduction of lentiviral shRNA or by selective eIF2α phosphatase inhibitor, salubrinal.
CONCLUSIONThis study provides the first evidence that ox-LDL induces apoptosis in vascular endothelial cells mediated largely via the PERK/eIF2α/CHOP ER-stress pathway. It adds new insights into the molecular mechanisms underlying the pathogenesis and progression of atherosclerosis.
Apoptosis ; Endoplasmic Reticulum Stress ; Eukaryotic Initiation Factor-2 ; genetics ; metabolism ; Human Umbilical Vein Endothelial Cells ; metabolism ; Humans ; Lipoproteins, LDL ; genetics ; metabolism ; Signal Transduction ; Transcription Factor CHOP ; genetics ; metabolism ; eIF-2 Kinase ; genetics ; metabolism
4.Intrinsic Cellular Defenses against Virus Infection by Antiviral Type I Interferon.
Kyung Hyun BOO ; Joo Sung YANG
Yonsei Medical Journal 2010;51(1):9-17
Intrinsic cellular defenses are non-specific antiviral activities by recognizing pathogen-associated molecular patterns (PAMPs). Toll-like receptors (TLRs), one of the pathogen recognize receptor (PRR), sense various microbial ligands. Especially, TLR2, TLR3, TLR4, TLR7, TLR8 and TLR9 recognize viral ligands such as glycoprotein, single- or double-stranded RNA and CpG nucleotides. The binding of viral ligands to TLRs transmits its signal to Toll/interleukin-1 receptor (TIR) to activate transcription factors via signal transduction pathway. Through activation of transcription factors, such as interferon regulatory factor-3, 5, and 7 (IRF-3, 5, 7) or nuclear factor-kappaB (NF-kappaB), type I interferons are induced, and antiviral proteins such as myxovirus-resistance protein (Mx) GTPase, RNA-dependent Protein Kinase (PKR), ribonuclease L (RNase L), Oligo-adenylate Synthetase (OAS) and Interferon Stimulated Gene (ISG) are further expressed. These antiviral proteins play an important role of antiviral resistancy against several viral pathogens in infected cells and further activate innate immune responses.
Animals
;
GTP-Binding Proteins/metabolism
;
Humans
;
Interferon Regulatory Factors/metabolism
;
Interferon Type I/*metabolism/physiology
;
Models, Biological
;
NF-kappa B/metabolism
;
Toll-Like Receptors/metabolism
;
Virus Diseases/*immunology/*metabolism/virology
;
eIF-2 Kinase/metabolism
5.Coxsackievirus B3 Infection Triggers Autophagy through 3 Pathways of Endoplasmic Reticulum Stress.
Xiao Nuan LUO ; Hai Lan YAO ; Juan SONG ; Qin Qin SONG ; Bing Tian SHI ; Dong XIA ; Jun HAN
Biomedical and Environmental Sciences 2018;31(12):867-875
OBJECTIVE:
Autophagy is a highly conserved intracellular degradation pathway. Many picornaviruses induce autophagy to benefit viral replication, but an understanding of how autophagy occurs remains incomplete. In this study, we explored whether coxsackievirus B3 (CVB3) infection induced autophagy through endoplasmic reticulum (ER) stress.
METHODS:
In CVB3-infected HeLa cells, the specific molecules of ER stress and autophagy were detected using Western blotting, reverse transcription polymerase chain reaction (RT-PCR), and confocal microscopy. Then PKR-like ER protein kinase (PERK) inhibitor, inositol-requiring protein-1 (IRE1) inhibitor, or activating transcription factor-6 (ATF6) inhibitor worked on CVB3-infected cells, their effect on autophagy was assessed by Western blotting for detecting microtubule-associated protein light chain 3 (LC3).
RESULTS:
CVB3 infection induced ER stress, and ER stress sensors PERK/eIF2α, IRE1/XBP1, and ATF6 were activated. CVB3 infection increased the accumulation of green fluorescent protein (GFP)-LC3 punctuation and induced the conversion from LC3-I to phosphatidylethanolamine-conjugated LC3-1 (LC3-II). CVB3 infection still decreased the expression of mammalian target of rapamycin (mTOR) and p-mTOR. Inhibition of PERK, IRE1, or ATF6 significantly decreased the ratio of LC3-II to LC3-I in CVB3-infected HeLa cells.
CONCLUSION
CVB3 infection induced autophagy through ER stress in HeLa cells, and PERK, IRE1, and ATF6a pathways participated in the regulation of autophagy. Our data suggested that ER stress may inhibit mTOR signaling pathway to induce autophagy during CVB3 infection.
Activating Transcription Factor 6
;
metabolism
;
Autophagy
;
Coxsackievirus Infections
;
metabolism
;
Endoplasmic Reticulum Stress
;
Endoribonucleases
;
metabolism
;
Enterovirus B, Human
;
HeLa Cells
;
Humans
;
Protein-Serine-Threonine Kinases
;
metabolism
;
Signal Transduction
;
eIF-2 Kinase
;
metabolism
6.Low-dose radiation induces endoplasmic reticulum stress and activates PERK-CHOP signaling pathway in mouse testicular cells.
Fang FANG ; Ping-Sheng GONG ; Xiang-Fu SONG ; Shou-Liang GONG ; Zhi-Cheng WANG
National Journal of Andrology 2012;18(9):777-782
OBJECTIVETo explore the correlation of low-dose radiation with endoplasmic reticulum stress and the activation of the PERK-CHOP signaling pathway in mouse testicular cells.
METHODSHealthy Kunming mice were randomly assigned to time-effect (0, 3, 6, 12 and 24 h of irradiation at 75 mGy) and dose-effect (12 h of irradiation at 0, 50, 75, 100 and 200 mGy) groups. The contents of H202 and MDA were measured by colorimetry with the agent kits, the expressions of GRP78, PERK and CHOP mRNA detected by quantitative RT-PCR, and the levels of GRP7B, PERK, phosphorylated PERK (pho-PERK) and CHOP proteins determined by Western blotting and image analysis.
RESULTSAfter whole-body irradiation of the mice with 75 mGy, the content of H2 02 in the testis tissue was increased with time prolongation, while that of MDA decreased slightly at 3 and 6 h and then increased with the lengthening of time, both increased significantly at 12 and 24 h as compared with those at 0 h (P < 0. 05, P < 0. 01). Apart from reduced levels of GRP78 mRNA at 3 and 24 h and GRP78 protein at 6 h after irradiation, significant increases were found in the mRNA expressions of GRP78 at 12 h, PERK at 3,6, 12 and 24 hand CHOP at 12 and 24 h (P < 0.05, P < 0.01), as well as in the protein levels of GRP78 at 12 and 24 h, pho-PERK at 3, 12 and 24 h and CHOP at 3, 6, 12 and 24 h in comparison with those at 0 h (P < 0. 05, P < 0. 01). No obvious regularity was observed in the change of the PERK protein expression. After 12 h of whole-body irradiation, the content of H202 was increased at 50, 75 and 100 mGy, but decreased slightly at 200 mGy, while that of MDA was increased with dose increasing, with significant increases in the content of H2 02 at 75 and 100 mCy and in that of MDA at 75, 100 and 200 mGy as compared with the 0 mGy group. Apart from the reduced levels of GRP78 mRNA at 50 and 200 mCy, significant increases were found in the mRNA expressions of PERK at 75, 100 and 200 mGy and CHOP at 50, 75, 100 and 200 (P c 0. 05, P < 0.01) as well as in the protein levels of GRP78 at 100 and 200 mGy, pho-PERK at 50, 100 and 200 mGy and CHOP at 50, 75, 100 and 200 mCy as compared with those at 0 mGy (P < 0. 05, P < 0. 01). There were differences in the changes of different protein expressions, but no obvious regularity was seen in the change of the PERK protein expression.
CONCLUSIONLow-dose radiation can induce endoplasmic reticulum stress in mouse testicular cells, and activate the PERK-CHOP signaling pathway.
Animals ; Endoplasmic Reticulum Stress ; radiation effects ; Heat-Shock Proteins ; metabolism ; Male ; Mice ; Mice, Inbred Strains ; Radiation Dosage ; Radiation, Ionizing ; Signal Transduction ; radiation effects ; Testis ; cytology ; metabolism ; radiation effects ; Transcription Factor CHOP ; metabolism ; Whole-Body Irradiation ; eIF-2 Kinase ; metabolism
7.Interaction of Hsp40 with influenza virus M2 protein: implications for PKR signaling pathway.
Zhenhong GUAN ; Di LIU ; Shuofu MI ; Jie ZHANG ; Qinong YE ; Ming WANG ; George F GAO ; Jinghua YAN
Protein & Cell 2010;1(10):944-955
Influenza virus contains three integral membrane proteins: haemagglutinin, neuraminidase, and matrix protein (M1 and M2). Among them, M2 protein functions as an ion channel, important for virus uncoating in endosomes of virus-infected cells and essential for virus replication. In an effort to explore potential new functions of M2 in the virus life cycle, we used yeast two-hybrid system to search for M2-associated cellular proteins. One of the positive clones was identified as human Hsp40/Hdj1, a DnaJ/Hsp40 family protein. Here, we report that both BM2 (M2 of influenza B virus) and A/M2 (M2 of influenza A virus) interacted with Hsp40 in vitro and in vivo. The region of M2-Hsp40 interaction has been mapped to the CTD1 domain of Hsp40. Hsp40 has been reported to be a regulator of PKR signaling pathway by interacting with p58(IPK) that is a cellular inhibitor of PKR. PKR is a crucial component of the host defense response against virus infection. We therefore attempted to understand the relationship among M2, Hsp40 and p58(IPK) by further experimentation. The results demonstrated that both A/M2 and BM2 are able to bind to p58(IPK) in vitro and in vivo and enhance PKR autophosphorylation probably via forming a stable complex with Hsp40 and P58(IPK), and consequently induce cell death. These results suggest that influenza virus M2 protein is involved in p58(IPK) mediated PKR regulation during influenza virus infection, therefore affecting infected-cell life cycle and virus replication.
HSP40 Heat-Shock Proteins
;
genetics
;
metabolism
;
Humans
;
Orthomyxoviridae
;
genetics
;
metabolism
;
Phosphorylation
;
Protein Binding
;
genetics
;
Signal Transduction
;
genetics
;
Two-Hybrid System Techniques
;
Viral Matrix Proteins
;
metabolism
;
Virus Replication
;
genetics
;
Virus Uncoating
;
eIF-2 Kinase
;
metabolism
8.Human Leptin Protein Induces Proliferation of A549 Cells via Inhibition of PKR-Like ER Kinase and Activating Transcription Factor-6 Mediated Apoptosis.
Yonsei Medical Journal 2013;54(6):1407-1415
PURPOSE: To investigate the anti-apoptotic mechanism of leptin in non-small cell lung cancer. MATERIALS AND METHODS: The influences of leptin on apoptosis were investigated, analyzing the mechanism that triggers growth of A549 cells. The effects of leptin on cell proliferation were examined by XTT analysis. Leptin, C/EBP homologous protein (CHOP), phosphorylated-PKR-like ER kinase (p-Perk), inositol requiring proteins-1, spliced X-box transcription factor-1 (XBP1), cleaved activating transcription factor-6 (ATF6), eukaryotic translation initiation factor-2alpha, caspase-12 and CHOP protein were detected in four groups by western blot, and endoplasmic reticulum (ER) stress related mRNA were detected by reverse transcription PCR. RESULTS: The expression of leptin in A549 and leptin transfected cells inhibited cisplatin activated ER stress-associated mRNA transcription and protein activation. Two ER stress unfolded protein response pathways, PERK and ATF6, were involved, and XBP1 and tumor necrosis factor receptor-associated factor 2 (TRAF2) were increased significantly when treated with cisplatin in A549-siRNA against leptin cells. Furthermore, CHOP expression was inhibited upon leptin expression in A549, LPT-PeP and LPT-EX cells. CONCLUSION: Leptin serves as an important factor that promotes the growth of A549 cells through blocking ER stress-mediated pathways. This blocking is triggered by p-Perk and ATF6 via inhibition of CHOP expression.
Activating Transcription Factor 6/genetics/*metabolism
;
Apoptosis/*drug effects/genetics
;
Blotting, Western
;
Cell Line, Tumor
;
Cell Proliferation/*drug effects
;
Humans
;
Leptin/*pharmacology
;
Reverse Transcriptase Polymerase Chain Reaction
;
eIF-2 Kinase/*metabolism
9.5-Hydroxymethylfurfural protects against ER stress-induced apoptosis in GalN/TNF-α-injured L02 hepatocytes through regulating the PERK-eIF2α signaling pathway.
Ze-Qun JIANG ; Yan-Xia MA ; Mu-Han LI ; Xiu-Qin ZHAN ; Xu ZHANG ; Ming-Yan WANG
Chinese Journal of Natural Medicines (English Ed.) 2015;13(12):896-905
5-Hydroxymethylfurfural (5-HMF), a water-soluble compound extracted from wine-processed Fructus corni, is a novel hepatic protectant for treating acute liver injury. The present study was designed to investigate the protective effect of 5-HMF in human L02 hepatocytes injured by D-galactosamine (GalN) and tumor necrosis factor-α (TNF-α) in vitro and to explore the underlying mechanisms of action. Our results showed that 5-HMF caused significant increase in the viability of L02 cells injured by GalN/TNF-α, in accordance with a dose-dependent decrease in apoptotic cell death confirmed by morphological and flow cytometric analyses. Based on immunofluorescence and Western blot assays, we found that GalN/TNF-α induced ER stress in the cells, as indicated by the disturbance of intracellular Ca(2+) concentration, the activation of protein kinase RNA (PKR)-like ER kinase (PERK), phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α), and expression of ATF4 and CHOP proteins, which was reversed by 5-HMF pre-treatment in a dose-dependent manner. The anti-apoptotic effect of 5-HMF was further evidenced by balancing the expression of Bcl-2 family members. In addition, the knockdown of PERK suppressed the expression of phospho-PERK, phospho-eIF2α, ATF4, and CHOP, resulting in a significant decrease in cell apoptosis after the treatment with GalN/TNF-α. 5-HMF could enhance the effects of PERK knockdown, protecting the cells against the GalN/TNF-α insult. In conclusion, these findings demonstrate that 5-HMF can effectively protect GalN/TNF-α-injured L02 hepatocytes against ER stress-induced apoptosis through the regulation of the PERK-eIF2α signaling pathway, suggesting that it is a possible candidate for liver disease therapy.
Apoptosis
;
drug effects
;
Cornus
;
chemistry
;
Endoplasmic Reticulum Stress
;
drug effects
;
Eukaryotic Initiation Factor-2
;
genetics
;
metabolism
;
Furaldehyde
;
analogs & derivatives
;
pharmacology
;
Galactosamine
;
metabolism
;
Hepatocytes
;
cytology
;
drug effects
;
metabolism
;
Humans
;
Liver
;
cytology
;
drug effects
;
metabolism
;
Plant Extracts
;
pharmacology
;
Protective Agents
;
pharmacology
;
Signal Transduction
;
drug effects
;
Tumor Necrosis Factor-alpha
;
genetics
;
metabolism
;
eIF-2 Kinase
;
genetics
;
metabolism
10.Homocysteine activates T cells by enhancing endoplasmic reticulum-mitochondria coupling and increasing mitochondrial respiration.
Juan FENG ; Silin LÜ ; Yanhong DING ; Ming ZHENG ; Xian WANG
Protein & Cell 2016;7(6):391-402
Hyperhomocysteinemia (HHcy) accelerates atherosclerosis by increasing proliferation and stimulating cytokine secretion in T cells. However, whether homocysteine (Hcy)-mediated T cell activation is associated with metabolic reprogramming is unclear. Here, our in vivo and in vitro studies showed that Hcy-stimulated splenic T-cell activation in mice was accompanied by increased levels of mitochondrial reactive oxygen species (ROS) and calcium, mitochondrial mass and respiration. Inhibiting mitochondrial ROS production and calcium signals or blocking mitochondrial respiration largely blunted Hcy-induced T-cell interferon γ (IFN-γ) secretion and proliferation. Hcy also enhanced endoplasmic reticulum (ER) stress in T cells, and inhibition of ER stress with 4-phenylbutyric acid blocked Hcy-induced T-cell activation. Mechanistically, Hcy increased ER-mitochondria coupling, and uncoupling ER-mitochondria by the microtubule inhibitor nocodazole attenuated Hcy-stimulated mitochondrial reprogramming, IFN-γ secretion and proliferation in T cells, suggesting that juxtaposition of ER and mitochondria is required for Hcy-promoted mitochondrial function and T-cell activation. In conclusion, Hcy promotes T-cell activation by increasing ER-mitochondria coupling and regulating metabolic reprogramming.
Animals
;
Calcium
;
metabolism
;
Cell Proliferation
;
drug effects
;
Cells, Cultured
;
Endoplasmic Reticulum
;
metabolism
;
Endoplasmic Reticulum Stress
;
drug effects
;
Endoribonucleases
;
metabolism
;
Female
;
Homocysteine
;
toxicity
;
Interferon-gamma
;
analysis
;
Metabolic Engineering
;
Mice
;
Mice, Inbred C57BL
;
Mitochondria
;
drug effects
;
metabolism
;
Nocodazole
;
pharmacology
;
Phenylbutyrates
;
pharmacology
;
Protein-Serine-Threonine Kinases
;
metabolism
;
Proto-Oncogene Proteins c-akt
;
metabolism
;
Reactive Oxygen Species
;
metabolism
;
T-Lymphocytes
;
cytology
;
drug effects
;
metabolism
;
eIF-2 Kinase
;
metabolism