scholarly journals The TSC1 and TSC2 tumor suppressors are required for proper ER stress response and protect cells from ER stress-induced apoptosis

2010 ◽  
Vol 18 (1) ◽  
pp. 133-144 ◽  
Author(s):  
Y J Kang ◽  
M-K Lu ◽  
K-L Guan
2009 ◽  
Vol 32 (2) ◽  
pp. 251-257 ◽  
Author(s):  
Do-Sung Kim ◽  
Seul-Ki Jeong ◽  
Hyung-Ryong Kim ◽  
Dal-Sik Kim ◽  
Soo-Wan Chae ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2856-2856 ◽  
Author(s):  
Rekha Rao ◽  
Warren Fiskus ◽  
Ramesh Balusu ◽  
Hongwei Ma ◽  
James Bradner ◽  
...  

Abstract Abstract 2856 The proteasome inhibitor bortezpmib has been shown to markedly increase the intracellular levels of misfolded proteins, induce aggresome formation and cause endoplasmic reticulum (ER) stress, resulting in apoptosis of human Mantle Cell Lymphoma (MCL) cells. Consistent with this, Bortezomib displays clinical efficacy in patients with relapsed and refractory MCL. We have recently reported that the pan-histone deacetylase (HDAC) inhibitor panobinostat, by also inhibiting HDAC6, abrogates aggresome formation and induces Endoplasmic Stress (ER) stress, as well as potentiates bortezomib-induced apoptosis of MCL cells. Here, we determined the anti-MCL cell activity of an HDAC6-specific inhibitor, WT-161 alone and in combination with the novel, orally bio-available, proteasome inhibitor carfilzomib (Proteolix Inc.) against human, cultured and primary, patient-derived MCL cells. Treatment with WT-161 (0.1 to 1.0 uM) resulted in a dose-dependent increase in the acetylation of alpha-tubulin and heat shock protein (hsp) 90, without any appreciable increase in the levels of acetylated histone (H) 3. Consistent with WT-161 mediated hyperacetylation and inhibition of hsp90 chaperone function, treatment with WT-161 increased the intracellular levels of polyubiuitylated proteins in the cultured MCL JeKo-1 and Z138 cells. WT-161 was also noted to dose-dependently deplete the levels of cyclin D1 in the cultured MCL cells. Treatment with WT-161 also induced ER stress response in the MCL cells, demonstrated by increase in the protein levels of Glucose regulated protein (GRP) 78, phosphorylated eIF2 (eukaryotic initation factor 2) α, and induction of the pro-apoptotic transcription factor CHOP (CAAT/Enhancer Binding Protein Homologous Protein). We next determined the effects of co-treatment with WT-161 on carfilzomib-induced aggresome formation, ER stress response and apoptosis of the cultured and primary MCL cells. Co-treatment with WT-161 (0.25 uM) abrogated carfilzomib-induced aggresome formation in MCL cells, as evidenced by confocal immunofluorescent staining of aggresomes with anti-HDAC6 and anti-ubiquitin antibodies. Compared to each agent alone, co-treatment with WT-161 and carfilzomib induced more intracellular polyubiquitylated proteins and induced higher levels of CHOP in the cultured MCL cells. Co-treatment with WT-161 and carfilzomib also synergistically induced apoptosis of the cultured MCL cells (combination indices < 1.0). Notably, co-treatment with WT-161 and carfilzomib also synergistically induced apoptosis of primary MCL cells (combination indices < 1.0). These findings strongly support the in vivo testing of the combination of an HDAC6-specific inhibitor such as WT-161 with the proteasome inhibitor carfilzomib against human MCL cells. Disclosures: No relevant conflicts of interest to declare.


Antioxidants ◽  
2021 ◽  
Vol 10 (9) ◽  
pp. 1492
Author(s):  
Ilandarage Menu Neelaka Molagoda ◽  
Mirissa Hewage Dumindu Kavinda ◽  
Yung Hyun Choi ◽  
Hyesook Lee ◽  
Chang-Hee Kang ◽  
...  

Fine particulate matter (PM2.5) originates from the combustion of coal and is found in the exhaust of fumes of diesel vehicles. PM2.5 readily penetrates the skin via the aryl hydrocarbon receptor, causing skin senescence, inflammatory skin diseases, DNA damage, and carcinogenesis. In this study, we investigated whether fisetin, a bioactive flavonoid, prevents PM2.5-induced apoptosis in HaCaT human keratinocytes. The results demonstrated that fisetin significantly downregulated PM2.5-induced apoptosis at concentrations below 10 μM. Fisetin strongly inhibited the production of reactive oxygen species (ROS) and the expression of pro-apoptotic proteins. The PM2.5-induced apoptosis was associated with the induction of the endoplasmic reticulum (ER) stress response, mediated via the protein kinase R-like ER kinase (PERK)–eukaryotic initiation factor 2α (eIF2α)–activating transcription factor 4 (ATF4)–CCAAT-enhancer-binding protein (C/EBP) homologous protein (CHOP) axis. Additionally, the cytosolic Ca2+ levels were markedly increased following exposure to PM2.5. However, fisetin inhibited the expression of ER stress-related proteins, including 78 kDa glucose-regulated protein (GRP78), phospho-eIF2α, ATF4, and CHOP, and reduced the cytosolic Ca2+ levels. These data suggest that fisetin inhibits PM2.5-induced apoptosis by inhibiting the ER stress response and production of ROS.


2019 ◽  
Author(s):  
Rebecca A.S. Palu ◽  
Clement Y. Chow

ABSTRACTEndoplasmic reticulum (ER) stress-induced apoptosis is a primary cause and modifier of degeneration in a number of genetic disorders. Understanding how genetic variation between individuals influences the ER stress response and subsequent activation of apoptosis could improve individualized therapies and predictions of outcomes for patients. In this study, we find that the uncharacterized, membrane-bound metallopeptidase CG14516 in Drosophila melanogaster, which we rename as SUPpressor of ER stress-induced DEATH (superdeath), plays a role in modifying ER stress-induced apoptosis. We demonstrate that loss of superdeath reduces apoptosis and degeneration in the Rh1G69D model of ER stress through the JNK signaling cascade. This effect on apoptosis occurs without altering the activation of the unfolded protein response (IRE1 and PERK), suggesting that the beneficial pro-survival effects of this response are intact. Furthermore, we show that superdeath functions epistatically upstream of CDK5, a known JNK-activated pro-apoptotic factor in this model of ER stress. We demonstrate that superdeath is not only a modifier of this particular model, but functions as a general modifier of ER stress-induced apoptosis across different tissues and ER stresses. Finally, we present evidence of Superdeath localization to the endoplasmic reticulum membrane. While similar in sequence to a number of human metallopeptidases found in the plasma membrane and ER membrane, its localization suggests that superdeath is orthologous to ERAP1/2 in humans. Together, this study provides evidence that superdeath is a link between stress in the ER and activation of cytosolic apoptotic pathways.SIGNIFICANCE STATEMENTGenetic diseases display a great deal of variability in presentation, progression, and overall outcomes. Much of this variability is caused by differences in genetic background among patients. One process that commonly modifies degenerative disease is the endoplasmic reticulum (ER) stress response. Understanding the genetic sources of variation in the ER stress response could improve individual diagnosis and treatment decisions. In this study, we characterized one such modifier in Drosophila melanogaster, the membrane-bound metallopeptidase CG14516 (superdeath). Loss of this enzyme suppresses a model of ER stress-induced degeneration by reducing cell death without altering the beneficial activation of the unfolded protein response. Our findings make superdeath and its orthologues attractive therapeutic targets in degenerative disease.


Genetics ◽  
2020 ◽  
Vol 214 (4) ◽  
pp. 913-925 ◽  
Author(s):  
Rebecca A. S. Palu ◽  
Hans M. Dalton ◽  
Clement Y. Chow

Endoplasmic reticulum (ER) stress-induced apoptosis is a primary cause and modifier of degeneration in a number of genetic disorders. Understanding how genetic variation influences the ER stress response and subsequent activation of apoptosis could improve individualized therapies and predictions of outcomes for patients. In this study, we find that the uncharacterized, membrane-bound metallopeptidase CG14516 in Drosophila melanogaster, which we rename as SUPpressor of ER stress-induced DEATH (superdeath), plays a role in modifying ER stress-induced apoptosis. We demonstrate that loss of superdeath reduces apoptosis and degeneration in the Rh1G69D model of ER stress through the JNK signaling cascade. This effect on apoptosis occurs without altering the activation of the unfolded protein response (IRE1 and PERK), suggesting that the beneficial prosurvival effects of this response are intact. Furthermore, we show that superdeath functions epistatically upstream of CDK5—a known JNK-activated proapoptotic factor in this model of ER stress. We demonstrate that superdeath is not only a modifier of this particular model, but affects the general tolerance to ER stress, including ER stress-induced apoptosis. Finally, we present evidence of Superdeath localization to the ER membrane. While similar in sequence to a number of human metallopeptidases found in the plasma membrane and ER membrane, its localization suggests that superdeath is orthologous to ERAP1/2 in humans. Together, this study provides evidence that superdeath is a link between stress in the ER and activation of cytosolic apoptotic pathways.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 790-790
Author(s):  
Manujendra N Saha ◽  
Hua Jiang ◽  
Asuka Mukai ◽  
Hong Chang

Abstract Abstract 790 TP53 mutations or deletions are relatively rare in multiple myeloma (MM). Thus, restoration of p53 tumor suppressor function in MM by blocking the inhibitory role of MDM2 is a promising and applicable therapeutic strategy. RITA (Reactivation of p53 and Induction of Tumor-cell Apoptosis), binds to p53, prevents the p53-MDM2 interaction and induces accumulation of p53 in solid tumors and leukemic cells. However, whether RITA exerts anti-myeloma activity is not known. Therefore, we examined the apoptotic effect of RITA and molecular mechanisms of RITA-induced apoptosis in MM cells. Treatment of MM cells with RITA caused a dose-dependent inhibition of survival and an increase in apoptosis of cells harboring wild type (MM.1S, H929) but not mutant (U266, LP1) or null p53 (8226R5) suggesting that RITA-induced apoptosis in MM cells is p53-dependent. MM cells were sensitive to low doses of RITA, killing about 80% of MM.1S or H929 cells at 2 or 6 μM, respectively. In addition, RITA elicited a dose-dependent inhibition of myeloma cell growth in 4 of the 5 primary MM samples tested. Importantly, RITA did not have a significant inhibitory effect on the survival of bone marrow or peripheral blood mononuclear cells obtained from 5 healthy donors at the concentrations that induced apoptosis of MM cells, indicating a preferential killing of myeloma cells by this drug. Furthermore, the combination of RITA with an MDM2 antagonist, nutlin displayed a strong synergism on the killing of H929 or MM.1S cells (CI=0.56-0.82). Western blot analysis showed that RITA induced an increase in the expression of p53 only in cells harboring wild type p53 but not mutant p53. The activation of p53 induced by RITA was mediated through modulation of multiple apoptotic regulatory proteins, including up-regulation of a pro-apoptotic protein Noxa, down-regulation of an anti-apoptotic protein Mcl-1 and activation of caspase-8 and -3. RITA-induced caspase-mediated apoptosis was followed by extrinsic pathway as shown by decrease in the percentage of annexin V-positive cells (p<0.05) as well as inhibition of cleavage of PARP and caspase-3 and/or caspase-8 activation in MM.1S or H929 cells when treated with either pan-caspase inhibitor, Z-VAD-FMK or caspase-8 specific inhibitor, Z-IETD-FMK. To further delineate the mechanisms of apoptosis, we performed gene expression profiling and identified up-regulation of transcription factors including JUN, DDIT3, ATF3 and ATF4, and down-regulation of survival factors, MYC and IGF1R in RITA-induced MM.1S cells, which were validated by qRT-pCR. Up-regulations of DDIT3, ATF3 and ATF4 are thought to be associated with the induction of endoplasmic reticulum (ER)-stress response. To examine whether apoptosis induction by RITA in MM cells is mediated through ER-stress response pathway, cells were treated with either an inhibitor of ER-stress-induced apoptosis, salubrinal, or a specific inhibitor of c-Jun N-terminal kinase (JNK), SP600125. Treatment of MM.1S or H929 cells with these inhibitors resulted in a significant inhibition of apoptosis induced by RITA as evidenced by inhibition of cleavage of caspase-3 and PARP, and decrease in the percentage of annexin V-positive cells (p<0.05). However, the above phenomenon was not observed in MM cells harboring mutant or null p53. Taken together, our results identify RITA as a promising agent in treatment of MM with two potential mechanisms of action: 1) activation of the p53 apoptotic pathway through up-regulation of Noxa and down-regulation of Mcl-1; and 2) p53-mediated induction of the ER-stress response pathways that may inhibit MM proliferation or survival. Our data provide the rationale for the clinical evaluation of RITA as a potential novel therapeutic intervention in MM. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 26 (21) ◽  
pp. 7999-8010 ◽  
Author(s):  
Genqing Liang ◽  
Timothy E. Audas ◽  
Yu Li ◽  
Gregory P. Cockram ◽  
J. Doug Dean ◽  
...  

ABSTRACT Luman/CREB3 (also called LZIP) is an endoplasmic reticulum (ER) membrane-bound transcription factor which is believed to undergo regulated intramembrane proteolysis in response to cellular cues. We previously found that Luman activates transcription from the unfolded protein response element. Here we report the identification of Herp, a gene involved in ER stress-associated protein degradation (ERAD), as a direct target of Luman. We found that Luman was transcriptionally induced and proteolytically activated by the ER stress inducer thaspsigargin. Overexpression of Luman activated transcription of cellular Herp via ER stress response element II (ERSE-II; ATTGG-N-CCACG) in the promoter region. Mutagenesis studies and chromatin immunoprecipitation assays showed that Luman physically associates with the Herp promoter, specifically the second half-site (CCACG) of ERSE-II. Luman was also necessary for the full activation of Herp during the ER stress response, since Luman small interfering RNA knockdown or functional repression by a dominant negative mutant attenuated Herp gene expression. Like Herp, overexpression of Luman protected cells against ER stress-induced apoptosis. With Luman structurally similar to ATF6 but resembling XBP1 in DNA-binding specificities, we propose that Luman is a novel factor that plays a role in ERAD and a converging point for various signaling pathways channeling through the ER.


2019 ◽  
Vol 116 (27) ◽  
pp. 13384-13393 ◽  
Author(s):  
Ronald A. Panganiban ◽  
Hae-Ryung Park ◽  
Maoyun Sun ◽  
Maya Shumyatcher ◽  
Blanca E. Himes ◽  
...  

Sensing misfolded proteins in the endoplasmic reticulum (ER), cells initiate the ER stress response and, when overwhelmed, undergo apoptosis. However, little is known about how cells prevent excessive ER stress response and cell death to restore homeostasis. Here, we report the identification and characterization of cellular suppressors of ER stress-induced apoptosis. Using a genome-wide CRISPR library, we screen for genes whose inactivation further increases ER stress-induced up-regulation of C/EBP homologous protein 10 (CHOP)—the transcription factor central to ER stress-associated apoptosis. Among the top validated hits are two interacting components of the polycomb repressive complex (L3MBTL2 [L(3)Mbt-Like 2] and MGA [MAX gene associated]), and microRNA-124-3 (miR-124-3). CRISPR knockout of these genes increases CHOP expression and sensitizes cells to apoptosis induced by multiple ER stressors, while overexpression confers the opposite effects. L3MBTL2 associates with the CHOP promoter in unstressed cells to repress CHOP induction but dissociates from the promoter in the presence of ER stress, whereas miR-124-3 directly targets the IRE1 branch of the ER stress pathway. Our study reveals distinct mechanisms that suppress ER stress-induced apoptosis and may lead to a better understanding of diseases whose pathogenesis is linked to overactive ER stress response.


2016 ◽  
Vol 103 ◽  
pp. 17-28 ◽  
Author(s):  
Kapil Sharma ◽  
Mohammad Ishaq ◽  
Gaurav Sharma ◽  
Mohammad Aslam Khan ◽  
Rajesh Kumar Dutta ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document