251
|
Effects of Ginsenoside Rg3 on Inhibiting Differentiation, Adipogenesis, and ER Stress-Mediated Cell Death in Brown Adipocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6668665. [PMID: 33815558 PMCID: PMC7990545 DOI: 10.1155/2021/6668665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/10/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022]
Abstract
Objectives Ginsenoside Rg3 (Rg3), a main active component of Panax ginseng, has various therapeutic properties in literatures, and it has been studied for its potential use in obesity control due to its antiadipogenic effects in white adipocytes. However, little is known about its effects on brown adipocytes. Methods The mechanisms through which Rg3 inhibits differentiation, adipogenesis, and ER stress-mediated cell death in mouse primary brown adipocytes (MPBAs) are explored. Results Rg3 significantly induced cytotoxicity in differentiated MPBAs but not in undifferentiated MPBAs. Rg3 treatment downregulated the expression of differentiation and adipogenesis markers and the level of perilipin in MPBAs while upregulating the expression of lipolytic Kruppel-like factor genes. Rg3 also induced lipolysis and efflux of triglycerides from MPBAs and subsequently increased proinflammatory cytokine levels. Notably, Rg3 treatment resulted in elevation of ER stress and proapoptotic markers in MPBAs. Conclusions Our results demonstrate that Rg3 is able to selectively exert cytotoxicity in differentiated MPBAs while leaving undifferentiated MPBAs intact, resulting in the induction of ER stress and subsequent cell death in MPBAs via regulation of various genes related to adipocyte differentiation, adipogenesis, lipolysis, and inflammation. These results indicate that further studies on the potential therapeutic applications of Rg3 are warranted.
Collapse
|
252
|
Yang J, Ding L, Yu L, Wang Y, Ge M, Jiang Q, Chen Y. Nanomedicine enables autophagy-enhanced cancer-cell ferroptosis. Sci Bull (Beijing) 2021; 66:464-477. [PMID: 36654184 DOI: 10.1016/j.scib.2020.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/29/2020] [Accepted: 10/17/2020] [Indexed: 01/20/2023]
Abstract
Ferroptosis and autophagy, playing significant roles in tumor treatment, are two typical forms of the programmed cell death. However, the rational combination of ferroptosis and autophagy for synergistic tumor therapy is still highly challenging. Herein, we report on an intriguing nanomedicine strategy for achieving autophagy-enhanced ferroptosis on efficiently combating cancer, which was based on the construction of trehalose-loaded mSiO2@MnOx-mPEG (TreMMM) nanoparticles with satisfactory biocompatibility. The nanoparticles are endowed with high glutathione (GSH) consumption efficiency, thereby inducing cancer-cell ferroptosis via inactivating glutathione peroxidases 4 (GPX4). Subsequently, the TreMMM degradation due to the GSH depletion and pH sensitivity contributed to the trehalose release for inducing autophagy, promoting/enhancing ferroptosis by NCOA4-mediated degradation of ferritin. A substantial in vitro and in vivo antitumor effect was achieved by such an intriguing autophagy-enhanced ferroptosis. Therefore, the rational combination of GSH-consumption-induced ferroptosis and trehalose-induced autophagy by nanomedicine design provides an alternative but effective strategy for tumor treatment.
Collapse
Affiliation(s)
- Jiacai Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Ding
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Luodan Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Yuemei Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Quzi Jiang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
253
|
Extracellular α-Synuclein Modulates Iron Metabolism Related Proteins via Endoplasmic Reticulum Stress in MES23.5 Dopaminergic Cells. Neurochem Res 2021; 46:1502-1513. [PMID: 33704649 DOI: 10.1007/s11064-021-03292-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
Alpha-synuclein plays a vital role in the pathology of Parkinson's disease (PD). Spreading of α-synuclein in neighboring cells was believed to contribute to progression in PD. How α-synuclein transmission affects adjacent cells is not full elucidated. Here, we used recombinant α-synuclein to mimic intercellular transmitted α-synuclein in MES23.5 dopaminergic cells, to investigate whether and how it could modulate iron metabolism. The results showed that α-synuclein treatment up-regulated divalent metal transporter 1 (DMT1) and down-regulated iron transporter (FPN), also up-regulated iron regulatory protein 1 (IRP1) protein levels and hepcidin mRNA levels. Endocytosis inhibitor dynasore pretreatment completely abolished and even reversed the upregulation of DMT1 and IRP1 induced by α-synuclein, however, FPN down-regulation was partially blocked by dynasore. Autophagy-inducing agent rapamycin reversed DMT1 up-regulation and FPN down-regulation, and fully blocked the upregulation of IRP1. Elevated hepcidin levels induced by α-synuclein was fully blocked by dynasore pretreatment, however, even higher with rapamycin pretreatment. Alpha-synuclein treatment triggered endoplasmic reticulum (ER) stress. ER stress inducer thapsigargin induced similar responses elicited by α-synuclein. ER stress inhibitor salubrinal blocked the up-regulation of IRP1 and hepcidin, as well as DMT1 up-regulation and FPN down-regulation, also dramatically abolished cAMP-response elements binding protein phosphorylation induced by α-synuclein. Taken together, these finding indicated that extracellular α-synuclein could regulate cellular iron metabolism, probably mediated by ER stress. It provides novel evidence to elucidate the relationships between transmitted α-synuclein and iron metabolism disturbance in PD.
Collapse
|
254
|
EGFRvIII Promotes Cell Survival during Endoplasmic Reticulum Stress through a Reticulocalbin 1-Dependent Mechanism. Cancers (Basel) 2021; 13:cancers13061198. [PMID: 33801941 PMCID: PMC7999088 DOI: 10.3390/cancers13061198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary A key molecule, EGFRvIII has been shown to provide several growth advantages for brain tumors. However, we have found a new mechanism in which the EGFRvIII provides increased survival to brain cancer cells when under sub-optimal conditions. Specifically, we have found that the EGFRvIII drives the expression of a molecule called Reticulocalbin 1 (RCN1) and that RCN1 blocks cell stress and cell death, thereby allowing cells to survive and proliferate. Importantly, these findings will allow for the generation of drugs that block the function of EGFRvIII and RCN1 with the hope that these drugs will induce brain cancer cell death. Abstract Reticulocalbin 1 (RCN1) is an endoplasmic reticulum (ER)-residing protein, involved in promoting cell survival during pathophysiological conditions that lead to ER stress. However, the key upstream receptor tyrosine kinase that regulates RCN1 expression and its potential role in cell survival in the glioblastoma setting have not been determined. Here, we demonstrate that RCN1 expression significantly correlates with poor glioblastoma patient survival. We also demonstrate that glioblastoma cells with expression of EGFRvIII receptor also have high RCN1 expression. Over-expression of wildtype EGFR also correlated with high RCN1 expression, suggesting that EGFR and EGFRvIII regulate RCN1 expression. Importantly, cells that expressed EGFRvIII and subsequently showed high RCN1 expression displayed greater cell viability under ER stress compared to EGFRvIII negative glioblastoma cells. Consistently, we also demonstrated that RCN1 knockdown reduced cell viability and exogenous introduction of RCN1 enhanced cell viability following induction of ER stress. Mechanistically, we demonstrate that the EGFRvIII-RCN1-driven increase in cell survival is due to the inactivation of the ER stress markers ATF4 and ATF6, maintained expression of the anti-apoptotic protein Bcl-2 and reduced activity of caspase 3/7. Our current findings identify that EGFRvIII regulates RCN1 expression and that this novel association promotes cell survival in glioblastoma cells during ER stress.
Collapse
|
255
|
Chen Z, Zhang J, Xue H, Qian M, Guo X, Gao X, Xu J, Qi Y, Sun X, Li G. Nitidine Chloride Is a Potential Alternative Therapy for Glioma Through Inducing Endoplasmic Reticulum Stress and Alleviating Epithelial-Mesenchymal Transition. Integr Cancer Ther 2021; 19:1534735419900927. [PMID: 32129091 PMCID: PMC7057402 DOI: 10.1177/1534735419900927] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Malignant glioma is a lethal brain tumor that is highly resistant to standard therapy. Our research aims to explore the suppressive effects of nitidine chloride (NC) on gliomas and the mechanisms involved, showing that it is a potential agent for integrative therapy of gliomas. Methods: After glioma cells were treated with NC, several experiments were performed to evaluate NC’s antitumor effects. CCK-8 assay was used to detect viability. Transwell and 3-dimensional spheroid invasion assays were used to evaluate motility of glioma in vitro, and the sphere-formation assay showed NC’s influence on glioma stem cells. Apoptosis and intracellular reactive oxygen species were measured by means of flow cytometry. Subcellular structures were observed through transmission electron microscopy. Western blot analysis reflected expression of endoplasmic reticulum (ER) stress and epithelial-mesenchymal transition (EMT) marker proteins. An orthotopic xenograft model was established to investigate the tumor suppressive effects in vivo. Results: Nitidine chloride inhibited glioma cell migration and invasion in vitro, downregulated the EMT proteins, and suppressed sphere formation of glioma stem cells. Furthermore, NC induced persistent ER stress that contributed to apoptosis and reactive oxygen species production. The xenograft model showed that NC effectively restricted glioma growth and invasion in vivo. Furthermore, we confirmed the signaling pathways that ER stress downregulates C/EBPβ and slug, as well as inhibition of the AKT/GSK3β/β-catenin axis caused by NC, in U-87 MG. Conclusion: We demonstrated that NC inhibits gliomas in vitro and in vivo by activating ER stress and downregulating EMT, which provides a basis for glioma therapy.
Collapse
Affiliation(s)
- Zihang Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Jinsen Zhang
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China.,Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Mingyu Qian
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Xing Guo
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Xiao Gao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Jianye Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| | - Xiaopeng Sun
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China.,Dezhou People's Hospital, Dezhou, Shandong, People's Republic of China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, People's Republic of China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, People's Republic of China
| |
Collapse
|
256
|
Du R, Sullivan DK, Azizian NG, Liu Y, Li Y. Inhibition of ERAD synergizes with FTS to eradicate pancreatic cancer cells. BMC Cancer 2021; 21:237. [PMID: 33676427 PMCID: PMC7937230 DOI: 10.1186/s12885-021-07967-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC), one of the most lethal cancers, is driven by oncogenic KRAS mutations. Farnesyl thiosalicylic acid (FTS), also known as salirasib, is a RAS inhibitor that selectively dislodges active RAS proteins from cell membrane, inhibiting downstream signaling. FTS has demonstrated limited therapeutic efficacy in PDAC patients despite being well tolerated. Methods To improve the efficacy of FTS in PDAC, we performed a genome-wide CRISPR synthetic lethality screen to identify genetic targets that synergize with FTS treatment. Among the top candidates, multiple genes in the endoplasmic reticulum-associated protein degradation (ERAD) pathway were identified. The role of ERAD inhibition in enhancing the therapeutic efficacy of FTS was further investigated in pancreatic cancer cells using pharmaceutical and genetic approaches. Results In murine and human PDAC cells, FTS induced unfolded protein response (UPR), which was further augmented upon treatment with a chemical inhibitor of ERAD, Eeyarestatin I (EerI). Combined treatment with FTS and EerI significantly upregulated the expression of UPR marker genes and induced apoptosis in pancreatic cancer cells. Furthermore, CRISPR-based genetic ablation of the key ERAD components, HRD1 and SEL1L, sensitized PDAC cells to FTS treatment. Conclusion Our study reveals a critical role for ERAD in therapeutic response of FTS and points to the modulation of UPR as a novel approach to improve the efficacy of FTS in PDAC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07967-6.
Collapse
Affiliation(s)
- Rong Du
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.,Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Delaney K Sullivan
- UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Nancy G Azizian
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yuanhui Liu
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yulin Li
- Center for Immunotherapy Research, Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
257
|
Gallot YS, Bohnert KR. Confounding Roles of ER Stress and the Unfolded Protein Response in Skeletal Muscle Atrophy. Int J Mol Sci 2021; 22:2567. [PMID: 33806433 PMCID: PMC7961896 DOI: 10.3390/ijms22052567] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is an essential organ, responsible for many physiological functions such as breathing, locomotion, postural maintenance, thermoregulation, and metabolism. Interestingly, skeletal muscle is a highly plastic tissue, capable of adapting to anabolic and catabolic stimuli. Skeletal muscle contains a specialized smooth endoplasmic reticulum (ER), known as the sarcoplasmic reticulum, composed of an extensive network of tubules. In addition to the role of folding and trafficking proteins within the cell, this specialized organelle is responsible for the regulated release of calcium ions (Ca2+) into the cytoplasm to trigger a muscle contraction. Under various stimuli, such as exercise, hypoxia, imbalances in calcium levels, ER homeostasis is disturbed and the amount of misfolded and/or unfolded proteins accumulates in the ER. This accumulation of misfolded/unfolded protein causes ER stress and leads to the activation of the unfolded protein response (UPR). Interestingly, the role of the UPR in skeletal muscle has only just begun to be elucidated. Accumulating evidence suggests that ER stress and UPR markers are drastically induced in various catabolic stimuli including cachexia, denervation, nutrient deprivation, aging, and disease. Evidence indicates some of these molecules appear to be aiding the skeletal muscle in regaining homeostasis whereas others demonstrate the ability to drive the atrophy. Continued investigations into the individual molecules of this complex pathway are necessary to fully understand the mechanisms.
Collapse
Affiliation(s)
- Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris Saclay, 91025 Evry, France
| | - Kyle R. Bohnert
- Kinesiology Department, St. Ambrose University, Davenport, IA 52803, USA
| |
Collapse
|
258
|
Unraveling the Molecular Nexus between GPCRs, ERS, and EMT. Mediators Inflamm 2021; 2021:6655417. [PMID: 33746610 PMCID: PMC7943314 DOI: 10.1155/2021/6655417] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent a large family of transmembrane proteins that transduce an external stimulus into a variety of cellular responses. They play a critical role in various pathological conditions in humans, including cancer, by regulating a number of key processes involved in tumor formation and progression. The epithelial-mesenchymal transition (EMT) is a fundamental process in promoting cancer cell invasion and tumor dissemination leading to metastasis, an often intractable state of the disease. Uncontrolled proliferation and persistent metabolism of cancer cells also induce oxidative stress, hypoxia, and depletion of growth factors and nutrients. These disturbances lead to the accumulation of misfolded proteins in the endoplasmic reticulum (ER) and induce a cellular condition called ER stress (ERS) which is counteracted by activation of the unfolded protein response (UPR). Many GPCRs modulate ERS and UPR signaling via ERS sensors, IRE1α, PERK, and ATF6, to support cancer cell survival and inhibit cell death. By regulating downstream signaling pathways such as NF-κB, MAPK/ERK, PI3K/AKT, TGF-β, and Wnt/β-catenin, GPCRs also upregulate mesenchymal transcription factors including Snail, ZEB, and Twist superfamilies which regulate cell polarity, cytoskeleton remodeling, migration, and invasion. Likewise, ERS-induced UPR upregulates gene transcription and expression of proteins related to EMT enhancing tumor aggressiveness. Though GPCRs are attractive therapeutic targets in cancer biology, much less is known about their roles in regulating ERS and EMT. Here, we will discuss the interplay in GPCR-ERS linked to the EMT process of cancer cells, with a particular focus on oncogenes and molecular signaling pathways.
Collapse
|
259
|
Yang J, Ding L, Yu L, Wang Y, Ge M, Jiang Q, Chen Y. Nanomedicine enables autophagy-enhanced cancer-cell ferroptosis. Sci Bull (Beijing) 2021; 66:464-477. [DOI: doi.org/10.1016/j.scib.2020.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
|
260
|
Kitajima S, Sun W, Lee KL, Ho JC, Oyadomari S, Okamoto T, Masai H, Poellinger L, Kato H. A KDM6 inhibitor potently induces ATF4 and its target gene expression through HRI activation and by UTX inhibition. Sci Rep 2021; 11:4538. [PMID: 33633164 PMCID: PMC7907191 DOI: 10.1038/s41598-021-83857-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
UTX/KDM6A encodes a major histone H3 lysine 27 (H3K27) demethylase, and is frequently mutated in various types of human cancers. Although UTX appears to play a crucial role in oncogenesis, the mechanisms involved are still largely unknown. Here we show that a specific pharmacological inhibitor of H3K27 demethylases, GSK-J4, induces the expression of transcription activating factor 4 (ATF4) protein as well as the ATF4 target genes (e.g. PCK2, CHOP, REDD1, CHAC1 and TRIB3). ATF4 induction by GSK-J4 was due to neither transcriptional nor post-translational regulation. In support of this view, the ATF4 induction was almost exclusively dependent on the heme-regulated eIF2α kinase (HRI) in mouse embryonic fibroblasts (MEFs). Gene expression profiles with UTX disruption by CRISPR-Cas9 editing and the following stable re-expression of UTX showed that UTX specifically suppresses the expression of the ATF4 target genes, suggesting that UTX inhibition is at least partially responsible for the ATF4 induction. Apoptosis induction by GSK-J4 was partially and cell-type specifically correlated with the activation of ATF4-CHOP. These findings highlight that the anti-cancer drug candidate GSK-J4 strongly induces ATF4 and its target genes via HRI activation and raise a possibility that UTX might modulate cancer formation by regulating the HRI-ATF4 axis.
Collapse
Affiliation(s)
- Shojiro Kitajima
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Republic of Singapore ,grid.26091.3c0000 0004 1936 9959Institute for Advanced Biosciences, Keio University, Kakuganji 246-2, Mizukami, Tsuruoka, Yamagata 997-0052 Japan
| | - Wendi Sun
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Republic of Singapore
| | - Kian Leong Lee
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Republic of Singapore ,grid.428397.30000 0004 0385 0924Cancer & Stem Cell Biology Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Republic of Singapore
| | - Jolene Caifeng Ho
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Republic of Singapore
| | - Seiichi Oyadomari
- grid.267335.60000 0001 1092 3579Institute of Advanced Medical Sciences, Tokushima University, Tokushima, 770-8503 Japan
| | - Takashi Okamoto
- grid.260433.00000 0001 0728 1069Department of Molecular and Cellular Biology, Nagoya City University Graduate School of Medical Science, Mizuho-ku, Nagoya, 467-8601 Japan
| | - Hisao Masai
- grid.272456.0Genome Dynamics Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506 Japan
| | - Lorenz Poellinger
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Republic of Singapore ,Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Hiroyuki Kato
- grid.4280.e0000 0001 2180 6431Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599 Republic of Singapore ,grid.260433.00000 0001 0728 1069Department of Molecular and Cellular Biology, Nagoya City University Graduate School of Medical Science, Mizuho-ku, Nagoya, 467-8601 Japan ,grid.272456.0Genome Dynamics Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506 Japan
| |
Collapse
|
261
|
Gilardini Montani MS, Benedetti R, Piconese S, Pulcinelli FM, Timperio AM, Romeo MA, Masuelli L, Mattei M, Bei R, D'Orazi G, Cirone M. PGE2 Released by Pancreatic Cancer Cells Undergoing ER Stress Transfers the Stress to DCs Impairing Their Immune Function. Mol Cancer Ther 2021; 20:934-945. [PMID: 33632872 DOI: 10.1158/1535-7163.mct-20-0699] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/24/2020] [Accepted: 02/09/2021] [Indexed: 11/16/2022]
Abstract
This study shows that pancreatic cancer cells undergoing cell death by valproic acid (VPA) treatment activated dendritic cells (DCs) more efficiently than those treated with trichostatin A (TSA), as demonstrated by CD86 and CD80 surface expression. Surprisingly though, DCs cultured in the presence of supernatant derived from VPA-treated cancer cells showed a reduced allostimulatory capacity and an increased release of IL10 and IL8 cytokines in comparison with those exposed to TSA-treated cell culture supernatant. Searching for molecular mechanisms leading to such differences, we found that VPA treatment dysregulated choline metabolism and triggered a stronger endoplasmic reticulum (ER) stress in pancreatic cancer cells than TSA, upregulating CCAAT/enhancer-binding protein homologous protein, and activated cyclooxygenase-2, thus promoting the release of prostaglandin (PG) E2. Interestingly, dysfunctional DCs cultured in the presence of VPA-treated cells culture supernatant showed a higher level of intracellular reactive oxygen species, 4-hydroxy-trans-2-nonenal protein adducts, and ER stress, as evidenced by the upregulation of spliced X-box binding protein 1 (XBP1s), effects that were reduced when DCs were exposed to supernatant of cancer cells treated with Celecoxib before VPA. Celecoxib prevented PGE2 release, restoring the function of DCs exposed to VPA-treated cells culture supernatant, and a similar effect was obtained by silencing XBP1s in DCs treated with VPA-treated cells culture supernatant. These results suggest that PGE2 could be one of the yet unidentified factors able to transfer the stress from cancer cells to DCs, resulting in an impairment of their function.
Collapse
Affiliation(s)
- Maria Saveria Gilardini Montani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Rossella Benedetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Silvia Piconese
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.,Department of Internal Clinical, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Anna Maria Timperio
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Maria Anele Romeo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Maurizio Mattei
- Centro di Servizi Interdipartimentale-Stazione per la Tecnologia Animale, University of Rome "Tor Vergata," Rome, Italy; Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome 'Tor Vergata,' Rome, Italy
| | - Gabriella D'Orazi
- Department of Research, IRCCS Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy. .,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
262
|
Straughn AR, Kelm NQ, Kakar SS. Withaferin A and Ovarian Cancer Antagonistically Regulate Skeletal Muscle Mass. Front Cell Dev Biol 2021; 9:636498. [PMID: 33718372 PMCID: PMC7947350 DOI: 10.3389/fcell.2021.636498] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/05/2021] [Indexed: 01/06/2023] Open
Abstract
Cachexia is a complex wasting syndrome that overwhelmingly affects the majority of late-stage cancer patients. Additionally, there are currently no efficacious therapeutic agents to treat the muscle atrophy induced by the cancer. While several preclinical studies have investigated the molecular signals orchestrating cachexia, very little information exists pertaining to ovarian cancer and the associated cachexia. Work from our lab has recently demonstrated that the steroidal lactone Withaferin A (WFA) is capable of attenuating the atrophying effects of ovarian cancer in a preclinical mouse model. However, it remained to be determined whether WFA's effect was in response to its anti-tumorigenic properties, or if it was capable of targeting skeletal muscle directly. The purpose of this study was to uncover whether WFA was capable of regulating muscle mass under tumor-free and tumor-bearing conditions. Treatment with WFA led to an improvement in functional muscle strength and mass under tumor-bearing and naïve conditions. WFA and ovarian cancer were observed to act antagonistically upon critical skeletal muscle regulatory systems, notably myogenic progenitors and proteolytic degradation pathways. Our results demonstrated for the first time that, while WFA has anti-tumorigenic properties, it also exerts hypertrophying effects on skeletal muscle mass, suggesting that it could be an anti-cachectic agent in the settings of ovarian cancer.
Collapse
Affiliation(s)
- Alex R. Straughn
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Natia Q. Kelm
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Sham S. Kakar
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Department of Physiology, University of Louisville, Louisville, KY, United States
| |
Collapse
|
263
|
de la Harpe A, Beukes N, Frost CL. CBD activation of TRPV1 induces oxidative signaling and subsequent ER stress in breast cancer cell lines. Biotechnol Appl Biochem 2021; 69:420-430. [PMID: 33604949 DOI: 10.1002/bab.2119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum (ER) stress is an imbalance between the protein-folding load and capacity of ER. It can be induced by various physiological conditions, activating the unfolded protein response (UPR) to re-establish homeostasis, promoting cell survival. Under severe or chronic stress, apoptosis is induced. Normal cells generally do not experience continuous ER stress induction. The stressful conditions experienced in the tumor microenvironment facilitate chronic ER stress and UPR activation, which plays a pivotal role in tumour survival. Exacerbation of pre-existing ER stress can trigger cancer cell death, with a minimal effect on normal cells. Current literature suggests that cannabinoid treatment may induce cancer cell death via ER stress; however, little is known about the mechanisms of induction. This study proposed that cannabidiol (CBD) mechanism occurred through the influx of Ca2+ via the TRPV1 receptor, and increasing reactive oxygen species (ROS) production affects protein folding and induces ER stress. ER stress was induced, and detection and quantification were completed using Thioflavin T staining and GRP78 by western blot analysis. The effect of cannabinoid treatment on ROS production and Ca2+ influx was measured. CBD was the most potent ER stress inducer, significantly increasing Ca2+ and ROS accumulation. Concomitant treatment with CBD and an antioxidant significantly increased cell viability and decreased ER stress induction in the MCF7 cell line. Concomitant treatment with a TRPV1 antagonist increased viability in this cell line. In conclusion, the data suggested that CBD induced ER stress via Ca2+ influx through the TRPV1 receptor, thereby elevating intracellular ROS levels and disrupting protein folding.
Collapse
Affiliation(s)
- Amy de la Harpe
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Natasha Beukes
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| | - Carminita L Frost
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth, South Africa
| |
Collapse
|
264
|
Qu J, Zou T, Lin Z. The Roles of the Ubiquitin-Proteasome System in the Endoplasmic Reticulum Stress Pathway. Int J Mol Sci 2021; 22:1526. [PMID: 33546413 PMCID: PMC7913544 DOI: 10.3390/ijms22041526] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is a highly dynamic organelle in eukaryotic cells, which is essential for synthesis, processing, sorting of protein and lipid metabolism. However, the cells activate a defense mechanism called endoplasmic reticulum stress (ER stress) response and initiate unfolded protein response (UPR) as the unfolded proteins exceed the folding capacity of the ER due to the environmental influences or increased protein synthesis. ER stress can mediate many cellular processes, including autophagy, apoptosis and senescence. The ubiquitin-proteasome system (UPS) is involved in the degradation of more than 80% of proteins in the cells. Today, increasing numbers of studies have shown that the two important components of UPS, E3 ubiquitin ligases and deubiquitinases (DUBs), are tightly related to ER stress. In this review, we summarized the regulation of the E3 ubiquitin ligases and DUBs in ER stress.
Collapse
Affiliation(s)
| | | | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China; (J.Q.); (T.Z.)
| |
Collapse
|
265
|
LW-213 induces cell apoptosis in human cutaneous T-cell lymphomas by activating PERK-eIF2α-ATF4-CHOP axis. Acta Pharmacol Sin 2021; 42:290-300. [PMID: 32747719 DOI: 10.1038/s41401-020-0466-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/22/2020] [Indexed: 11/08/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is characterized by a heterogeneous group of extranodal non-Hodgkin lymphomas, in which monoclonal T lymphocytes infiltrate the skin. LW-213, a derivative of wogonin, was found to induce cell apoptosis in chronic myeloid leukemia (CML). In this study, we investigated the effects of LW-213 on CTCL cells and the underlying mechanisms. We showed that LW-213 (1-25 μM) dose-dependently inhibited human CTCL cell lines (Hut-102, Hut-78, MyLa, and HH) with IC50 values of around 10 μM, meanwhile it potently inhibited primary leukemia cells derived from peripheral blood of T-cell lymphoma patients. We revealed that LW-213-induced apoptosis was accompanied by ROS formation and the release of calcium from endoplasmic reticulum (ER) through IP3R-1channel. LW-213 selectively activated CHOP and induced apoptosis in Hut-102 cells via activating PERK-eIF2α-ATF4 pathway. Interestingly, the degree of apoptosis and expression of ER stress-related proteins were alleviated in the presence of either N-acetyl cysteine (NAC), an ROS scavenger, or 2-aminoethyl diphenylborinate (2-APB), an IP3R-1 inhibitor, implicating ROS/calcium-dependent ER stress in LW-213-induced apoptosis. In NOD/SCID mice bearing Hut-102 cell line xenografts, administration of LW-213 (10 mg/kg, ip, every other day for 4 weeks) markedly inhibited the growth of Hut-102 derived xenografts and prolonged survival. In conclusion, our study provides a new insight into the mechanism of LW-213-induced apoptosis, suggesting the potential of LW-213 as a promising agent against CTCL.
Collapse
|
266
|
Huang J, Pan H, Wang J, Wang T, Huo X, Ma Y, Lu Z, Sun B, Jiang H. Unfolded protein response in colorectal cancer. Cell Biosci 2021; 11:26. [PMID: 33514437 PMCID: PMC7844992 DOI: 10.1186/s13578-021-00538-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a gastrointestinal malignancy originating from either the colon or the rectum. A growing number of researches prove that the unfolded protein response (UPR) is closely related to the occurrence and progression of colorectal cancer. The UPR has three canonical endoplasmic reticulum (ER) transmembrane protein sensors: inositol requiring kinase 1 (IRE1), pancreatic ER eIF2α kinase (PERK), and activating transcription factor 6 (ATF6). Each of the three pathways is closely associated with CRC development. The three pathways are relatively independent as well as interrelated. Under ER stress, the activated UPR boosts the protein folding capacity to maximize cell adaptation and survival, whereas sustained or excessive ER triggers cell apoptosis conversely. The UPR involves different stages of CRC pathogenesis, promotes or hinders the progression of CRC, and will pave the way for novel therapeutic and diagnostic approaches. Meanwhile, the correlation between different signal branches in UPR and the switch between the adaptation and apoptosis pathways still need to be further investigated in the future.
Collapse
Affiliation(s)
- Jingjing Huang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Huayang Pan
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Jinge Wang
- The Second Affiliated Hospital & College of Nursing, Harbin Medical University, Harbin, People's Republic of China
| | - Tong Wang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Xiaoyan Huo
- Pediatrics Department of The First Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Yong Ma
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Zhaoyang Lu
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Bei Sun
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China
| | - Hongchi Jiang
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, People's Republic of China.
| |
Collapse
|
267
|
Fu X, Cui J, Meng X, Jiang P, Zheng Q, Zhao W, Chen X. Endoplasmic reticulum stress, cell death and tumor: Association between endoplasmic reticulum stress and the apoptosis pathway in tumors (Review). Oncol Rep 2021; 45:801-808. [PMID: 33469681 PMCID: PMC7859917 DOI: 10.3892/or.2021.7933] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
External and internal stimuli are often involved in the pathogenesis of tumors, and the deterioration of endoplasmic reticulum (ER) function within cells is also an important etiological factor of tumorigenesis resulting in the impairment of the endoplasmic reticulum, which is termed ER stress. The ER is an organelle that serves a crucial role in the process of protein synthesis and maturation, and also acts as a reservoir of calcium to maintain intracellular Ca2+ homeostasis. ER stress has been revealed to serve a critical role in tumorigenesis. In the present review, the association between ER stress‑related pathways and tumor cell apoptosis is examined. Primarily, the role of ER stress in tumor cell apoptosis is discussed, and it is stipulated that ER stress, induced by drugs both directly and indirectly, promotes tumor cell apoptosis.
Collapse
Affiliation(s)
- Xiaojing Fu
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Juanjuan Cui
- Qingdao Municipal Hospital, Qingdao (Group), Qingdao, Shandong 266071, P.R. China
| | - Xiangjun Meng
- Qingdao Mental Health Center, Qingdao, Shandong 266071, P.R. China
| | - Piyu Jiang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Qiuling Zheng
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Wenwen Zhao
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
268
|
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is caused by the accumulation of fat in over 5% of hepatocytes in the absence of alcohol consumption. NAFLD is considered the hepatic manifestation of metabolic syndrome (MS). Recently, an expert consensus suggested as more appropriate the term MAFLD (metabolic-associated fatty liver disease). Insulin resistance (IR) plays a key role in the development of NAFLD, as it causes an increase in hepatic lipogenesis and an inhibition of adipose tissue lipolysis. Beyond the imbalance of adipokine levels, the increase in the mass of visceral adipose tissue also determines an increase in free fatty acid (FFA) levels. In turn, an excess of FFA is able to determine IR through the inhibition of the post-receptor insulin signal. Adipocytes secrete chemokines, which are able to enroll macrophages inside the adipose tissue, responsible, in turn, for the increased levels of TNF-α. The latter, as well as resistin and other pro-inflammatory cytokines such as IL-6, enhances insulin resistance and correlates with endothelial dysfunction and an increased cardiovascular (CV) risk. In this review, the role of diet, intestinal microbiota, genetic and epigenetic factors, low-degree chronic systemic inflammation, mitochondrial dysfunction, and endoplasmic reticulum stress on NAFLD have been addressed. Finally, the clinical impact of NAFLD on cardiovascular and renal outcomes, and its direct link with type 2 diabetes have been discussed.
Collapse
|
269
|
Harnessing the Co-vulnerabilities of Amino Acid-Restricted Cancers. Cell Metab 2021; 33:9-20. [PMID: 33406406 PMCID: PMC7837405 DOI: 10.1016/j.cmet.2020.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/18/2022]
Abstract
Sustained proliferative potential of cancer cells creates heightened energetic and biosynthetic demands. The resulting overt dependence of cancer cells on unperturbed nutrient supply has prompted a widespread interest in amino acid restriction strategies as potential cancer therapeutics. However, owing to rapid signaling and metabolic reprogramming in cancer cells, the prospects for success of amino acid restriction approaches remain unclear. We thus recognize that the identification of co-vulnerabilities of amino acid-restricted cancers may inform actionable targets for effective combined interventions. In this perspective, we outline the current state of key cellular mechanisms underlying adaptation to amino acid restriction and discuss the role of signal transduction pathways governing cancer cell resistance to amino acid restriction, with potential ramifications for the design of future therapeutic efforts.
Collapse
|
270
|
Cho J, Min HY, Lee HJ, Hyun SY, Sim JY, Noh M, Hwang SJ, Park SH, Boo HJ, Lee HJ, Hong S, Park RW, Shin YK, Hung MC, Lee HY. RGS2-mediated translational control mediates cancer cell dormancy and tumor relapse. J Clin Invest 2021; 131:136779. [PMID: 33393490 PMCID: PMC7773398 DOI: 10.1172/jci136779] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Slow-cycling/dormant cancer cells (SCCs) have pivotal roles in driving cancer relapse and drug resistance. A mechanistic explanation for cancer cell dormancy and therapeutic strategies targeting SCCs are necessary to improve patient prognosis, but are limited because of technical challenges to obtaining SCCs. Here, by applying proliferation-sensitive dyes and chemotherapeutics to non-small cell lung cancer (NSCLC) cell lines and patient-derived xenografts, we identified a distinct SCC subpopulation that resembled SCCs in patient tumors. These SCCs displayed major dormancy-like phenotypes and high survival capacity under hostile microenvironments through transcriptional upregulation of regulator of G protein signaling 2 (RGS2). Database analysis revealed RGS2 as a biomarker of retarded proliferation and poor prognosis in NSCLC. We showed that RGS2 caused prolonged translational arrest in SCCs through persistent eukaryotic initiation factor 2 (eIF2α) phosphorylation via proteasome-mediated degradation of activating transcription factor 4 (ATF4). Translational activation through RGS2 antagonism or the use of phosphodiesterase 5 inhibitors, including sildenafil (Viagra), promoted ER stress-induced apoptosis in SCCs in vitro and in vivo under stressed conditions, such as those induced by chemotherapy. Our results suggest that a low-dose chemotherapy and translation-instigating pharmacological intervention in combination is an effective strategy to prevent tumor progression in NSCLC patients after rigorous chemotherapy.
Collapse
Affiliation(s)
- Jaebeom Cho
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Ho Jin Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Seung Yeob Hyun
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Jeong Yeon Sim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Myungkyung Noh
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
| | - Su Jung Hwang
- College of Pharmacy, Inje University, Gimhae, Gyungnam, Republic of Korea
| | - Shin-Hyung Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Hye-Jin Boo
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Hyo-Jong Lee
- College of Pharmacy, Inje University, Gimhae, Gyungnam, Republic of Korea
| | - Sungyoul Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, and Cell & Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Young Kee Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, and
| |
Collapse
|
271
|
Minnee E, Faller WJ. Translation initiation and its relevance in colorectal cancer. FEBS J 2021; 288:6635-6651. [PMID: 33382175 PMCID: PMC9291299 DOI: 10.1111/febs.15690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 01/08/2023]
Abstract
Protein synthesis is one of the most essential processes in every kingdom of life, and its dysregulation is a known driving force in cancer development. Multiple signaling pathways converge on the translation initiation machinery, and this plays a crucial role in regulating differential gene expression. In colorectal cancer, dysregulation of initiation results in translational reprogramming, which promotes the selective translation of mRNAs required for many oncogenic processes. The majority of upstream mutations found in colorectal cancer, including alterations in the WNT, MAPK, and PI3K\AKT pathways, have been demonstrated to play a significant role in translational reprogramming. Many translation initiation factors are also known to be dysregulated, resulting in translational reprogramming during tumor initiation and/or maintenance. In this review, we outline the role of translational reprogramming that occurs during colorectal cancer development and progression and highlight some of the most critical factors affecting the etiology of this disease.
Collapse
Affiliation(s)
- Emma Minnee
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - William James Faller
- Division of Oncogenomics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
272
|
Carlos AJ, Ha DP, Yeh DW, Van Krieken R, Tseng CC, Zhang P, Gill P, Machida K, Lee AS. The chaperone GRP78 is a host auxiliary factor for SARS-CoV-2 and GRP78 depleting antibody blocks viral entry and infection. J Biol Chem 2021; 296:100759. [PMID: 33965375 PMCID: PMC8102082 DOI: 10.1016/j.jbc.2021.100759] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/27/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the COVID-19 global pandemic, utilizes the host receptor angiotensin-converting enzyme 2 (ACE2) for viral entry. However, other host factors might also play important roles in SARS-CoV-2 infection, providing new directions for antiviral treatments. GRP78 is a stress-inducible chaperone important for entry and infectivity for many viruses. Recent molecular docking analyses revealed putative interaction between GRP78 and the receptor-binding domain (RBD) of the SARS-CoV-2 Spike protein (SARS-2-S). Here we report that GRP78 can form a complex with SARS-2-S and ACE2 on the surface and at the perinuclear region typical of the endoplasmic reticulum in VeroE6-ACE2 cells and that the substrate-binding domain of GRP78 is critical for this interaction. In vitro binding studies further confirmed that GRP78 can directly bind to the RBD of SARS-2-S and ACE2. To investigate the role of GRP78 in this complex, we knocked down GRP78 in VeroE6-ACE2 cells. Loss of GRP78 markedly reduced cell surface ACE2 expression and led to activation of markers of the unfolded protein response. Treatment of lung epithelial cells with a humanized monoclonal antibody (hMAb159) selected for its safe clinical profile in preclinical models depleted cell surface GRP78 and reduced cell surface ACE2 expression, as well as SARS-2-S-driven viral entry and SARS-CoV-2 infection in vitro. Our data suggest that GRP78 is an important host auxiliary factor for SARS-CoV-2 entry and infection and a potential target to combat this novel pathogen and other viruses that utilize GRP78 in combination therapy.
Collapse
Affiliation(s)
- Anthony J Carlos
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Da-Wei Yeh
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Richard Van Krieken
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Chun-Chih Tseng
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Pu Zhang
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Parkash Gill
- USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Keigo Machida
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
273
|
Adaptation to Endoplasmic Reticulum Stress Enhances Resistance of Oral Cancer Cells to Cisplatin by Up-Regulating Polymerase η and Increasing DNA Repair Efficiency. Int J Mol Sci 2020; 22:ijms22010355. [PMID: 33396303 PMCID: PMC7794796 DOI: 10.3390/ijms22010355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Endoplasmic reticulum (ER) stress response is an adaptive program to cope with cellular stress that disturbs the function and homeostasis of ER, which commonly occurs during cancer progression to late stage. Late-stage cancers, mostly requiring chemotherapy, often develop treatment resistance. Chemoresistance has been linked to ER stress response; however, most of the evidence has come from studies that correlate the expression of stress markers with poor prognosis or demonstrate proapoptosis by the knockdown of stress-responsive genes. Since ER stress in cancers usually persists and is essentially not induced by genetic manipulations, we used low doses of ER stress inducers at levels that allowed cell adaptation to occur in order to investigate the effect of stress response on chemoresistance. We found that prolonged tolerable ER stress promotes mesenchymal-epithelial transition, slows cell-cycle progression, and delays the S-phase exit. Consequently, cisplatin-induced apoptosis was significantly decreased in stress-adapted cells, implying their acquisition of cisplatin resistance. Molecularly, we found that proliferating cell nuclear antigen (PCNA) ubiquitination and the expression of polymerase η, the main polymerase responsible for translesion synthesis across cisplatin-DNA damage, were up-regulated in ER stress-adaptive cells, and their enhanced cisplatin resistance was abrogated by the knockout of polymerase η. We also found that a fraction of p53 in stress-adapted cells was translocated to the nucleus, and that these cells exhibited a significant decline in the level of cisplatin-DNA damage. Consistently, we showed that the nuclear p53 coincided with strong positivity of glucose-related protein 78 (GRP78) on immunostaining of clinical biopsies, and the cisplatin-based chemotherapy was less effective for patients with high levels of ER stress. Taken together, this study uncovers that adaptation to ER stress enhances DNA repair and damage tolerance, with which stressed cells gain resistance to chemotherapeutics.
Collapse
|
274
|
Thapsigargin-From Traditional Medicine to Anticancer Drug. Int J Mol Sci 2020; 22:ijms22010004. [PMID: 33374919 PMCID: PMC7792614 DOI: 10.3390/ijms22010004] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
A sesquiterpene lactone, thapsigargin, is a phytochemical found in the roots and fruits of Mediterranean plants from Thapsia L. species that have been used for centuries in folk medicine to treat rheumatic pain, lung diseases, and female infertility. More recently thapsigargin was found to be a potent cytotoxin that induces apoptosis by inhibiting the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) pump, which is necessary for cellular viability. This biological activity encouraged studies on the use of thapsigargin as a novel antineoplastic agent, which were, however, hampered due to high toxicity of this compound to normal cells. In this review, we summarized the recent knowledge on the biological activity and molecular mechanisms of thapsigargin action and advances in the synthesis of less-toxic thapsigargin derivatives that are being developed as novel anticancer drugs.
Collapse
|
275
|
Human embryonic stem cell-derived organoid retinoblastoma reveals a cancerous origin. Proc Natl Acad Sci U S A 2020; 117:33628-33638. [PMID: 33318192 PMCID: PMC7776986 DOI: 10.1073/pnas.2011780117] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
As a genetic malignancy, retinoblastoma (Rb) is caused by RB1 mutations; however, its developmental origin and drug agents for human Rb remain largely unexplored. Here we describe an innovative Rb organoid model derived from human embryonic stem cells with a biallelic mutagenesis of the RB1 gene. We identify tumorigenic growth in the Rb organoids, as well as properties consistent with human primary Rb. We confirm that the Rb cell of origin stemmed from ARR3+ maturing cone precursor cells and SYK inhibitors displaying a significant therapeutic response. Our elegant in-dish Rb organoid model can be used to efficiently and effectively dissect the origin of Rb and mechanisms of Rb tumorigenesis, as well as screen novel therapies. Retinoblastoma (Rb) is the most prevalent intraocular malignancy in children, with a worldwide survival rate <30%. We have developed a cancerous model of Rb in retinal organoids derived from genetically engineered human embryonic stem cells (hESCs) with a biallelic mutagenesis of the RB1 gene. These organoid Rbs exhibit properties highly consistent with Rb tumorigenesis, transcriptome, and genome-wide methylation. Single-cell sequencing analysis suggests that Rb originated from ARR3-positive maturing cone precursors during development, which was further validated by immunostaining. Notably, we found that the PI3K-Akt pathway was aberrantly deregulated and its activator spleen tyrosine kinase (SYK) was significantly up-regulated. In addition, SYK inhibitors led to remarkable cell apoptosis in cancerous organoids. In conclusion, we have established an organoid Rb model derived from genetically engineered hESCs in a dish that has enabled us to trace the cell of origin and to test novel candidate therapeutic agents for human Rb, shedding light on the development and therapeutics of other malignancies.
Collapse
|
276
|
Activation of the IRE1 RNase through remodeling of the kinase front pocket by ATP-competitive ligands. Nat Commun 2020; 11:6387. [PMID: 33318494 PMCID: PMC7736581 DOI: 10.1038/s41467-020-19974-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
Inositol-Requiring Enzyme 1 (IRE1) is an essential component of the Unfolded Protein Response. IRE1 spans the endoplasmic reticulum membrane, comprising a sensory lumenal domain, and tandem kinase and endoribonuclease (RNase) cytoplasmic domains. Excess unfolded proteins in the ER lumen induce dimerization and oligomerization of IRE1, triggering kinase trans-autophosphorylation and RNase activation. Known ATP-competitive small-molecule IRE1 kinase inhibitors either allosterically disrupt or stabilize the active dimeric unit, accordingly inhibiting or stimulating RNase activity. Previous allosteric RNase activators display poor selectivity and/or weak cellular activity. In this study, we describe a class of ATP-competitive RNase activators possessing high selectivity and strong cellular activity. This class of activators binds IRE1 in the kinase front pocket, leading to a distinct conformation of the activation loop. Our findings reveal exquisitely precise interdomain regulation within IRE1, advancing the mechanistic understanding of this important enzyme and its investigation as a potential small-molecule therapeutic target. The RNase activity of Inositol-Requiring Enzyme 1 (IRE1) can be allosterically regulated by ATP-competitive inhibitors of the IRE1 kinase domain. Here, the authors identify ATP-competitive IRE1 RNase activators with improved selectivity and cellular activity, and elucidate their mechanism of action.
Collapse
|
277
|
Liu Y, Tan Z, Yang Y. Negative feedback and modern anti-cancer strategies targeting the ER stress response. FEBS Lett 2020; 594:4247-4265. [PMID: 33206409 DOI: 10.1002/1873-3468.14000] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum (ER) stress is a cell state in which misfolded or unfolded proteins are aberrantly accumulated in the ER. ER stress induces an evolutionarily conserved adaptive response, named the ER stress response, that deploys a self-regulated machinery to maintain cellular proteostasis. However, compared to its well-established canonical activation mechanism, the negative feedback mechanisms regulating the ER stress response remain unclear and no accepted methods or markers have been established. Several studies have documented that both endogenous and exogenous insults can induce ER stress in cancer. Based on this evidence, small molecule inhibitors targeting ER stress response have been designed to kill cancer cells, with some of them showing excellent curative effects. Here, we review recent advances in our understanding of negative feedback of the ER stress response and compare the markers used to date. We also summarize therapeutic inhibitors targeting ER stress response and highlight the promises and challenges ahead.
Collapse
Affiliation(s)
- Yaofu Liu
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Zhenzhi Tan
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| |
Collapse
|
278
|
Characterization of constitutive ER-phagy of excess membrane proteins. PLoS Genet 2020; 16:e1009255. [PMID: 33275594 PMCID: PMC7744050 DOI: 10.1371/journal.pgen.1009255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/16/2020] [Accepted: 11/05/2020] [Indexed: 11/19/2022] Open
Abstract
Thirty percent of all cellular proteins are inserted into the endoplasmic reticulum (ER), which spans throughout the cytoplasm. Two well-established stress-induced pathways ensure quality control (QC) at the ER: ER-phagy and ER-associated degradation (ERAD), which shuttle cargo for degradation to the lysosome and proteasome, respectively. In contrast, not much is known about constitutive ER-phagy. We have previously reported that excess of integral-membrane proteins is delivered from the ER to the lysosome via autophagy during normal growth of yeast cells. Whereas endogenously expressed ER resident proteins serve as cargos at a basal level, this level can be induced by overexpression of membrane proteins that are not ER residents. Here, we characterize this pathway as constitutive ER-phagy. Constitutive and stress-induced ER-phagy share the basic macro-autophagy machinery including the conserved Atgs and Ypt1 GTPase. However, induction of stress-induced autophagy is not needed for constitutive ER-phagy to occur. Moreover, the selective receptors needed for starvation-induced ER-phagy, Atg39 and Atg40, are not required for constitutive ER-phagy and neither these receptors nor their cargos are delivered through it to the vacuole. As for ERAD, while constitutive ER-phagy recognizes cargo different from that recognized by ERAD, these two ER-QC pathways can partially substitute for each other. Because accumulation of membrane proteins is associated with disease, and constitutive ER-phagy players are conserved from yeast to mammalian cells, this process could be critical for human health.
Collapse
|
279
|
Zhao T, Du J, Zeng H. Interplay between endoplasmic reticulum stress and non-coding RNAs in cancer. J Hematol Oncol 2020; 13:163. [PMID: 33267910 PMCID: PMC7709275 DOI: 10.1186/s13045-020-01002-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
To survive, cancer cells are subjected to various internal and external adverse factors, including genetic mutations, hypoxia, nutritional deficiencies, and drug toxicity. All of these factors result in the accumulation of unfolded proteins in the endoplasmic reticulum, which leads to a condition termed endoplasmic reticulum stress (ER stress) and triggers the unfolded protein response (UPR). UPR downstream components strictly control transcription and translation reprogramming to ensure selective gene expression, including that of non-coding RNA (ncRNAs), to adapt to adverse environments. NcRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play important roles in regulating target gene expression and protein translation, and their aberrant expression is related to tumor development. Dysregulation of ncRNAs is involved in the regulation of various cellular characteristics of cancer cells, including growth, apoptosis, metastasis, angiogenesis, drug sensitivity, and tumor stem cell properties. Notably, ncRNAs and ER stress can regulate each other and collaborate to determine the fate of tumor cells. Therefore, investigating the interaction between ER stress and ncRNAs is crucial for developing effective cancer treatment and prevention strategies. In this review, we summarize the ER stress-triggered UPR signaling pathways involved in carcinogenesis followed by the mutual regulation of ER stress and ncRNAs in cancer, which provide further insights into the understanding of tumorigenesis and therapeutic strategies.
Collapse
Affiliation(s)
- Tianming Zhao
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China
| | - Juan Du
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
280
|
Dolly A, Dumas J, Servais S. Cancer cachexia and skeletal muscle atrophy in clinical studies: what do we really know? J Cachexia Sarcopenia Muscle 2020; 11:1413-1428. [PMID: 33053604 PMCID: PMC7749617 DOI: 10.1002/jcsm.12633] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/24/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Research investigators have shown a growing interest in investigating alterations underlying skeletal muscle wasting in patients with cancer. However, skeletal muscle dysfunctions associated with cancer cachexia have mainly been studied in preclinical models. In the present review, we summarize the results of clinical studies in which skeletal muscle biopsies were collected from cachectic vs. non-cachectic cancer patients. Most of these studies suggest the presence of significant physiological alterations in skeletal muscle from cachectic cancer patients. We suggest a hypothesis, which connects structural and metabolic parameters that may, at least in part, be responsible for the skeletal muscle atrophy characteristic of cancer cachexia. Finally, we discuss the importance of a better standardization of the diagnostic criteria for cancer cachexia, as well as the requirement for additional clinical studies to improve the robustness of these conclusions.
Collapse
Affiliation(s)
- Adeline Dolly
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| | - Jean‐François Dumas
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| | - Stéphane Servais
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| |
Collapse
|
281
|
Zachariah M, Maamoun H, Milano L, Rayman MP, Meira LB, Agouni A. Endoplasmic reticulum stress and oxidative stress drive endothelial dysfunction induced by high selenium. J Cell Physiol 2020; 236:4348-4359. [PMID: 33241572 DOI: 10.1002/jcp.30175] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022]
Abstract
Selenium is an essential trace element important for human health. A balanced intake is, however, crucial to maximize the health benefits of selenium. At physiological concentrations, selenium mediates antioxidant, anti-inflammatory, and pro-survival actions. However, supra-nutritional selenium intake was associated with increased diabetes risk leading potentially to endothelial dysfunction, the initiating step in atherosclerosis. High selenium causes apoptosis in cancer cells via endoplasmic reticulum (ER) stress, a mechanism also implicated in endothelial dysfunction. Nonetheless, whether ER stress drives selenium-induced endothelial dysfunction, remains unknown. Here, we investigated the effects of increasing concentrations of selenium on endothelial cells. High selenite reduced nitric oxide bioavailability and impaired angiogenesis. High selenite also induced ER stress, increased reactive oxygen species (ROS) production, and apoptosis. Pretreatment with the chemical chaperone, 4-phenylbutyrate, prevented the toxic effects of selenium. Our findings support a model where high selenite leads to endothelial dysfunction through activation of ER stress and increased ROS production. These results highlight the importance of tailoring selenium supplementation to achieve maximal health benefits and suggest that prophylactic use of selenium supplements as antioxidants may entail risk.
Collapse
Affiliation(s)
- Matshediso Zachariah
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Hatem Maamoun
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Larissa Milano
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Margaret P Rayman
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Lisiane B Meira
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
282
|
Zhang J, Ye ZW, Chen W, Culpepper J, Jiang H, Ball LE, Mehrotra S, Blumental-Perry A, Tew KD, Townsend DM. Altered redox regulation and S-glutathionylation of BiP contribute to bortezomib resistance in multiple myeloma. Free Radic Biol Med 2020; 160:755-767. [PMID: 32937189 PMCID: PMC7704679 DOI: 10.1016/j.freeradbiomed.2020.09.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
Multiple myeloma (MM) cells have high rates of secretion of proteins rich in disulfide bonds and depend upon compartmentalized redox balance for accurate protein folding. The proteasome inhibitor bortezomib (Btz) is a successful frontline treatment for the disease, but its long-term efficacy is restricted by the acquisition of resistance. We found that MM cell lines resistant to Btz maintain high levels of oxidative stress and are cross resistant to endoplasmic reticulum (ER) stress-inducing agents thapsigargin (ThG), and tunicamycin (TuM). Moreover, cells expressing high/wild type levels of glutathione S-transferase P (GSTP) are more resistant than Gstp1/p2 knockout cells. In agreement, basal levels of S-glutathionylated proteins and redox regulation enzymes, including GSTP are elevated at mRNA and protein levels in resistant cells. GSTP mediated S-glutathionylation (SSG) regulates the activities of a number of redox active ER proteins. Here we demonstrated that the post-translational modification determines the balance between foldase and ATPase activities of the binding immunoglobulin protein (BiP), with Cys41-SSG important for ATPase, and Cys420-SSG for foldase. BiP expression and S-glutathionylation are increased in clinical specimens of bone marrow from MM patients compared to non-cancerous samples. Preventing S-glutathionylation in MM cells with a GSTP specific inhibitor restored BiP activities and reversed resistance to Btz. Therefore, S-glutathionylation of BiP confers pro-survival advantages and represents a novel mechanism of drug resistance in MM cells. We conclude that altered GSTP expression leads to S-glutathionylation of BiP, and contributes to acquired resistance to Btz in MM.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA.
| | - Zhi-Wei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Wei Chen
- Clinical Research Center, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, 1-1 Zhongfu Road, Nangjing, 21003, China
| | - John Culpepper
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Haiming Jiang
- Intensive Care Unit, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu Road, Muping District, Yantai City, Shandong, 264100, PR China
| | - Lauren E Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, 86 Jonathan Lucas Street, HCC512H, Charleston, SC, 29425, USA
| | - Anna Blumental-Perry
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, MSC 509/BSB 358, Charleston, SC, 29425, USA
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, 274 Calhoun Street, MSC 141, Charleston, SC, 29425, USA.
| |
Collapse
|
283
|
Grandjean JMD, Wiseman RL. Small molecule strategies to harness the unfolded protein response: where do we go from here? J Biol Chem 2020; 295:15692-15711. [PMID: 32887796 PMCID: PMC7667976 DOI: 10.1074/jbc.rev120.010218] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/02/2020] [Indexed: 12/31/2022] Open
Abstract
The unfolded protein response (UPR) plays a central role in regulating endoplasmic reticulum (ER) and global cellular physiology in response to pathologic ER stress. The UPR is comprised of three signaling pathways activated downstream of the ER membrane proteins IRE1, ATF6, and PERK. Once activated, these proteins initiate transcriptional and translational signaling that functions to alleviate ER stress, adapt cellular physiology, and dictate cell fate. Imbalances in UPR signaling are implicated in the pathogenesis of numerous, etiologically-diverse diseases, including many neurodegenerative diseases, protein misfolding diseases, diabetes, ischemic disorders, and cancer. This has led to significant interest in establishing pharmacologic strategies to selectively modulate IRE1, ATF6, or PERK signaling to both ameliorate pathologic imbalances in UPR signaling implicated in these different diseases and define the importance of the UPR in diverse cellular and organismal contexts. Recently, there has been significant progress in the identification and characterization of UPR modulating compounds, providing new opportunities to probe the pathologic and potentially therapeutic implications of UPR signaling in human disease. Here, we describe currently available UPR modulating compounds, specifically highlighting the strategies used for their discovery and specific advantages and disadvantages in their application for probing UPR function. Furthermore, we discuss lessons learned from the application of these compounds in cellular and in vivo models to identify favorable compound properties that can help drive the further translational development of selective UPR modulators for human disease.
Collapse
Affiliation(s)
- Julia M D Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA.
| |
Collapse
|
284
|
Abstract
Post-translational modifications of cellular substrates with ubiquitin and ubiquitin-like proteins (UBLs), including ubiquitin, SUMOs, and neural precursor cell-expressed developmentally downregulated protein 8, play a central role in regulating many aspects of cell biology. The UBL conjugation cascade is initiated by a family of ATP-dependent enzymes termed E1 activating enzymes and executed by the downstream E2-conjugating enzymes and E3 ligases. Despite their druggability and their key position at the apex of the cascade, pharmacologic modulation of E1s with potent and selective drugs has remained elusive until 2009. Among the eight E1 enzymes identified so far, those initiating ubiquitylation (UBA1), SUMOylation (SAE), and neddylation (NAE) are the most characterized and are implicated in various aspects of cancer biology. To date, over 40 inhibitors have been reported to target UBA1, SAE, and NAE, including the NAE inhibitor pevonedistat, evaluated in more than 30 clinical trials. In this Review, we discuss E1 enzymes, the rationale for their therapeutic targeting in cancer, and their different inhibitors, with emphasis on the pharmacologic properties of adenosine sulfamates and their unique mechanism of action, termed substrate-assisted inhibition. Moreover, we highlight other less-characterized E1s-UBA6, UBA7, UBA4, UBA5, and autophagy-related protein 7-and the opportunities for targeting these enzymes in cancer. SIGNIFICANCE STATEMENT: The clinical successes of proteasome inhibitors in cancer therapy and the emerging resistance to these agents have prompted the exploration of other signaling nodes in the ubiquitin-proteasome system including E1 enzymes. Therefore, it is crucial to understand the biology of different E1 enzymes, their roles in cancer, and how to translate this knowledge into novel therapeutic strategies with potential implications in cancer treatment.
Collapse
Affiliation(s)
- Samir H Barghout
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (S.H.B., A.D.S.); Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada (S.H.B., A.D.S.); and Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt (S.H.B.)
| | - Aaron D Schimmer
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada (S.H.B., A.D.S.); Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada (S.H.B., A.D.S.); and Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt (S.H.B.)
| |
Collapse
|
285
|
Zhou X, Zhang X, Wu Z, Xu X, Guo M, Zhai X, Zuo D, Wu Y. The novel ALK inhibitor ZX-29 induces apoptosis through inhibiting ALK and inducing ROS-mediated endoplasmic reticulum stress in Karpas299 cells. J Biochem Mol Toxicol 2020; 35:e22666. [PMID: 33140567 DOI: 10.1002/jbt.22666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 11/08/2022]
Abstract
It is a well-known fact that 60%-85% of anaplastic large cell lymphoma (ALCL) is mainly driven by the anaplastic lymphoma kinase (ALK) fusion protein. Although ALK-positive ALCL patients respond significantly to ALK inhibitors, the development of resistance is inevitable, which requires the development of new therapeutic strategies for ALK-positive ALCL. Here, we investigated the anticancer activities of N-(2((5-chloro-2-((2-methoxy-6-(4-methylpiperazin-1-yl)pyridin-3yl)amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (ZX-29), a newly synthesized ALK inhibitor, against nucleophosmin-ALK-positive cell line Karpas299. We demonstrated that ZX-29 decreased Karpas299 cells growth and had better cytotoxicity than ceritinib, which was mediated through downregulating the expression of ALK and related proteins, inducing cell cycle arrest, and promoting cell apoptosis. Moreover, ZX-29-induced cell apoptosis by inducing endoplasmic reticulum stress (ERS). In addition, ZX-29 increased the generation of reactive oxygen species (ROS), and cells pretreatment with N-acetyl- l-cysteine could attenuate ZX-29-induced cell apoptosis and ERS. Taken together, ZX-29 inhibited Karpas299 cell proliferation and induced apoptosis through inhibiting ALK and its downstream protein expression and inducing ROS-mediated ERS. Therefore, our results provide evidence for a novel antitumor candidate for the further investigation.
Collapse
Affiliation(s)
- Xuejiao Zhou
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaoning Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhuzhu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaobo Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ming Guo
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
286
|
Zhang LN, Xia YZ, Zhang C, Zhang H, Luo JG, Yang L, Kong LY. Vielanin K enhances doxorubicin-induced apoptosis via activation of IRE1α- TRAF2 - JNK pathway and increases mitochondrial Ca 2 + influx in MCF-7 and MCF-7/MDR cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153329. [PMID: 32896708 DOI: 10.1016/j.phymed.2020.153329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 07/05/2020] [Accepted: 08/09/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Therapeutic failure and drug resistance are common and have important implications in the poor prognosis of advanced breast cancer. It is necessary to acquire a natural product to overcome the resistance of cancer and increase the sensitivity of drug-resistant cells to anticancer agents. PURPOSE To demonstrate whether the compound Vielanin K (VK) has the potential to increase the sensitivity of MCF-7 and MCF-7/MDR cells to anticancer agents. METHODS Cell viability and proliferative capacity were determined by MTT, colony formation and EdU assays. Apoptosis and Ca2+ accumulation were evaluated by flow cytometry. Then, proteins were detected by immunoblotting, and gene expression levels were explored by qRT-PCR. RESULTS In MCF-7 and corresponding MDR cells, VK increased the fluorescence intensity of Rho123, but not CFDA. VK treatment did not affect the protein expression of P-gp, MRP1 or BCRP. VK treatment enhanced the DOX-induced apoptotic cascade, while VK combined with DOX increased JNK phosphorylation by activating the IRE1α-TRAF2 signaling pathway. In addition, Ca2+ was released from the endoplasmic reticulum following combination treatment, thereby giving rise to mitochondrial apoptosis. Silencing IRE1α and JNK with small interfering RNA (siRNA) efficiently attenuated combination treatment-induced apoptosis. These effects caused mitochondrial depolarization and reduced viability in MCF-7 and corresponding MCF-7/MDR cells. CONCLUSION VK combined with DOX increases the apoptosis of MCF-7 and corresponding MCF-7/MDR cells by activating ER stress and mitochondrial apoptosis via IRE1α-TRAF2-JNK signaling.
Collapse
Affiliation(s)
- Li-Na Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China
| | - Yuan-Zheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China
| | - Hao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China
| | - Jian-Guang Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China.
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nan Jing 210009, China.
| |
Collapse
|
287
|
Elfiky AA, Baghdady AM, Ali SA, Ahmed MI. GRP78 targeting: Hitting two birds with a stone. Life Sci 2020; 260:118317. [PMID: 32841659 PMCID: PMC7442953 DOI: 10.1016/j.lfs.2020.118317] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/22/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Glucose regulating protein 78 (GRP78) is one member of the Heat Shock Protein family of chaperone proteins (HSPA5) found in eukaryotes. It acts as the master of the Unfolded Protein Response (UPR) process in the lumen of the Endoplasmic Reticulum (ER). SCOPE Under the stress of unfolded proteins, GRP78 binds to the unfolded proteins to prevent misfolding, while under the load of the unfolded protein, it drives the cell to autophagy or apoptosis. Several attempts reported the overexpression of GRP78 on the cell membrane of cancer cells and cells infected with viruses or fungi. MAJOR CONCLUSIONS Cell-surface GRP78 is used as a cancer cell target in previous studies. Additionally, GRP78 is used as a drug target to stop the progression of cancer cells by different compounds, including peptides, antibodies, and some natural compounds. Additionally, it can be used as a protein target to reduce the infectivity of different viruses, including the pandemic SARS-CoV-2. Besides, GRP78 targeting is used in diagnosis and imaging modalities using radionuclides. GENERAL SIGNIFICANCE This review summarizes the various attempts that used GRP78 both in therapy (fighting cancer, viral and fungal infections) and diagnosis (imaging).
Collapse
|
288
|
Wu C, Chen W, Yu F, Yuan Y, Chen Y, Hurst DR, Li Y, Li L, Liu Z. Long Noncoding RNA HITTERS Protects Oral Squamous Cell Carcinoma Cells from Endoplasmic Reticulum Stress-Induced Apoptosis via Promoting MRE11-RAD50-NBS1 Complex Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002747. [PMID: 33240783 PMCID: PMC7675039 DOI: 10.1002/advs.202002747] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Indexed: 02/05/2023]
Abstract
Recent studies have proven that long noncoding RNAs (lncRNAs) exhibit regulatory functions of both DNA damage response (DDR) and endoplasmic reticulum (ER) stress. Herein, ER stress-induced lncRNA transcriptomic changes are reported in human oral squamous cell carcinoma (OSCC) cells and a novel lncRNA HITTERS ( H ERPUD1 intronic transcript of ER stress) is identified as the most significantly upregulated lncRNA. It is shown that HITTERS is a nucleus-located lncRNA including two transcript variants. HITTERS lacks an independent promoter but shares the same promoter with HERPUD1. HITTERS is transcriptionally regulated by Activating Transcription Factor (ATF) 6, ATF4, X-Box Binding Protein 1 (XBP1), and DNA methylation. In human OSCC tissues, HITTERS is significantly correlated with OSCC clinicopathological features and prognosis. Gain- and loss-of-function studies reveal that HITTERS promotes OSCC proliferation and invasion via influencing the expression of growth factor receptors and the downstream pathways. Once ER stress is triggered, HITTERS significantly attenuates ER stress-induced apoptosis both in vivo and in vitro. Mechanically, HITTERS functions as RNA scaffold to promote MRE11-RAD50-NBS1 complex formation in the repair of ER stress-induced DNA damage. To sum up, this study presents a novel lncRNA, namely HITTERS, which links ER stress and DDR together in OSCC.
Collapse
Affiliation(s)
- Chenzhou Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Wen Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of EndodonticsWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Yihang Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Yafei Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Douglas R. Hurst
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAL35294USA
| | - Yi Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| | - Zhe Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Head and Neck OncologyWest China Hospital of StomatologySichuan UniversityNumber 14, Unit 3, Renmin Nan RoadChengduSichuan610041China
| |
Collapse
|
289
|
Jiang Z, Zhang G, Huang L, Yuan Y, Wu C, Li Y. Transmissible Endoplasmic Reticulum Stress: A Novel Perspective on Tumor Immunity. Front Cell Dev Biol 2020; 8:846. [PMID: 33117793 PMCID: PMC7575690 DOI: 10.3389/fcell.2020.00846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/06/2020] [Indexed: 02/05/2023] Open
Abstract
As the first compartment of the protein secretory pathway, the endoplasmic reticulum (ER) acts as a protein synthesis factory, maintaining proteostasis and ER homeostasis. However, a variety of intrinsic and extrinsic perturbations, such as cancer, can disrupt the homeostasis and result in a large accumulation of misfolded/unfolded proteins in the ER lumen, thereby provoking a specific cellular state addressed as “ER stress”. Then the unfolded protein response (UPR), an adaptive signaling pathway, is triggered to address the stress and restore the homeostasis. A novel aspect of ER stress is that it can be transmitted from cancer cells to tumor-infiltrating myeloid cells through certain cancer cell-released soluble factors, which is termed as transmissible ER stress (TERS) or ER stress resonance (ERSR). In this review, we provide a comprehensive overview of the link between cancer and ER stress as well as the possible soluble factors mediating TERS. We further elaborate the cell-extrinsic effects of TERS on tumor immunity, and how it indirectly modulates cancer development and progression, which is expected to add a new dimension to anticancer therapy.
Collapse
Affiliation(s)
- Zhou Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Geru Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yihang Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
290
|
Geldanamycin-Derived HSP90 Inhibitors Are Synthetic Lethal with NRF2. Mol Cell Biol 2020; 40:MCB.00377-20. [PMID: 32868290 DOI: 10.1128/mcb.00377-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 08/24/2020] [Indexed: 12/30/2022] Open
Abstract
Activating mutations in KEAP1-NRF2 are frequently found in tumors of the lung, esophagus, and liver, where they are associated with aggressive growth, resistance to cancer therapies, and low overall survival. Despite the fact that NRF2 is a validated driver of tumorigenesis and chemotherapeutic resistance, there are currently no approved drugs which can inhibit its activity. Therefore, there is an urgent clinical need to identify NRF2-selective cancer therapies. To this end, we developed a novel synthetic lethal assay, based on fluorescently labeled isogenic wild-type and Keap1 knockout cell lines, in order to screen for compounds which selectively kill cells in an NRF2-dependent manner. Through this approach, we identified three compounds based on the geldanamycin scaffold which display synthetic lethality with NRF2. Mechanistically, we show that products of NRF2 target genes metabolize the quinone-containing geldanamycin compounds into more potent HSP90 inhibitors, which enhances their cytotoxicity while simultaneously restricting the synthetic lethal effect to cells with aberrant NRF2 activity. As all three of the geldanamycin-derived compounds have been used in clinical trials, they represent ideal candidates for drug repositioning to target the currently untreatable NRF2 activity in cancer.
Collapse
|
291
|
Nomura Y, Sylvester CF, Nguyen LO, Kandeel M, Hirata Y, Mungrue IN, Oh-Hashi K. Characterization of the 5'-flanking region of the human and mouse CHAC1 genes. Biochem Biophys Rep 2020; 24:100834. [PMID: 33102815 PMCID: PMC7573368 DOI: 10.1016/j.bbrep.2020.100834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/09/2020] [Indexed: 11/29/2022] Open
Abstract
The Unfolded Protein Response pathway is a conserved signaling mechanism having important roles in cellular physiology and is perturbed accompanying disease. We previously identified the novel UPR target gene CHAC1, a direct target of ATF4, downstream of PERK-EIF2A and activated by the UPR pathway. CHAC1 enzyme directs catalysis of γ-linked glutamate bonds within specific molecular targets. CHAC1 is the first enzyme characterized that can catalyze intracellular glutathione degradation in eukaryotes, having implications for regulation of oxidative stress. DDIT3 (CHOP) is a terminal UPR transcription factor, regulated by ATF4 and an output promoting cell death signaling. Herein we examine the relationship of CHOP controlling CHAC1 transcription in humans and mice. We note parallel induction of CHOP and CHAC1 in human cells after agonist induced UPR. Expanding upon previous reports, we define transcriptional induction of CHAC1 in humans and mice driven by ATF4 through a synergistic relationship with conserved ATF/CRE and CARE DNA sequences of the CHAC1 promoter. Using this system, we also tested effects of CHOP on CHAC1 transcription, and binding at the CHAC1 ATF/CRE using IM-EMSA. These data indicate a novel inhibitory effect of CHOP on CHAC1 transcription, which was ablated in the absence of the ATF/CRE control element. While direct binding of ATF4 to CHAC1 promoter sequences was confirmed, binding of CHOP to the CHAC1 ATF/CRE was not evident at baseline or after UPR induction. These data reveal CHAC1 as a novel CHOP inhibited target gene, acting through an upstream ATF/CRE motif via an indirect mechanism.
Collapse
Affiliation(s)
- Yuki Nomura
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Charity F Sylvester
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, 1901, Perdido St, New Orleans, LA, USA
| | - Lisa O Nguyen
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, 1901, Perdido St, New Orleans, LA, USA
| | - Mahmoud Kandeel
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, King Faisal University, Hofuf, Alahsa, 31982, Saudi Arabia.,Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Yoko Hirata
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.,Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Imran N Mungrue
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, 1901, Perdido St, New Orleans, LA, USA
| | - Kentaro Oh-Hashi
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.,Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
292
|
Cui L, Li C, Chen B, Huang H, Xia Q, Li X, Shen Z, Ge Z, Wang Y. Surface functionalized red fluorescent dual-metallic Au/Ag nanoclusters for endoplasmic reticulum imaging. Mikrochim Acta 2020; 187:606. [PMID: 33052480 DOI: 10.1007/s00604-020-04585-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/01/2020] [Indexed: 11/30/2022]
Abstract
An efficient method is reported to prepare endoplasmic reticulum-targetable dual-metallic gold-silver nanoclusters, denoted as ER-Au/Ag nanoclusters (NCs), by virtue of a rationally designed molecular ligand. The prepared ER-Au/Ag NCs possesses red-emitting fluorescence with a strong emission at 622 nm and a high fluorescence quantum yield of 5.1%, which could avoid the influence of biological auto-fluorescence. Further investigation results showed that ER-Au/Ag NCs exhibited superior photostability, minimal cytotoxicity, and ER-targeting capability. Enabled by these meritorious features, ER-Au/Ag NCs have been successfully employed for long-term bioimaging of ER in living cells.Graphical abstract A sensitive non-enzymatic fluorescent glucose probe-based ZnO nanorod decorated with Au nanoparticles.
Collapse
Affiliation(s)
- Lifeng Cui
- Department of Environmental Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Chengyun Li
- Department of Environmental Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Biyun Chen
- Nanhu College, Jiaxing University, Jiaxing, 314001, China
| | - Hong Huang
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China.
| | - Qineng Xia
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Xi Li
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Zhangfeng Shen
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Zhigang Ge
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Yangang Wang
- College of Biological, Chemical Science and Engineering, Jiaxing University, Jiaxing, 314001, China.
| |
Collapse
|
293
|
Pandey S, Nandi A, Basu S, Ballav N. Inducing endoplasmic reticulum stress in cancer cells using graphene oxide-based nanoparticles. NANOSCALE ADVANCES 2020; 2:4887-4894. [PMID: 36132889 PMCID: PMC9417581 DOI: 10.1039/d0na00338g] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/17/2020] [Indexed: 06/10/2023]
Abstract
The endoplasmic reticulum is one of the vital organelles primarily involved in protein synthesis, folding, and transport and lipid biosynthesis. However, in cancer cells its functions are dysregulated leading to ER stress. ER stress is now found to be closely associated with hallmarks of cancer and has subsequently emerged as an alluring target in cancer therapy. However, specific targeting of the ER in a cancer cell milieu remains a challenge. To address this, in this report we have engineered ER-targeted self-assembled 3D spherical graphene oxide nanoparticles (ER-GO-NPs) encompassing dual ER stress inducers, doxorubicin and cisplatin. DLS, FESEM and AFM techniques revealed that the nanoparticles were spherical in shape with a sub 200 nm diameter. Confocal microscopy confirmed the specific homing of these ER-GO-NPs into the subcellular ER within 3 h. A combination of gel electrophoresis, confocal microscopy and flow cytometry studies revealed that these ER-GO-NPs induced ER stress mediated apoptosis in HeLa cells. Interestingly, the nanoparticles also activated autophagy which was inhibited through the cocktail treatment with ER-GO-NPs and chloroquine (CQ). At the same time these ER-GO-NPs were found to be efficient in prompting ER stress associated apoptosis in breast, lung and drug resistant triple negative breast cancer cell lines as well. We envision that these ER specific self-assembled graphene oxide nanoparticles can serve as a platform to exploit ER stress and its associated unfolded protein response (UPR) as a target resulting in promising therapeutic outcomes in cancer therapy.
Collapse
Affiliation(s)
- Shalini Pandey
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr Homi Bhabha Road, Pashan Pune Maharashtra 411008 India
| | - Aditi Nandi
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr Homi Bhabha Road, Pashan Pune Maharashtra 411008 India
| | - Sudipta Basu
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| | - Nirmalya Ballav
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr Homi Bhabha Road, Pashan Pune Maharashtra 411008 India
| |
Collapse
|
294
|
Choudhury A, Renjilian E, Asan O. Use of machine learning in geriatric clinical care for chronic diseases: a systematic literature review. JAMIA Open 2020; 3:459-471. [PMID: 33215079 PMCID: PMC7660963 DOI: 10.1093/jamiaopen/ooaa034] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/26/2020] [Accepted: 07/11/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives Geriatric clinical care is a multidisciplinary assessment designed to evaluate older patients’ (age 65 years and above) functional ability, physical health, and cognitive well-being. The majority of these patients suffer from multiple chronic conditions and require special attention. Recently, hospitals utilize various artificial intelligence (AI) systems to improve care for elderly patients. The purpose of this systematic literature review is to understand the current use of AI systems, particularly machine learning (ML), in geriatric clinical care for chronic diseases. Materials and Methods We restricted our search to eight databases, namely PubMed, WorldCat, MEDLINE, ProQuest, ScienceDirect, SpringerLink, Wiley, and ERIC, to analyze research articles published in English between January 2010 and June 2019. We focused on studies that used ML algorithms in the care of geriatrics patients with chronic conditions. Results We identified 35 eligible studies and classified in three groups: psychological disorder (n = 22), eye diseases (n = 6), and others (n = 7). This review identified the lack of standardized ML evaluation metrics and the need for data governance specific to health care applications. Conclusion More studies and ML standardization tailored to health care applications are required to confirm whether ML could aid in improving geriatric clinical care.
Collapse
Affiliation(s)
- Avishek Choudhury
- School of Systems and Enterprises, Stevens Institute of Technology, Hoboken, New Jersey, USA
| | - Emily Renjilian
- School of Systems and Enterprises, Stevens Institute of Technology, Hoboken, New Jersey, USA
| | - Onur Asan
- School of Systems and Enterprises, Stevens Institute of Technology, Hoboken, New Jersey, USA
| |
Collapse
|
295
|
Milan M, Balestrieri C, Alfarano G, Polletti S, Prosperini E, Nicoli P, Spaggiari P, Zerbi A, Cirulli V, Diaferia GR, Natoli G. Pancreatic Cancer Cells Require the Transcription Factor MYRF to Maintain ER Homeostasis. Dev Cell 2020; 55:398-412.e7. [PMID: 32997974 DOI: 10.1016/j.devcel.2020.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 01/01/2023]
Abstract
Many tumors of endodermal origin are composed of highly secretory cancer cells that must adapt endoplasmic reticulum (ER) activity to enable proper folding of secreted proteins and prevent ER stress. We found that pancreatic ductal adenocarcinomas (PDACs) overexpress the myelin regulatory factor (MYRF), an ER membrane-associated transcription factor (TF) released by self-cleavage. MYRF was expressed in the well-differentiated secretory cancer cells, but not in the poorly differentiated quasi-mesenchymal cells that coexist in the same tumor. MYRF expression was controlled by the epithelial identity TF HNF1B, and it acted to fine-tune the expression of genes encoding highly glycosylated, cysteine-rich secretory proteins, thus preventing ER overload. MYRF-deficient PDAC cells showed signs of ER stress, impaired proliferation, and an inability to form spheroids in vitro, while in vivo they generated highly secretory but poorly proliferating and hypocellular tumors. These data indicate a role of MYRF in the control of ER homeostasis in highly secretory PDAC cells.
Collapse
Affiliation(s)
- Marta Milan
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Chiara Balestrieri
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Gabriele Alfarano
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Sara Polletti
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Elena Prosperini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Paola Nicoli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Paola Spaggiari
- Humanitas Clinical Research Center IRCCS, Rozzano, Milano, Italy
| | - Alessandro Zerbi
- Humanitas Clinical Research Center IRCCS, Rozzano, Milano, Italy; Humanitas University, Pieve Emanuele, Milano, Italy
| | - Vincenzo Cirulli
- Departments of Medicine and Pharmacology, Institute for Stem Cell and Regenerative Medicine, UW Diabetes Institute, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Giuseppe R Diaferia
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy.
| | - Gioacchino Natoli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy; Humanitas University, Pieve Emanuele, Milano, Italy.
| |
Collapse
|
296
|
Wang X, Bai Y, Zhang F, Yang Y, Feng D, Li A, Yang Z, Li D, Tang Y, Wei X, Wei W, Han P. Targeted Inhibition of P4HB Promotes Cell Sensitivity to Gemcitabine in Urothelial Carcinoma of the Bladder. Onco Targets Ther 2020; 13:9543-9558. [PMID: 33061438 PMCID: PMC7532080 DOI: 10.2147/ott.s267734] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/15/2020] [Indexed: 02/05/2023] Open
Abstract
Background Bladder cancer (BC) is a common malignancy worldwide that accounts for 3% of global cancer diagnoses. Chemotherapy resistance limits the therapeutic effect of chemotherapeutic agents in patients with BC. Prolyl 4-hydroxylase, beta polypeptide (P4HB) is an endoplasmic reticulum (ER) chaperone that is upregulated in bladder cancer tissues (The Cancer Genome Atlas, TCGA datasets). Knockdown or suppression of P4HB exerts anticancer activity and sensitizes cells to chemotherapy in various types of cancer. Purpose We aimed to investigate whether the inhibition of P4HB enhances the anticancer efficacy of gemcitabine (GEM) in BC cells and to study the underlying molecular mechanisms. Patients and Methods The P4HB mRNA expression levels of 411 BC patients from the TCGA database and P4HB expression level of eighty BC paraffin-embedded samples detected by immunohistochemistry (IHC) staining were used for clinical feature and prognostic analyses. Bioinformatics analysis was utilized for the mechanistic investigation. Highly P4HB-expressed BC cell lines (T24 and 5637) treated with P4HB inhibitor (Bacitracin, BAC) were used to study the effects of BAC on the sensitivity of BC cells to GEM and the potential mechanism. P4HB inhibition experiments were performed in highly P4HB-expressed BC cells, and cell viability, colony formation, cell cycle, reactive oxygen species (ROS), apoptosis and pathway proteins were assessed in T24 and 5637 cells. Results Western blot analysis showed that P4HB expression was significantly higher in BC tissues than in paired normal tissues. IHC showed that patients with high P4HB expression had a poorer overall survival (OS) rate than those with low P4HB expression. Furthermore, increased P4HB expression was demonstrated to be an independent prognostic marker for BC. Functionally, P4HB inhibition by BAC decreased the cell proliferation ability in vitro. Moreover, BAC treatment sensitized BC cells to GEM. Molecular mechanism analysis indicated that inhibition of P4HB by BAC treatment enhanced the anticancer effects of GEM through increasing cellular ROS content and promoting cell apoptosis and PERK/eIF2α/ATF4/CHOP signaling. Conclusion High P4HB expression was significantly correlated with poor prognosis in BC patients. Inhibition of P4HB by BAC decreased the cell proliferation ability and sensitized BC cells to GEM by activating apoptosis and the PERK/eIF2α/ATF4/CHOP pathways.
Collapse
Affiliation(s)
- Xiaoming Wang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Yunjin Bai
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Facai Zhang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Yubo Yang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Dechao Feng
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Ao Li
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Zhiqiang Yang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Dengxiong Li
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Yin Tang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Xin Wei
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Wuran Wei
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| | - Ping Han
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People's Republic of China
| |
Collapse
|
297
|
Shen T, Li Y, Liang S, Chen Z. XBP1 negatively regulates CENPF expression via recruiting ATF6α to the promoter during ER stress. Cancer Cell Int 2020; 20:459. [PMID: 32973403 PMCID: PMC7507253 DOI: 10.1186/s12935-020-01553-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/11/2020] [Indexed: 12/20/2022] Open
Abstract
Background Centromere protein F (CENPF) is a key component of the kinetochore complex involved in mitosis, cell differentiation and cellular response to stresses. However, the alteration of CENPF in response to endoplasmic reticulum (ER) stress has not been well described. In the present study, we investigate CENPF regulation in response to ER stress. Methods Quantitative real-time polymerase chain reaction and western blotting were used to determine CENPF expression under ER stress. Luciferase activity analysis was performed to investigate the promoter regions contributing to CENPF transcription in response to TG. Chromatin immunoprecipitation (ChIP) and ChIP Re-IP assays were used to determine if X-box binding protein 1 (XBP1) and/or activating transcription factor 6α (ATF6α) bind in the CENPF promoter region. Cell apoptosis and proliferation were analyzed using TUNEL, cell growth and clonogenic assays. Results CENPF expression is dramatically reduced under ER stress induced by thapsigargin (TG), brefeldin A (BFA), or tunicamycin (TM) and this downregulation of CENPF expression was dependent on XBP1 and ATF6α. Luciferase activity analysis of the truncated CENPF promoter indicates that regions from bases - 679 to - 488 and from - 241 to - 78 in the CENPF promoter were sensitive to TG treatment. Additionally, ChIP and ChIP Re-IP assays reveal that XBP1 and ATF6α were assembled on the same regions of CENPF promoter. Notably, we identify two XBP1 binding sequences at positions - 567 and - 192, to which XBP1 binding was enhanced by TG. Finally, CENPF overexpression inhibits cell apoptosis and promotes cell proliferation in response to ER stress. Conclusion In summary, these results demonstrate that ER stress plays a crucial role in CENPF expression, and XBP1 may up-regulate DNA-binding affinities after TG treatment to the promoter of CENPF. These findings may contribute to the understanding of the molecular mechanism of CENPF regulation.
Collapse
Affiliation(s)
- Tao Shen
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 People's Republic of China
| | - Yan Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122 People's Republic of China.,Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 USA
| | - Shuang Liang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455 USA
| | - Zhiguang Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, 110004 People's Republic of China
| |
Collapse
|
298
|
RSK2 protects human breast cancer cells under endoplasmic reticulum stress through activating AMPKα2-mediated autophagy. Oncogene 2020; 39:6704-6718. [PMID: 32958832 DOI: 10.1038/s41388-020-01447-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 01/07/2023]
Abstract
Autophagy can protect stressed cancer cell by degradation of damaged proteins and organelles. However, the regulatory mechanisms behind this cellular process remain incompletely understood. Here, we demonstrate that RSK2 (p90 ribosomal S6 kinase 2) plays a critical role in ER stress-induced autophagy in breast cancer cells. We demonstrated that the promotive effect of RSK2 on autophagy resulted from directly binding of AMPKα2 in nucleus and phosphorylating it at Thr172 residue. IRE1α, an ER membrane-associated protein mediating unfolded protein response (UPR), is required for transducing the signal for activation of ERK1/2-RSK2 under ER stress. Suppression of autophagy by knockdown of RSK2 enhanced the sensitivity of breast cancer cells to ER stress both in vitro and in vivo. Furthermore, we demonstrated that inhibition of RSK2-mediated autophagy rendered breast cancer cells more sensitive to paclitaxel, a chemotherapeutic agent that induces ER stress-mediated cell death. This study identifies RSK2 as a novel controller of autophagy in tumor cells and suggests that targeting RSK2 can be exploited as an approach to reinforce the efficacy of ER stress-inducing agents against cancer.
Collapse
|
299
|
Kranz P, Sänger C, Wolf A, Baumann J, Metzen E, Baumann M, Göpelt K, Brockmeier U. Tumor cells rely on the thiol oxidoreductase PDI for PERK signaling in order to survive ER stress. Sci Rep 2020; 10:15299. [PMID: 32943707 PMCID: PMC7499200 DOI: 10.1038/s41598-020-72259-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Upon ER stress cells activate the unfolded protein response through PERK, IRE1 and ATF6. Remarkable effort has been made to delineate the downstream signaling of these three ER stress sensors after activation, but upstream regulation at the ER luminal site still remains mostly undefined. Here we report that the thiol oxidoreductase PDI is mandatory for activation of the PERK pathway in HEK293T as well as in human pancreatic, lung and colon cancer cells. Under ER stress, depletion of PDI selectively abrogated eIF2α phosphorylation, induction of ATF4, CHOP and even BiP. Furthermore, we could demonstrate that PDI prevented degradation of activated PERK by the 26S proteasome and therefore contributes to maintained PERK signaling. As a result of decreased PERK activity, PDI depleted cells showed an increased vulnerability to ER stress induced by chemicals or ionizing radiation in 2D as well as in 3D culture models. We conclude that PDI is an obligatory regulator of the PERK pathway with future therapy implications.
Collapse
Affiliation(s)
- Philip Kranz
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany
| | | | - Alexandra Wolf
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany
| | - Jennifer Baumann
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany
| | - Eric Metzen
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany
| | - Melanie Baumann
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany
| | - Kirsten Göpelt
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany
| | - Ulf Brockmeier
- Institut für Physiologie, Universität Duisburg-Essen, Duisburg, Germany. .,Department of Neurology, University Hospital Essen, University Duisburg-Essen, Duisburg, Germany.
| |
Collapse
|
300
|
Xin W, Zhang M, Yu Y, Li S, Ma C, Zhang J, Jiang Y, Li Y, Zheng X, Zhang L, Zhao X, Pei X, Zhu D. BCAT1 binds the RNA-binding protein ZNF423 to activate autophagy via the IRE1-XBP-1-RIDD axis in hypoxic PASMCs. Cell Death Dis 2020; 11:764. [PMID: 32938905 PMCID: PMC7494854 DOI: 10.1038/s41419-020-02930-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/24/2022]
Abstract
Abnormal functional changes in pulmonary artery smooth muscle cells are the main causes of many lung diseases. Among, autophagy plays a crucial role. However, the specific molecular regulatory mechanism of autophagy in PASMCs remains unclear. Here, we first demonstrate that BCAT1 played a key role in the autophagy of hypoxic PASMCs and hypoxic model rats. BCAT1-induced activation and accumulation of the autophagy signaling proteins BECN1 and Atg5 by the endoplasmic reticulum (ER) stress pathway. Interestingly, we discovered that BCAT1 bound IRE1 on the ER to activate expression of its downstream pathway XBP-1-RIDD axis to activate autophagy. More importantly, we identified an RNA-binding protein, zinc finger protein 423, which promoted autophagy by binding adenylate/uridylate (AU)-rich elements in the BCAT1 mRNA 3′-untranslated region. Overall, our results identify BCAT1 as a potential therapeutic target for the clinical treatment of lung diseases and reveal a novel posttranscriptional regulatory mechanism and signaling pathway in hypoxia-induced PASMC autophagy.
Collapse
Affiliation(s)
- Wei Xin
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Min Zhang
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China.,Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China
| | - Yang Yu
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Songlin Li
- College of Pharmacy, Harbin University of Commerce, Harbin, 150076, P.R. China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Junting Zhang
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Yuan Jiang
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Yiying Li
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Xiaodong Zheng
- Department of Genetic and Cell Biology, Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Xijuan Zhao
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Xuzhong Pei
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China.,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China
| | - Daling Zhu
- College of Pharmacy, Harbin Medical University, Harbin, 150081, P.R. China. .,Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, P.R. China. .,State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Daqing, 163319, P.R. China. .,Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin, 150081, P.R. China.
| |
Collapse
|