1
|
Vo VTA, Tran LN, Bui TT, Lee HW, Jeong Y. Etoposide-induced protein 2.4 homolog promotes argininosuccinate synthase 1 and cancer cell survival upon arginine deprivation. Cell Mol Biol Lett 2025; 30:52. [PMID: 40253325 PMCID: PMC12008907 DOI: 10.1186/s11658-025-00726-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/03/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Arginine auxotrophy has been reported in a subset of cancers with inherently defective de novo arginine synthesis. However, the use of arginine deprivation therapy seems to be unequally effective, partially owing to the resistance acquired by cancer cells. Study of underlying factors involved in this response thus becomes of utmost importance. Meanwhile, the function of etoposide-induced 2.4 homolog (EI24) in cancer metabolism, and specifically in arginine metabolism, remains unknown. METHODS EI24 was overexpressed in cancer cells using a doxycycline-inducible system or adenovirus transduction, while siRNA was used to knockdown EI24. Amino acid(s) deprivation medium was exploited with a cell viability assay to check the reliance of cancer cell survival on arginine. Protein expression and activation were examined through western blot and co-immunoprecipitation blot. Furthermore, global and specific protein translation were assessed through the SUnSET assay and polysome fractionation analysis. Gene expression and arginine level were downloaded from public cancer datasets for in silico validation including gene set enrichment and survival analysis to objectively evaluate the association between EI24 and arginine metabolism. RESULTS EI24 promoted cancer survival under arginine starvation. Mechanistically, EI24 replenished translation of argininosuccinate synthase 1 (ASS1) by inducing the inactive S-nitrosylated form of phosphatase and tensin homolog (PTEN), leading to release of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) axis. This tumor-promoting action of EI24 could be found in multiple ASS1-deficient cancer cells regardless of p53 status. Furthermore, expression of EI24 was linked to enrichment of arginine metabolism pathway as well as poor survival of patients with cancer across various cancer types, suggesting its role in cancer resistance to arginine deprivation. CONCLUSIONS This study is the first to report the role of EI24 in promoting cancer survival via translational regulation of the metabolic enzyme ASS1, thus paving a route for further investigation into the link between EI24 and cancer metabolism.
Collapse
Affiliation(s)
- Vu T A Vo
- Department of Biochemistry, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
- Organelle Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Le Nhat Tran
- Department of Biochemistry, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
- Organelle Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Thu Thanh Bui
- Department of Biochemistry, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
- Organelle Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yangsik Jeong
- Department of Biochemistry, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea.
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea.
- Organelle Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea.
- Institute of Mitochondrial Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea.
- ONCOin, Ltd., Startup cube #2 - 204, 1 Kangwondaehakgil, Chuncheon, Republic of Korea.
| |
Collapse
|
2
|
Wang M, Zheng H, Chen J, Tang Y, Feng M, Li L. ZnO nanoparticles impair autophagic flux and cell viability through the TRIM16-NRF2-p62 pathway in inflammatory keratinocytes. Food Chem Toxicol 2023; 182:114177. [PMID: 37977258 DOI: 10.1016/j.fct.2023.114177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/08/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Zinc oxide nanoparticles (ZnO NPs) are widely used in sunscreen, cosmetics, and topical drugs. Most previous studies have confirmed the safety of ZnO NPs applied to normal skin; however, little is known about the safety and potential toxicity of ZnO NPs applied to inflamed skin. This study aimed to evaluate the exposure risk of ZnO NPs in the treatment of inflammatory skin diseases. METHODS Normal human and tumor necrosis factor-α (TNF-α)-induced inflammatory keratinocytes were incubated with ZnO NPs to assess their toxic effects on cell viability and autophagy signaling pathway. Tandem mass tag (TMT)-based proteomics analysis was used to identify differentially expressed proteins following incubation of inflammatory keratinocytes with ZnO NPs. Protein expression was assessed by Western blot, and double fluorescent labeling and siRNA-knockdown further elucidated the role of the TRIM16-NRF2-p62 pathway in mediating the effects of ZnO NP. RESULTS In TNF-α-induced inflammatory keratinocytes, ZnO NPs activated cytoprotective autophagy and mediated p62-related autophagic flux block, thereby reducing the viability of inflammatory keratinocytes. Additionally, TRIM16-NRF2 was essential in ZnO NP-mediated autophagy flux block and cell viability reduction in inflammatory keratinocytes. Inhibition of the TRIM16-NRF2 pathway reduced p62 levels, alleviated autophagy flux blockade, and slightly restored the viability of inflammatory keratinocytes. CONCLUSION ZnO NPs activated protective cell autophagy. Blockade of autophagy flux mediated by the TRIM16-NRF2-p62 pathway led to decreased cell viability. This study provided a deeper understanding of the toxicity mechanism of ZnO NPs in inflammatory keratinocytes.
Collapse
Affiliation(s)
- Menglei Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, China
| | - Huanxin Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, China
| | - Jiawen Chen
- Department of Dermatology, Nanfang Hospital, Southern Medical University, China
| | - Yingmei Tang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, China
| | - Meixin Feng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, China
| | - Li Li
- Department of Dermatology, Nanfang Hospital, Southern Medical University, China.
| |
Collapse
|
3
|
Visintin R, Ray SK. Intersections of Ubiquitin-Proteosome System and Autophagy in Promoting Growth of Glioblastoma Multiforme: Challenges and Opportunities. Cells 2022; 11:cells11244063. [PMID: 36552827 PMCID: PMC9776575 DOI: 10.3390/cells11244063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a brain tumor notorious for its propensity to recur after the standard treatments of surgical resection, ionizing radiation (IR), and temozolomide (TMZ). Combined with the acquired resistance to standard treatments and recurrence, GBM is an especially deadly malignancy with hardly any worthwhile treatment options. The treatment resistance of GBM is influenced, in large part, by the contributions from two main degradative pathways in eukaryotic cells: ubiquitin-proteasome system (UPS) and autophagy. These two systems influence GBM cell survival by removing and recycling cellular components that have been damaged by treatments, as well as by modulating metabolism and selective degradation of components of cell survival or cell death pathways. There has recently been a large amount of interest in potential cancer therapies involving modulation of UPS or autophagy pathways. There is significant crosstalk between the two systems that pose therapeutic challenges, including utilization of ubiquitin signaling, the degradation of components of one system by the other, and compensatory activation of autophagy in the case of proteasome inhibition for GBM cell survival and proliferation. There are several important regulatory nodes which have functions affecting both systems. There are various molecular components at the intersections of UPS and autophagy pathways that pose challenges but also show some new therapeutic opportunities for GBM. This review article aims to provide an overview of the recent advancements in research regarding the intersections of UPS and autophagy with relevance to finding novel GBM treatment opportunities, especially for combating GBM treatment resistance.
Collapse
Affiliation(s)
- Rhett Visintin
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
- Correspondence: ; Tel.: +1-803-216-3420; Fax: +1-803-216-3428
| |
Collapse
|
4
|
Shin WH, Park JH, Chung KC. The central regulator p62 between ubiquitin proteasome system and autophagy and its role in the mitophagy and Parkinson's disease. BMB Rep 2020. [PMID: 31818366 PMCID: PMC6999829 DOI: 10.5483/bmbrep.2020.53.1.283] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) and autophagy are two major degradative pathways of proteins in eukaryotic cells. As about 30% of newly synthesized proteins are known to be misfolded under normal cell conditions, the precise and timely operation of the UPS and autophagy to remove them as well as their tightly controlled regulation, is so important for proper cell function and survival. In the UPS, target proteins are labeled by small proteins called ubiquitin, which are then transported to the proteasome complex for degradation. Alternatively, many greatly damaged proteins are believed to be delivered to the lysosome for autophagic degradation. Although these autophagy and UPS pathways have not been considered to be directly related, many recent studies proposed their close link and dynamic interconversion. In this review, we’ll focus on the several regulatory molecules that function in both UPS and autophagy and their crosstalk. Among the proposed multiple modulators, we will take a closer look at the so-called main connector of UPS-autophagy regulation, p62. Last, the functional role of p62 in the mitophagy and its implication for the pathogenesis of Parkinson’s disease, one of the major neurodegenerative diseases, will be briefly reviewed.
Collapse
Affiliation(s)
- Woo Hyun Shin
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Joon Hyung Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
5
|
Hwang M, Jun DW, Kang EH, Yoon KA, Cheong H, Kim YH, Lee CH, Kim S. EI24, as a Component of Autophagy, Is Involved in Pancreatic Cell Proliferation. Front Oncol 2019; 9:652. [PMID: 31396480 PMCID: PMC6664870 DOI: 10.3389/fonc.2019.00652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/03/2019] [Indexed: 01/29/2023] Open
Abstract
Autophagy is a highly conserved cellular process in which cytoplasmic materials are degraded and recycled as energy sources when nutrient supplies are lacking. Established tumor cells require autophagy for cell growth and tumor promotion. In particular, the survival of pancreatic tumor cells appears to be strongly dependent on autophagy, referred to as autophagy addiction. This dependency of pancreatic tumor cells on autophagy may be a candidate target for pancreatic tumor therapy. EI24 (etoposide-induced gene 2.4 kb; PIG8, p53-induced gene 8) acts as a tumor suppressor, inhibiting cell growth and inducing apoptosis in breast, cervical, and prostate cancer cells. However, recent papers have reported that EI24 is an essential component of the autophagy pathway. This newly discovered role of EI24 as a component of autophagy may act as a tumor promoter, which is contradictory to its known role as a tumor suppressor. We investigated the role of EI24 as a component of autophagy in pancreatic tumor cell proliferation. Here, we demonstrated that knockdown of EI24 using siRNA in pancreatic tumor cells led to impaired autophagy at a late step (increase in LC3-II and accumulation of p62 and autolysosomes). EI24 deficiency in pancreatic tumor cell lines inhibited cell proliferation. We confirmed that loss of EI24 inhibited pancreatic cell proliferation using the CRISPR-Cas9 system. However, loss of EI24 in other cell lines did not affect cell proliferation. Taken together, our results suggest that EI24 acts as a tumor promoter in pancreatic tumor cells, and studying the role of EI24 in reference to its cellular context may lead to a useful therapeutic target.
Collapse
Affiliation(s)
- Mihwa Hwang
- National Cancer Center, Research Institute, Goyang-si, South Korea
| | - Dong Wha Jun
- National Cancer Center, Research Institute, Goyang-si, South Korea
| | - Eun Hye Kang
- National Cancer Center, Research Institute, Goyang-si, South Korea
| | - Kyong-Ah Yoon
- College of Veterinary Medicine, Konkuk University, Seoul, South Korea
| | - Heesun Cheong
- National Cancer Center, Research Institute, Goyang-si, South Korea
| | - Yun-Hee Kim
- National Cancer Center, Research Institute, Goyang-si, South Korea
| | - Chang-Hun Lee
- National Cancer Center, Research Institute, Goyang-si, South Korea
| | - Sunshin Kim
- National Cancer Center, Research Institute, Goyang-si, South Korea
| |
Collapse
|
6
|
Nam TW, Park SY, Lee JH, Roh JI, Lee HW. Effect of EI24 expression on the tumorigenesis of Apc Min/+ colorectal cancer mouse model. Biochem Biophys Res Commun 2019; 514:1087-1092. [PMID: 31097220 DOI: 10.1016/j.bbrc.2019.04.186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/27/2019] [Indexed: 12/14/2022]
Abstract
Etoposide-induced 2.4 kb transcript (EI24, also known as PIG8) is a p53 target gene involved in cell growth suppression and apoptosis and known to be frequently altered in human cancers. Although EI24 expression is decreased in various cancers and is associated with colorectal cancer progression and metastasis, the physiological function of EI24 in colorectal cancer is yet unclear. We generated an Ei24 conditional transgenic (Tg) mouse to study the therapeutic effects of Ei24 in vivo and evaluated whether Ei24 plays a role of a tumor suppressor using Ei24 Tg mouse crossed with ApcMin/+ mouse, which develops multiple intestinal adenomas. The overexpression of Ei24 failed to cause any notable difference in the number of polyps, lengths of the intestine and spleen, and survival rate between ApcMin/+ and ApcMin/+Ei24 Tg mice. Ei24 plays no significant role in colon cancer caused by the substitutional mutation of Apc in mice. Therefore, our result dismisses the hypothesized direct link between ApcMin/+ mutation and Ei24 expression in colorectal cancer model.
Collapse
Affiliation(s)
- Tae Wook Nam
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Song Yi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae Hoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae Il Roh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
7
|
EI24 Suppresses Tumorigenesis in Pancreatic Cancer via Regulating c-Myc. Gastroenterol Res Pract 2018; 2018:2626545. [PMID: 30369947 PMCID: PMC6189671 DOI: 10.1155/2018/2626545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/22/2018] [Accepted: 08/23/2018] [Indexed: 02/08/2023] Open
Abstract
The EI24 autophagy-associated transmembrane protein is frequently associated with tumor growth and patient survival. In the present study, we found that EI24 was downregulated in pancreatic ductal adenocarcinoma (PDAC) tissues compared with adjacent normal tissues and was associated with cancer cell differentiation. Overexpression of EI24 suppressed cancer cell growth in vitro and in vivo and induced cell cycle S phase arrest, with no impact on caspase-dependent apoptosis. EI24 overexpression also resulted in reduced c-Myc expression, an oncogene in PDAC, accompanied with increased LC3B-II formation, increased Beclin-1, and diminished p62. Together, we propose that EI24 suppresses cell proliferation and prompts cell cycle arrest in pancreatic cancer cells by activating the autophagic lysosomal degradation of c-Myc. Our results suggest a potential mechanism underlying the antitumor effects of EI24 in PDAC and provide insight into the crosstalk between autophagy and cell proliferation involving a possible EI24/Beclin-1/p62/c-Myc signaling pathway.
Collapse
|
8
|
Kocaturk NM, Gozuacik D. Crosstalk Between Mammalian Autophagy and the Ubiquitin-Proteasome System. Front Cell Dev Biol 2018; 6:128. [PMID: 30333975 PMCID: PMC6175981 DOI: 10.3389/fcell.2018.00128] [Citation(s) in RCA: 297] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
Autophagy and the ubiquitin-proteasome system (UPS) are the two major intracellular quality control and recycling mechanisms that are responsible for cellular homeostasis in eukaryotes. Ubiquitylation is utilized as a degradation signal by both systems, yet, different mechanisms are in play. The UPS is responsible for the degradation of short-lived proteins and soluble misfolded proteins whereas autophagy eliminates long-lived proteins, insoluble protein aggregates and even whole organelles (e.g., mitochondria, peroxisomes) and intracellular parasites (e.g., bacteria). Both the UPS and selective autophagy recognize their targets through their ubiquitin tags. In addition to an indirect connection between the two systems through ubiquitylated proteins, recent data indicate the presence of connections and reciprocal regulation mechanisms between these degradation pathways. In this review, we summarize these direct and indirect interactions and crosstalks between autophagy and the UPS, and their implications for cellular stress responses and homeostasis.
Collapse
Affiliation(s)
- Nur Mehpare Kocaturk
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Devrim Gozuacik
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul, Turkey
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, Turkey
| |
Collapse
|
9
|
DNA damage triggers tubular endoplasmic reticulum extension to promote apoptosis by facilitating ER-mitochondria signaling. Cell Res 2018; 28:833-854. [PMID: 30030520 PMCID: PMC6063967 DOI: 10.1038/s41422-018-0065-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/23/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022] Open
Abstract
The endoplasmic reticulum (ER) is composed of the nuclear envelope, perinuclear sheets and a peripheral tubular network. The peripheral ER and mitochondria form tight contacts at specific subdomains, which coordinate the functions of the two organelles and are required for multiple cellular processes such as Ca2+ transfer and apoptosis. However, it is largely unknown how ER morphology and ER-mitochondria signaling are dynamically regulated under different physiological or pathological conditions such as DNA damage. Here we show that the peripheral, tubular ER undergoes significant extension in response to DNA damage, and that this process is dependent on p53-mediated transcriptional activation of the ER-shaping proteins REEP1, REEP2 and EI24 (alias PIG8). This promotes the formation of ER-mitochondria contacts through EI24 and the mitochondrial outer membrane protein VDAC2, facilitates Ca2+ transfer from ER to mitochondria and promotes DNA damage-induced apoptosis. Thus, we identify a unique DNA damage response pathway involving alterations in ER morphology, ER-mitochondria signaling, and apoptosis.
Collapse
|
10
|
Yuan L, Wang H, Liu Q, Wang Z, Zhang M, Zhao Y, Liang K, Chen L, Xu T, Xu P. Etoposide-induced protein 2.4 functions as a regulator of the calcium ATPase and protects pancreatic β-cell survival. J Biol Chem 2018; 293:10128-10140. [PMID: 29769309 DOI: 10.1074/jbc.ra118.002399] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/07/2018] [Indexed: 12/22/2022] Open
Abstract
Calcium homeostasis is essential for maintaining the viability and function of pancreatic β cells and plays a key role in preventing the development of diabetes. Decreased levels of ATPase sarcoplasmic/endoplasmic reticulum Ca2+-transporting 2 (ATP2a2), the main calcium pump in β cells, are often found in individuals with diabetes and in diabetic animal models. However, the regulators of ATP2a2 and the molecular mechanisms responsible for controlling ATP2a2 activity remain unclear. Etoposide-induced protein 2.4 (Ei24) is also down-regulated in β cells of diabetic individuals, whereas the effect of decreased Ei24 level on β-cell function is not clarified. Here, using Cre-LoxP and CRISPR/Cas9-based genomic knockout (KO) approaches to generate pancreatic β cell-specific Ei24 KO mice and pancreatic β-cell lines, we found that Ei24 regulates ATP2a2 activity. Specifically, we observed that Ei24 binds to ATP2a2 through Ei24 residues 293-299, which we named here the ATP2a2-interacting region (AIR). Loss of Ei24 inactivated ATP2a2, disrupted calcium homeostasis, and deactivated the calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2)-AMP-activated protein kinase (AMPK) pathway. Elevation of calcium concentration in the endoplasmic reticulum or agonist-induced AMPK activation rescued pancreatic β-cell survival and improved glucose tolerance of Ei24 KO mice. Our findings indicate that targeting the Ei24-ATP2a2 interaction to increase ATP2a2 activity can protect pancreatic β cells and improve glucose homeostasis in diabetic models, suggesting that Ei24 could potentially serve as a target to prevent or manage diabetes.
Collapse
Affiliation(s)
- Lin Yuan
- From the Key Laboratory of RNA Biology and
| | - Huiyu Wang
- From the Key Laboratory of RNA Biology and.,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| | - Qi Liu
- From the Key Laboratory of RNA Biology and.,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| | - Zhe Wang
- From the Key Laboratory of RNA Biology and.,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| | | | - Yan Zhao
- the National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101
| | - Kuo Liang
- the Department of General Surgery, XuanWu Hospital, Capital Medical University, Beijing 100053, and
| | - Liangyi Chen
- the State Key Laboratory of Biomembrane and Membrane Biotechnology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Tao Xu
- the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101, .,the National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101
| | - Pingyong Xu
- From the Key Laboratory of RNA Biology and .,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| |
Collapse
|
11
|
Nam T, Han JH, Devkota S, Lee HW. Emerging Paradigm of Crosstalk between Autophagy and the Ubiquitin-Proteasome System. Mol Cells 2017; 40:897-905. [PMID: 29237114 PMCID: PMC5750708 DOI: 10.14348/molcells.2017.0226] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/13/2017] [Accepted: 11/23/2017] [Indexed: 02/08/2023] Open
Abstract
Cellular protein homeostasis is maintained by two major degradation pathways, namely the ubiquitin-proteasome system (UPS) and autophagy. Until recently, the UPS and autophagy were considered to be largely independent systems targeting proteins for degradation in the proteasome and lysosome, respectively. However, the identification of crucial roles of molecular players such as ubiquitin and p62 in both of these pathways as well as the observation that blocking the UPS affects autophagy flux and vice versa has generated interest in studying crosstalk between these pathways. Here, we critically review the current understanding of how the UPS and autophagy execute coordinated protein degradation at the molecular level, and shed light on our recent findings indicating an important role of an autophagy-associated transmembrane protein EI24 as a bridging molecule between the UPS and autophagy that functions by regulating the degradation of several E3 ligases with Really Interesting New Gene (RING)-domains.
Collapse
Affiliation(s)
- Taewook Nam
- Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 03722,
Republic of Korea
| | - Jong Hyun Han
- Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 03722,
Republic of Korea
| | - Sushil Devkota
- Section of Cell and Developmental Biology, University of California San Diego, La Jolla, CA,
USA
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 03722,
Republic of Korea
| |
Collapse
|
12
|
Ali M, Devkota S, Roh JI, Lee J, Lee HW. Telomerase reverse transcriptase induces basal and amino acid starvation-induced autophagy through mTORC1. Biochem Biophys Res Commun 2016; 478:1198-204. [DOI: 10.1016/j.bbrc.2016.08.094] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 01/06/2023]
|
13
|
Devkota S, Jeong H, Kim Y, Ali M, Roh JI, Hwang D, Lee HW. Functional characterization of EI24-induced autophagy in the degradation of RING-domain E3 ligases. Autophagy 2016; 12:2038-2053. [PMID: 27541728 PMCID: PMC5103340 DOI: 10.1080/15548627.2016.1217371] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Historically, the ubiquitin-proteasome system (UPS) and autophagy pathways were believed to be independent; however, recent data indicate that these pathways engage in crosstalk. To date, the players mediating this crosstalk have been elusive. Here, we show experimentally that EI24 (EI24, autophagy associated transmembrane protein), a key component of basal macroautophagy/autophagy, degrades 14 physiologically important E3 ligases with a RING (really interesting new gene) domain, whereas 5 other ligases were not degraded. Based on the degradation results, we built a statistical model that predicts the RING E3 ligases targeted by EI24 using partial least squares discriminant analysis. Of 381 RING E3 ligases examined computationally, our model predicted 161 EI24 targets. Those targets are primarily involved in transcription, proteolysis, cellular bioenergetics, and apoptosis and regulated by TP53 and MTOR signaling. Collectively, our work demonstrates that EI24 is an essential player in UPS-autophagy crosstalk via degradation of RING E3 ligases. These results indicate a paradigm shift regarding the fate of E3 ligases.
Collapse
Affiliation(s)
- Sushil Devkota
- a Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center , Yonsei University , Seoul , Republic of Korea
| | - Hyobin Jeong
- b Department of New Biology and Center for Plant Aging Research , Institute for Basic Science, DGIST , Daegu , Republic of Korea
| | - Yunmi Kim
- a Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center , Yonsei University , Seoul , Republic of Korea
| | - Muhammad Ali
- a Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center , Yonsei University , Seoul , Republic of Korea
| | - Jae-Il Roh
- a Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center , Yonsei University , Seoul , Republic of Korea
| | - Daehee Hwang
- b Department of New Biology and Center for Plant Aging Research , Institute for Basic Science, DGIST , Daegu , Republic of Korea
| | - Han-Woong Lee
- a Department of Biochemistry, College of Life Science and Biotechnology and Yonsei Laboratory Animal Research Center , Yonsei University , Seoul , Republic of Korea
| |
Collapse
|
14
|
Choi JM, Jang JY, Choi YR, Kim HR, Cho BC, Lee HW. Reduced expression of EI24 confers resistance to gefitinib through IGF-1R signaling in PC9 NSCLC cells. Lung Cancer 2015; 90:175-81. [PMID: 26342551 DOI: 10.1016/j.lungcan.2015.08.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 08/20/2015] [Accepted: 08/27/2015] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Lung cancer is the commonly diagnosed cancer and is the leading cause of cancer-related mortality worldwide. The most prevalent form of lung cancer is NSCLC, comprising 80% of all lung cancer cases, and epidermal growth factor receptor (EGFR) is frequently mutated in NSCLC. EI24 is a p53-responsive gene and plays an important role in tumor suppression. In the current study, we found that reduced expression of EI24 conferred resistance to EGFR-tyrosine-kinase inhibitor (TKI) in NSCLC cells. MATERIALS AND METHODS The correlation between EI24 expression and EGFR-TKI drug resistance in EGFR-driven tumors were determined from microarray datasets. The phospho-protein expression profiles of receptor tyrosine kinases and protein kinases were examined using antibody arrays method in PC9 cells expressing shRNAs targeting EI24 and gefitinib-resistant PC9-GR cells expressing exogenous EI24. RESULTS AND CONCLUSIONS The EGFR-TKI resistant clones had reduced expression of EI24 mRNA compared to the sensitive clones, and EI24 knockdown rendered sensitive cells resistant to EGFR-TKI. Receptor tyrosine kinase screening revealed the involvement of a kinase switch in EI24-mediated regulation of drug sensitivity. We found that EI24 modulates the insulin growth factor-1 receptor (IGF-1R) pathway through the induction of IGF-1. Combination treatment with EGFR and IGF-1R inhibitors significantly reduced the viability of EI24 knockdown-induced resistant cell lines compared to single-agent treatments. We also showed that low EI24 and high IGF-1R expressions in lung cancer patients were correlated with reduced overall survival. Taken together, these results suggest a potential role for EI24 as a biomarker of drug resistance, and indicate that combination therapy with EGFR and IGF-1R inhibitors would be effective in NSCLC patients with low EI24 expression.
Collapse
Affiliation(s)
- Jung-Min Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Seoul, South Korea
| | - Ji-Young Jang
- Department of Biochemistry, College of Life Science and Biotechnology, Seoul, South Korea
| | - Yu-Ra Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Seoul, South Korea
| | - Hye Ryun Kim
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, South Korea; Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Byoung Chul Cho
- Yonsei Cancer Center, Division of Medical Oncology, Yonsei University College of Medicine, Seoul, South Korea; Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Seoul, South Korea; Laboratory Animal Research Center, Yonsei University, Seoul, South Korea.
| |
Collapse
|
15
|
Choi JM, Devkota S, Sung YH, Lee HW. EI24 regulates epithelial-to-mesenchymal transition and tumor progression by suppressing TRAF2-mediated NF-κB activity. Oncotarget 2014; 4:2383-96. [PMID: 24280371 PMCID: PMC3926834 DOI: 10.18632/oncotarget.1434] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Tumor metastasis is a multistep process that requires the concerted activity of discrete biological functions. The epithelial-to-mesenchymal transition (EMT) is the most critical mechanism implicated in tumor progression that is controlled by the inflammatory microenvironment. Understanding how an inflammatory microenvironment is maintained and contributes to tumor progression will be crucial for the development of new effective therapies. Here, we report that etoposide induced 2.4 (EI24) has a multifaceted role against tumor progression that is regulated by both EMT and inflammation. Decreased expression levels of EI24 in epithelial tumor cells induced EMT in association with increased cell motility and invasiveness and resistance to anoikis. Overexpression of EI24 resulted in the opposite cell biological characteristics and suppressed in vivo metastatic behavior. EI24 attenuated NF-κB activity by binding to the Complex I component TRAF2 and inducing its lysosome-dependent degradation, leading to transcriptional alterations of EMT-and inflammation-related genes. Analysis of clinical samples demonstrated that reduced EI24 expression and copy number was positively correlated with tumor malignancy and poor prognosis. Collectively, these findings establish EI24 as a critical suppressor of tumor progression and implicate EI24 expression level in malignant tumors as a useful therapeutic and diagnostic marker.
Collapse
Affiliation(s)
- Jung-Min Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | | | | | | |
Collapse
|
16
|
Sung YH, Jin Y, Kang Y, Devkota S, Lee J, Roh JI, Lee HW. Ei24, a novel E2F target gene, affects p53-independent cell death upon ultraviolet C irradiation. J Biol Chem 2013; 288:31261-7. [PMID: 24014029 DOI: 10.1074/jbc.m113.477570] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The deficiency of retinoblastoma (Rb) gene deregulates E2F transcription factors and thus induces E2F target genes directly or p53 target genes indirectly via mouse p19(Arf) (or p14(ARF) in humans), an E2F target gene. Here, we identified that etoposide-induced 2.4 mRNA (Ei24)/p53-induced gene 8 (Pig8), a p53 target gene involved in apoptosis and autophagy, was up-regulated in Rb(-/-) mouse embryonic fibroblasts (MEFs). The Ei24 promoter was activated by E2F1 via multiple E2F-responsive elements, independently of the previously reported p53-responsive element. Chromatin immunoprecipitation assays revealed that E2F1 directly acts on the mouse Ei24 promoter. We observed that Ei24 expression was suppressed in p53(-/-) MEFs upon UVC irradiation, which was exacerbated in p53(-/-) E2f1(-/-) MEFs, supporting the positive role of E2F1 on Ei24 transcription. Furthermore, Ei24 knockdown sensitized p53(-/-) MEFs against UVC irradiation. Together, our data indicate that Ei24 is a novel E2F target gene contributing to the survival of p53-deficient cells upon UVC irradiation and thus may have a potential significance as a therapeutic target of certain chemotherapy for treating p53-deficient tumors.
Collapse
Affiliation(s)
- Young Hoon Sung
- From the Department of Biochemistry, College of Life Science and Biotechnology, and Laboratory Animal Research Center, Yonsei University, Seoul 120-749, Korea and
| | | | | | | | | | | | | |
Collapse
|