Down regulation of DJ-1 enhances cell death by oxidative stress, ER stress, and proteasome inhibition

2003 ◽  
Vol 312 (4) ◽  
pp. 1342-1348 ◽  
Author(s):  
Takanori Yokota ◽  
Kanako Sugawara ◽  
Kaoru Ito ◽  
Ryosuke Takahashi ◽  
Hiroyoshi Ariga ◽  
...  

2006 ◽  
Vol 26 (8) ◽  
pp. 3071-3084 ◽  
Author(s):  
Ping Hu ◽  
Zhang Han ◽  
Anthony D. Couvillon ◽  
Randal J. Kaufman ◽  
John H. Exton

ABSTRACT NF-κB is critical for determining cellular sensitivity to apoptotic stimuli by regulating both mitochondrial and death receptor apoptotic pathways. The endoplasmic reticulum (ER) emerges as a new apoptotic signaling initiator. However, the mechanism by which ER stress activates NF-κB and its role in regulation of ER stress-induced cell death are largely unclear. Here, we report that, in response to ER stress, IKK forms a complex with IRE1α through the adapter protein TRAF2. ER stress-induced NF-κB activation is impaired in IRE1α knockdown cells and IRE1α−/− MEFs. We found, however, that inhibiting NF-κB significantly decreased ER stress-induced cell death in a caspase-8-dependent manner. Gene expression analysis revealed that ER stress-induced expression of tumor necrosis factor alpha (TNF-α) was IRE1α and NF-κB dependent. Blocking TNF receptor 1 signaling significantly inhibited ER stress-induced cell death. Further studies suggest that ER stress induces down-regulation of TRAF2 expression, which impairs TNF-α-induced activation of NF-κB and c-Jun N-terminal kinase and turns TNF-α from a weak to a powerful apoptosis inducer. Thus, ER stress induces two signals, namely TNF-α induction and TRAF2 down-regulation. They work in concert to amplify ER-initiated apoptotic signaling through the membrane death receptor.



2000 ◽  
Vol 20 (10) ◽  
pp. 1467-1473 ◽  
Author(s):  
Jeffrey N. Keller ◽  
Feng F. Huang ◽  
Hong Zhu ◽  
Jin Yu ◽  
Ye-Shih Ho ◽  
...  

Numerous studies indicate a role for oxidative stress in the neuronal degeneration and cell death that occur during ischemia–reperfusion injury. Recent data suggest that inhibition of the proteasome may be a means by which oxidative stress mediates neuronal cell death. In the current study, the authors demonstrate that there is a time-dependent decrease in proteasome activity, which is not associated with decreased expression of proteasome subunits, after cerebral ischemia–reperfusion injury. To determine the role of oxidative stress in mediating proteasome inhibition, ischemia–reperfusion studies were conducted in mice that either overexpressed the antioxidant enzyme glutathione peroxidase [GPX 1(+)], or were devoid of glutathione peroxidase activity (GPX −/−). After ischemia–reperfusion, GPX 1(+) mice displayed decreased infarct size, attenuated neurologic impairment, and reduced levels of proteasome inhibition compared with either GPX −/− or wild type mice. In addition, GPX 1(+) mice displayed lower levels of 4-hydroxynonenal-modified proteasome subunits after ischemia–reperfusion injury. Together, these data indicate that proteasome inhibition occurs during cerebral ischemia–reperfusion injury and is mediated, at least in part, by oxidative stress.



2014 ◽  
Vol 4 (1) ◽  
Author(s):  
Sunita Maharjan ◽  
Masahide Oku ◽  
Masashi Tsuda ◽  
Jun Hoseki ◽  
Yasuyoshi Sakai


2010 ◽  
Vol 426 (1) ◽  
pp. 43-53 ◽  
Author(s):  
Craig W. Younce ◽  
Pappachan E. Kolattukudy

MCP-1 (monocyte chemotactic protein-1) plays a critical role in the development of heart failure that is known to involve apoptosis. How MCP-1 contributes to cell death involved in the development of heart disease is not understood. In the present study we show that MCP-1 causes death in cardiac myoblasts, H9c2 cells, by inducing oxidative stress which causes ER stress leading to autophagy via a novel zinc-finger protein, MCPIP (MCP-1-induced protein). MCPIP expression caused cell death, and knockdown of MCPIP attenuated MCP-1induced cell death. It caused induction of iNOS (inducible NO synthase), translocation of the NADPH oxidase subunit phox47 from the cytoplasm to the membrane, production of ROS (reactive oxygen species), and induction of ER (endoplasmic reticulum) stress markers HSP40 (heat-shock protein 40), PDI (protein disulfide-isomerase), GRP78 (guanine-nucleotide-releasing protein 78) and IRE1α (inositol-requiring enzyme 1α). It also caused autophagy, as indicated by beclin-1 induction, cleavage of LC3 (microtubule-associated protein 1 light chain 3) and autophagolysosome formation, and apoptosis, as indicated by caspase 3 activation and TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling) assay. Inhibitors of oxidative stress, including CeO2 nanoparticles, inhibited ROS formation, ER stress, autophagy and cell death. Specific inhibitors of ER stress inhibited autophagy and cell death as did knockdown of the ER stress signalling protein IRE1. Knockdown of beclin-1 and autophagy inhibitors prevented cell death. This cell death involved caspase 2 and caspase 12, as specific inhibitors of these caspases prevented MCPIP-induced cell death. Microarray analysis showed that MCPIP expression caused induction of a variety of genes known to be involved in cell death. MCPIP caused activation of JNK (c-Jun N-terminal kinase) and p38 and induction of p53 and PUMA (p53 up-regulated modulator of apoptosis). Taken together, these results suggest that MCPIP induces ROS/RNS (reactive nitrogen species) production that causes ER stress which leads to autophagy and apoptosis through caspase 2/12 and IRE1α–JNK/p38–p53–PUMA pathway. These results provide the first molecular insights into the mechanism by which elevated MCP-1 levels associated with chronic inflammation may contribute to the development of heart failure.



2013 ◽  
Vol 304 (7) ◽  
pp. C636-C655 ◽  
Author(s):  
Bhavana Chhunchha ◽  
Nigar Fatma ◽  
Eri Kubo ◽  
Prerana Rai ◽  
Sanjay P. Singh ◽  
...  

Oxidative stress and endoplasmic reticulum (ER) stress are emerging as crucial events in the etiopathology of many neurodegenerative diseases. While the neuroprotective contributions of the dietary compound curcumin has been recognized, the molecular mechanisms underlying curcumin's neuroprotection under oxidative and ER stresses remains elusive. Herein, we show that curcumin protects HT22 from oxidative and ER stresses evoked by the hypoxia (1% O2 or CoCl2 treatment) by enhancing peroxiredoxin 6 (Prdx6) expression. Cells exposed to CoCl2 displayed reduced expression of Prdx6 with higher reactive oxygen species (ROS) expression and activation of NF-κB with IκB phosphorylation. When NF-κB activity was blocked by using SN50, an inhibitor of NF-κB, or cells treated with curcumin, the repression of Prdx6 expression was restored, suggesting the involvement of NF-κB in modulating Prdx6 expression. These cells were enriched with an accumulation of ER stress proteins, C/EBP homologous protein (CHOP), GRP/78, and calreticulin, and had activated states of caspases 12, 9, and 3. Reinforced expression of Prdx6 in HT22 cells by curcumin reestablished survival signaling by reducing propagation of ROS and blunting ER stress signaling. Intriguingly, knockdown of Prdx6 by antisense revealed that loss of Prdx6 contributed to cell death by sustaining enhanced levels of ER stress-responsive proapoptotic proteins, which was due to elevated ROS production, suggesting that Prdx6 deficiency is a cause of initiation of ROS-mediated ER stress-induced apoptosis. We propose that using curcumin to reinforce the naturally occurring Prdx6 expression and attenuate ROS-based ER stress and NF-κB-mediated aberrant signaling improves cell survival and may provide an avenue to treat and/or postpone diseases associated with ROS or ER stress.



Antioxidants ◽  
2021 ◽  
Vol 10 (10) ◽  
pp. 1503
Author(s):  
Hae-Ahm Lee ◽  
Ki-Back Chu ◽  
Eun-Kyung Moon ◽  
Fu-Shi Quan

Histone deacetylase inhibitors (HDACi) are emerging as anti-hepatocellular carcinoma (HCC) agents. However, the molecular mechanisms underlying HDACi-induced sensitization to oxidative stress and cell death of HCC remain elusive. We hypothesized that HDACi reduces the anti-oxidative stress capacity of HCC, rendering it more susceptible to oxidative stress and cell death. Change in the transcriptome of HCC was analyzed by RNA-seq and validated using real-time quantitative polymerase chain reaction (qPCR) and Western blot. Cell death of HCC was analyzed by fluorescence-activated cell sorting (FACS). Protein localization and binding on the target gene promoters were investigated by immunofluorescence (IF) and chromatin immunoprecipitation (ChIP), respectively. Glutathione peroxidase 8 (GPX8) was highly down-regulated in HCC upon oxidative stress and HDACi co-treatment. Oxidative stress and HDACi enhanced the expression and transcriptional activities of ER-stress-related genes. N-acetyl-cysteine (NAC) supplementation reversed the oxidative stress and HDACi-induced apoptosis in HCC. HDACi significantly enhanced the effect of ER stressors on HCC cell death. GPX8 overexpression reversed the activation of ER stress signaling and apoptosis induced by oxidative stress and HDACi. In conclusion, HDACi suppresses the expression of GPX8, which sensitizes HCC to ER stress and apoptosis by oxidative stress.



Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5800-5800
Author(s):  
Yin-Chen Chou ◽  
Chia-Wei Chen ◽  
Yuan-Yeh Kuo ◽  
Liang-In Lin ◽  
Chung-Yi Hu

Abstract Introduction: Acute lymphoblastic leukemias (ALLs) harboring t(9;22)(Ph+-ALL) are very high risk (VHR) ALL displaying poor clinical outcome irrespective of intensive chemotherapies plus tyrosine kinase inhibitor (TKI) treatment. HQ17(3)[10'(Z),13'(E),15'(E)-heptadecatrienyl hydroquinone] isolated from sap of the lacquer tree showed rapid (within 24hrs) and potent cytotoxic effect at micromolar concentration on several ALL cell lines, including Imatinib-refractory Ph+-ALL SUP-B15 cells, but spared normal PB leukocytes, and showed nontoxic in experimental rats after 28-day injection. Therefore HQ17(3) presents as a potential anti-leukemic agents and provide a platform for exploring anti-leukemic adjuvants. Our previous study showed HQ17(3)-induced rapid cell demise, characterized by oxidative stress, mitochondrial membrane potential disturbance, loss of membrane integrity, and nuclear DNA fragmentation. HQ17(3)-induced cell death is a caspase-independent program, and is different from the RIP1-mediated controlled necroptosis since both pan-caspase inhibitor and RIP-1 inhihitor failed to protect SUP-B15 cells from death. The ER stress markers (chaperon Grp78 and phosphorylated-eIF2α) were up-regulated as early as 5hrs after HQ17(3) treatment. Here we aim to illustrate the characters of the HQ17(3)-induced non-classical death on Ph+-SUP-B15 cells, focus on ER stress-associated mitochondrial Ca2+ homeostasis. Methods: Cell death and changes of mitochondria in response to HQ17(3) w/wo inhibitors were analyzed. Cells were stained by Annexin V/PI and analyzed by flow cytometry for cell death. Mitochondria mass, mitochondrial Ca2+ accumulation was detected by fluorescent Mitotracker Green and Rhod-2 probes, respectively. Mitochondrial superoxide was measured by Mitosox stain. Western blot analysis was used to analyze MFN1/2, OPA1 (mitochondrial markers). Nuclear accumulation of apoptosis inducing factor (AIF), co-localization of mitochondrial COX-IV and LC3-II (mitophagy) were revealed by immunofluorescence stain and confocal microscopy. Results: We showed mitochondrial Ca2+ accumulation at the early time when ER stress occurred (Fig 1), accompanied by mitochondrial superoxide elevation, followed by loss of mitochondrial membrane potential (MMP) and nuclear translocation of apoptosis-inducing factor (AIF). HQ17(3) treatment lead to decreased mitochondrial proteins MFN1/2 and OPA1, while Mitotracker Green stain showed significant loss of mitochondrial mass preceded cell death, indicating damaged mitochondria underwent fission followed by mitophagy. Immunofluorescence stain showed evidence of mitophagy (COX IV and LC3B co-localization). Calpain-1 inhibitor PD150606 blocked AIF nuclear translocation but only slightly reduced the HQ17(3)-induced cell death (Fig 2). Further, Ca2+ chelator Bapta-AM prevented mitochondrial superoxide production, MMP loss, mitophagy (Fig 3), and rescued cell death (Fig 1) more effectively. Conclusion: In Ph+-ALL SUP-B15 cells, HQ17(3) induce ER stress by yet-defined mechanism, this mobilizes Ca2+ to mitochondria and acts in multi-facet: a) results in AIF cleavage and translocation to mediate nuclear chromatin fragmentation, b) Ca2+-overload leads to oxidative stress and perturbs mitochondria integrity, c) damaged mitochondria trigger extensive mitophagy and cell death ensues. Therefore, agents that help elicit similar intricate effector network associated with ER/mitochondria stress will have potential to be adjuvants in aiding control of the Ph+ VHR-ALL cells refractory to conventional chemotherapies and TKI regime. Disclosures No relevant conflicts of interest to declare.



2014 ◽  
Vol 25 (9) ◽  
pp. 1411-1420 ◽  
Author(s):  
Nobuhiko Hiramatsu ◽  
Carissa Messah ◽  
Jaeseok Han ◽  
Matthew M. LaVail ◽  
Randal J. Kaufman ◽  
...  

Endoplasmic reticulum (ER) protein misfolding activates the unfolded protein response (UPR) to help cells cope with ER stress. If ER homeostasis is not restored, UPR promotes cell death. The mechanisms of UPR-mediated cell death are poorly understood. The PKR-like endoplasmic reticulum kinase (PERK) arm of the UPR is implicated in ER stress–induced cell death, in part through up-regulation of proapoptotic CCAAT/enhancer binding protein homologous protein (CHOP). Chop−/− cells are partially resistant to ER stress–induced cell death, and CHOP overexpression alone does not induce cell death. These findings suggest that additional mechanisms regulate cell death downstream of PERK. Here we find dramatic suppression of antiapoptosis XIAP proteins in response to chronic ER stress. We find that PERK down-regulates XIAP synthesis through eIF2α and promotes XIAP degradation through ATF4. Of interest, PERK's down-regulation of XIAP occurs independently of CHOP activity. Loss of XIAP leads to increased cell death, whereas XIAP overexpression significantly enhances resistance to ER stress–induced cell death, even in the absence of CHOP. Our findings define a novel signaling circuit between PERK and XIAP that operates in parallel with PERK to CHOP induction to influence cell survival during ER stress. We propose a “two-hit” model of ER stress–induced cell death involving concomitant CHOP up-regulation and XIAP down-regulation both induced by PERK.



2010 ◽  
Vol 79 (9) ◽  
pp. 1221-1230 ◽  
Author(s):  
Nicolas Dejeans ◽  
Nicolas Tajeddine ◽  
Raphaël Beck ◽  
Julien Verrax ◽  
Henryk Taper ◽  
...  


Sign in / Sign up

Export Citation Format

Share Document