1
|
Mazzolini L, Touriol C. PERK-Olating Through Cancer: A Brew of Cellular Decisions. Biomolecules 2025; 15:248. [PMID: 40001551 PMCID: PMC11852789 DOI: 10.3390/biom15020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The type I protein kinase PERK is an endoplasmic reticulum (ER) transmembrane protein that plays a multifaceted role in cancer development and progression, influencing tumor growth, metastasis, and cellular stress responses. The activation of PERK represents one of the three signaling pathways induced during the unfolded protein response (UPR), which is triggered, in particular, in tumor cells that constitutively experience various intracellular and extracellular stresses that impair protein folding within the ER. PERK activation can lead to both pro-survival and proapoptotic outcomes, depending on the cellular context and the extent of ER stress. It helps the reprogramming of the gene expression in cancer cells, thereby ensuring survival in the face of oncogenic stress, such as replicative stress and DNA damage, and also microenvironmental challenges, including hypoxia, angiogenesis, and metastasis. Consequently, PERK contributes to tumor initiation, transformation, adaptation to the microenvironment, and chemoresistance. However, sustained PERK activation in cells can also impair cell proliferation and promote apoptotic death by various interconnected processes, including mitochondrial dysfunction, translational inhibition, the accumulation of various cellular stresses, and the specific induction of multifunctional proapoptotic factors, such as CHOP. The dual role of PERK in promoting both tumor progression and suppression makes it a complex target for therapeutic interventions. A comprehensive understanding of the intricacies of PERK pathway activation and their impact is essential for the development of effective therapeutic strategies, particularly in diseases like cancer, where the ER stress response is deregulated in most, if not all, of the solid and liquid tumors. This article provides an overview of the knowledge acquired from the study of animal models of cancer and tumor cell lines cultured in vitro on PERK's intracellular functions and their impact on cancer cells and their microenvironment, thus highlighting potential new therapeutic avenues that could target this protein.
Collapse
|
2
|
Wu HY, Cao SY, Xu ZG, Wang T, Ji GW, Wang K. Construction of a radiogenomic signature based on endoplasmic reticulum stress for predicting prognosis and systemic combination therapy response in hepatocellular carcinoma. BMC Cancer 2025; 25:131. [PMID: 39849389 PMCID: PMC11756198 DOI: 10.1186/s12885-025-13433-4] [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: 09/28/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common tumors worldwide. Various factors in the tumor environment (TME) can lead to the activation of endoplasmic reticulum stress (ERS), thereby affecting the occurrence and development of tumors. The objective of our study was to develop and validate a radiogenomic signature based on ERS to predict prognosis and systemic combination therapy response. METHODS Using data from The Cancer Genome Atlas Program (TCGA) as a training cohort and data from International cancer genome consortium (ICGC) as a testing cohort. Univariate Cox regression and multivariate Cox regression analysis were used to identify prognostic-related genes and construct a model. HCC single-cell data obtained from Gene Expression Omnibus (GEO) were used to map gene signatures and explore inter-cellular signaling communications. Finally, a radiogenomic signature was used to predict the objective response rate (ORR) and overall survival (OS). RESULTS A total of four gene signatures related to ERS, including Stanniocalcin-2 (STC2), Melanoma-Associated Antigen 3 (MAGEA3), BR Serine/Threonine-Protein Kinase 2 (BRSK2), DEAD/H-Box Helicase 11 (DDX11) were identified. Macrophages were significantly different between high-risk and low-risk groups. The high-risk group showed higher targeting programmed cell death-1 (PD-1) and mutated tumor protein p53 (TP53) scores. Drug sensitivity analysis found that most sensitive drugs target the phosphatidylinositol 3-kinase/ mechanistic target of rapamycin (PI3K/mTOR) signaling pathway. Further research revealed the expression of STC2 in the endothelial cells (ECs), particularly plasmalemma vesicle associated protein (PLVAP) + ECs, and may regulate the reprogramming and function of macrophages. Furthermore, we identified nine radiomic features and established a radiogenomic signature based on ERS that can predict prognosis and response to systemic combination therapy. This signature can guide systemic combination therapy for patients with unresectable HCC. CONCLUSIONS We established an ERS prognostic model that can predict patient prognosis. We also found that ERS is closely related to the TME and is mainly manifested in the interaction between tumor-associated endothelial cells (TAEs) and tumor-associated macrophages (TAMs). Moreover, we constructed a radiogenomic signature based on the ERS. This signature can guide subsequent combination therapy for patients with unresectable HCC.
Collapse
Affiliation(s)
- Huai-Yu Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China
- NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, People's Republic of China
| | - Shu-Ya Cao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China
- NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, People's Republic of China
| | - Zheng-Gang Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China
- NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, People's Republic of China
| | - Tian- Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China
- NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, People's Republic of China
| | - Gu-Wei Ji
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China.
- NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, People's Republic of China.
| | - Ke Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, People's Republic of China.
- NHC Key laboratory of Hepatobiliary cancers, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Tak J, Kim YS, Kim SG. Roles of X-box binding protein 1 in liver pathogenesis. Clin Mol Hepatol 2025; 31:1-31. [PMID: 39355873 PMCID: PMC11791611 DOI: 10.3350/cmh.2024.0441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024] Open
Abstract
The prevalence of drug-induced liver injury (DILI) and viral liver infections presents significant challenges in modern healthcare and contributes to considerable morbidity and mortality worldwide. Concurrently, metabolic dysfunctionassociated steatotic liver disease (MASLD) has emerged as a major public health concern, reflecting the increasing rates of obesity and leading to more severe complications such as fibrosis and hepatocellular carcinoma. X-box binding protein 1 (XBP1) is a distinct transcription factor with a basic-region leucine zipper structure, whose activity is regulated by alternative splicing in response to disruptions in endoplasmic reticulum (ER) homeostasis and the unfolded protein response (UPR) activation. XBP1 interacts with a key signaling component of the highly conserved UPR and is critical in determining cell fate when responding to ER stress in liver diseases. This review aims to elucidate the emerging roles and molecular mechanisms of XBP1 in liver pathogenesis, focusing on its involvement in DILI, viral liver infections, MASLD, fibrosis/cirrhosis, and liver cancer. Understanding the multifaceted functions of XBP1 in these liver diseases offers insights into potential therapeutic strategies to restore ER homeostasis and mitigate liver damage.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|
4
|
Zhang H, Tang M, Liu Q, Wu D, Sun B, Dong J, Guan L, Luo J, Zeng M. PAT exposure caused human hepatocytes apoptosis and induced mice subacute liver injury by activating oxidative stress and the ERS-associated PERK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177003. [PMID: 39433224 DOI: 10.1016/j.scitotenv.2024.177003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
With the widespread use of antimony compounds in synthetic materials and processing, the occupational exposure and environmental pollution caused by antimony have attracted the attention of researchers. Studies have shown that antimony compounds can cause liver damage, but the mechanism has not yet been elucidated. In this study, we used the trivalent potassium antimony tartrate (PAT) to infect L02 hepatocytes and Kunming (KM) mice to establish an antimony-induced apoptosis model of L02 cells and a subacute liver injury model of KM mice. We found that PAT exposure caused hepatocyte apoptosis and was accompanied by oxidative stress and endoplasmic reticulum stress (ERS), and the ERS-associated PERK pathway was activated. Further experimental results showed that N-acetyl-l-cysteine (NAC) pretreatment or silencing of the PERK gene in L02 cells reduced PAT-induced apoptosis. The activity of SOD and CAT in treated L02 cells was increased, the malondialdehyde content in L02 cells and liver tissues was decreased, and the content of ERS-related proteins GRP78 and CHOP, as well as the content of PERK-pathway-related proteins p-PERK/PERK, p-eif2α/eif2α and ATF4 protein were significantly reduced. Overall, PAT exposure triggered hepatocyte apoptosis and liver injury by inducing oxidative stress and activating the ERS-associated PERK pathway; however, this effect could be alleviated by NAC intervention or silencing of PERK in hepatocytes.
Collapse
Affiliation(s)
- Hualing Zhang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Meng Tang
- Center for Disease Control and Prevention, Jiulongpo District, Chongqing 400050, PR China
| | - Qin Liu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Die Wu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Bing Sun
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jingbang Dong
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Lan Guan
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jianlan Luo
- Institute of Geophysical & Geochemical Exploration of Hunan, Changsha 411100, PR China
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| |
Collapse
|
5
|
Zhou S, Cheng K, Peng Y, Liu Y, Hu Q, Zeng S, Qi X, Yu L. Regulation mechanism of endoplasmic reticulum stress on metabolic enzymes in liver diseases. Pharmacol Res 2024; 207:107332. [PMID: 39089398 DOI: 10.1016/j.phrs.2024.107332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The endoplasmic reticulum (ER) plays a pivotal role in protein folding and secretion, Ca2+ storage, and lipid synthesis in eukaryotic cells. When the burden of protein synthesis and folding required to be handled exceeds the processing capacity of the ER, the accumulation of misfolded/unfolded proteins triggers ER stress. In response to short-term ER stress, the unfolded protein response (UPR) is activated to allow cells to survive. When ER stress is severe and sustained, it typically provokes cell death through multiple approaches. It is well documented that ER stress and metabolic deregulation are functionally intertwined, both are considered contributing factors to the pathogenesis of liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), ischemia/reperfusion (I/R) injury, viral hepatitis, liver fibrosis, and hepatocellular carcinoma (HCC). Hepatocytes are rich in smooth and rough ER, which harbor metabolic enzymes that are capable of sensing alterations in various nutritional status and external stimuli. Extensive research has focused on the molecular mechanism linking ER stress with metabolic enzymes. The purpose of this review is to summarize the current knowledge regarding the effects of ER stress on metabolic enzymes in various liver diseases and to provide potential therapeutic strategies for chronic liver diseases via targeting UPR.
Collapse
Affiliation(s)
- Shaojun Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Kaiwen Cheng
- Medical Research Center, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing 312000, China
| | - Yi Peng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuxi Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qingqing Hu
- The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Jinhua 322023, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xuchen Qi
- Department of Pharmacy, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing 312000, China; Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China.
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China; Department of Pharmacy, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing 312000, China; Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou 310024, China; Department of Pharmacy, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
6
|
Hu Q, Zhang L, Tao Y, Xie S, Wang A, Luo C, Yang R, Shen Z, He B, Fang Y, Chen P. Semaglutide Ameliorates Hepatocyte Steatosis in a Cell Co-Culture System by Downregulating the IRE1α-XBP1-C/EBPα Signaling Pathway in Macrophages. Pharmacology 2024; 110:26-35. [PMID: 39089233 DOI: 10.1159/000540654] [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: 11/06/2023] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is currently the most common type of chronic liver disease. Semaglutide is a glucose-lowering drug administered for the treatment of type 2 diabetes mellitus (T2DM) and is clinically effective in the treatment of NAFLD. X-box binding protein 1 (XBP1) is related to the pathogenesis of both NAFLD and T2DM. The aim of the present study was to demonstrate whether the underlying mechanism of semaglutide treatment for NAFLD is via downregulation of the inositol-requiring transmembrane kinase/endonuclease-1α (IRE1α)-XBP1-CCAAT/enhancer binding protein α (C/EBPα) signaling pathway in macrophages. METHODS In the present study, NAFLD cell modeling was induced by oleic acid (0.4 mm) and palmitic acid (0.2 mm). Hepatocytes (AML12) and macrophages (RAW264.7) were co-cultured in 6-well Transwell plates. Semaglutide (60 or 140 nm) was administrated for 24 h, while pioglitazone (2 μm) and toyocamycin (200 nm) were used as a positive control drug and a XBP1 inhibitor, respectively. Autophagy and apoptosis of AML12 cells were detected by transmission electron microscopy and Western blotting (WB). Hepatocyte steatosis was evaluated by adopting total intracellular triglyceride determination, analysis of the relative expression of proteins and genes associated with lipid metabolism and hepatocyte Oil red O staining. Detection of inflammation factors was conducted by ELISA and WB. To explore the underlying mechanism of NAFLD treatment with semaglutide, the relative expression of related proteins and genes were tested. RESULTS Our study demonstrated that semaglutide treatment improved autophagy and inhibited apoptosis of hepatocytes, while notably ameliorating steatosis of hepatocytes. In addition, inflammation was attenuated in the NAFLD cell co-culture model after semaglutide administration. Semaglutide also significantly reduced the protein and gene expression levels of the IRE1α-XBP1-C/EBPα signaling pathway in macrophages. CONCLUSION Semaglutide partially ameliorated NAFLD by downregulating the IRE1α-XBP1-C/EBPα signaling pathway in macrophages. These findings may provide a potential theoretical basis for semaglutide therapy for NAFLD.
Collapse
Affiliation(s)
- Qin Hu
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Zhang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - YiTing Tao
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - ShuangLin Xie
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - AiYun Wang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Caiying Luo
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - RenHua Yang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Zhiqiang Shen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Bo He
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Yu Fang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Peng Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
- College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| |
Collapse
|
7
|
Lu HJ, Koju N, Sheng R. Mammalian integrated stress responses in stressed organelles and their functions. Acta Pharmacol Sin 2024; 45:1095-1114. [PMID: 38267546 PMCID: PMC11130345 DOI: 10.1038/s41401-023-01225-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024]
Abstract
The integrated stress response (ISR) triggered in response to various cellular stress enables mammalian cells to effectively cope with diverse stressful conditions while maintaining their normal functions. Four kinases (PERK, PKR, GCN2, and HRI) of ISR regulate ISR signaling and intracellular protein translation via mediating the phosphorylation of eukaryotic translation initiation factor 2 α (eIF2α) at Ser51. Early ISR creates an opportunity for cells to repair themselves and restore homeostasis. This effect, however, is reversed in the late stages of ISR. Currently, some studies have shown the non-negligible impact of ISR on diseases such as ischemic diseases, cognitive impairment, metabolic syndrome, cancer, vanishing white matter, etc. Hence, artificial regulation of ISR and its signaling with ISR modulators becomes a promising therapeutic strategy for relieving disease symptoms and improving clinical outcomes. Here, we provide an overview of the essential mechanisms of ISR and describe the ISR-related pathways in organelles including mitochondria, endoplasmic reticulum, Golgi apparatus, and lysosomes. Meanwhile, the regulatory effects of ISR modulators and their potential application in various diseases are also enumerated.
Collapse
Affiliation(s)
- Hao-Jun Lu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Nirmala Koju
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
8
|
Hazari Y, Chevet E, Bailly-Maitre B, Hetz C. ER stress signaling at the interphase between MASH and HCC. Hepatology 2024:01515467-990000000-00844. [PMID: 38626349 DOI: 10.1097/hep.0000000000000893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/28/2024] [Indexed: 04/18/2024]
Abstract
HCC is the most frequent primary liver cancer with an extremely poor prognosis and often develops on preset of chronic liver diseases. Major risk factors for HCC include metabolic dysfunction-associated steatohepatitis, a complex multifactorial condition associated with abnormal endoplasmic reticulum (ER) proteostasis. To cope with ER stress, the unfolded protein response engages adaptive reactions to restore the secretory capacity of the cell. Recent advances revealed that ER stress signaling plays a critical role in HCC progression. Here, we propose that chronic ER stress is a common transversal factor contributing to the transition from liver disease (risk factor) to HCC. Interventional strategies to target the unfolded protein response in HCC, such as cancer therapy, are also discussed.
Collapse
Affiliation(s)
- Younis Hazari
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Department of Biotechnology, University of Kashmir, Srinagar, India
| | - Eric Chevet
- Inserm U1242, University of Rennes, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Béatrice Bailly-Maitre
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1065, Université Côte d'Azur (UCA), Centre Méditerranéen de Médecine Moléculaire (C3M), 06204 Nice, France Team "Metainflammation and Hematometabolism", Metabolism Department, France
- Université Côte d'Azur, INSERM, U1065, C3M, 06200 Nice, France
| | - Claudio Hetz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Buck Institute for Research on Aging, Novato, California, USA
| |
Collapse
|
9
|
Yuan S, She D, Jiang S, Deng N, Peng J, Ma L. Endoplasmic reticulum stress and therapeutic strategies in metabolic, neurodegenerative diseases and cancer. Mol Med 2024; 30:40. [PMID: 38509524 PMCID: PMC10956371 DOI: 10.1186/s10020-024-00808-9] [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: 12/15/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The accumulation of unfolded or misfolded proteins within the endoplasmic reticulum (ER), due to genetic determinants and extrinsic environmental factors, leads to endoplasmic reticulum stress (ER stress). As ER stress ensues, the unfolded protein response (UPR), comprising three signaling pathways-inositol-requiring enzyme 1, protein kinase R-like endoplasmic reticulum kinase, and activating transcription factor 6 promptly activates to enhance the ER's protein-folding capacity and restore ER homeostasis. However, prolonged ER stress levels propels the UPR towards cellular demise and the subsequent inflammatory cascade, contributing to the development of human diseases, including cancer, neurodegenerative disorders, and diabetes. Notably, increased expression of all three UPR signaling pathways has been observed in these pathologies, and reduction in signaling molecule expression correlates with decreased proliferation of disease-associated target cells. Consequently, therapeutic strategies targeting ER stress-related interventions have attracted significant research interest. In this review, we elucidate the critical role of ER stress in cancer, metabolic, and neurodegenerative diseases, offering novel therapeutic approaches for these conditions.
Collapse
Affiliation(s)
- Siqi Yuan
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Dan She
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Shangming Jiang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Nan Deng
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jiayi Peng
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
10
|
García-Carrillo R, Molina-Pelayo FA, Zarate-Lopez D, Cabrera-Aguilar A, Ortega-Domínguez B, Domínguez-López M, Chiquete-Félix N, Dagnino-Acosta A, Velasco-Loyden G, Chávez E, Castro-Sánchez L, de Sánchez VC. An adenosine derivative promotes mitochondrial supercomplexes reorganization and restoration of mitochondria structure and bioenergetics in a diethylnitrosamine-induced hepatocellular carcinoma model. Sci Rep 2024; 14:6348. [PMID: 38491051 PMCID: PMC10943223 DOI: 10.1038/s41598-024-56306-9] [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: 12/22/2023] [Accepted: 03/05/2024] [Indexed: 03/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) progression is associated with dysfunctional mitochondria and bioenergetics impairment. However, no data about the relationship between mitochondrial supercomplexes (hmwSC) formation and ATP production rates in HCC are available. Our group has developed an adenosine derivative, IFC-305, which improves mitochondrial function, and it has been proposed as a therapeutic candidate for HCC. We aimed to determine the role of IFC-305 on both mitochondrial structure and bioenergetics in a sequential cirrhosis-HCC model in rats. Our results showed that IFC-305 administration decreased the number and size of liver tumors, reduced the expression of tumoral markers, and reestablished the typical architecture of the hepatic parenchyma. The livers of treated rats showed a reduction of mitochondria number, recovery of the mtDNA/nDNA ratio, and mitochondrial length. Also, IFC-305 increased cardiolipin and phosphatidylcholine levels and promoted hmwSC reorganization with changes in the expression levels of hmwSC assembly-related genes. IFC-305 in HCC modified the expression of several genes encoding elements of electron transport chain complexes and increased the ATP levels by recovering the complex I, III, and V activity. We propose that IFC-305 restores the mitochondrial bioenergetics in HCC by normalizing the quantity, morphology, and function of mitochondria, possibly as part of its hepatic restorative effect.
Collapse
Grants
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- FOP02-2022-02 project 321696 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- Ciencia de Frontera-2019 project 501204 Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT)
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
- PAPIIT-UNAM project IN214419 Universidad Nacional Autónoma de México
Collapse
Affiliation(s)
- Rosendo García-Carrillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México
| | | | - David Zarate-Lopez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México
| | - Alejandro Cabrera-Aguilar
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Bibiana Ortega-Domínguez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Mariana Domínguez-López
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Natalia Chiquete-Félix
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Adan Dagnino-Acosta
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México
- CONAHCYT-Universidad de Colima, 28045, Colima, México
| | - Gabriela Velasco-Loyden
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Enrique Chávez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Luis Castro-Sánchez
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, 28045, Colima, México.
- CONAHCYT-Universidad de Colima, 28045, Colima, México.
| | - Victoria Chagoya de Sánchez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México.
| |
Collapse
|
11
|
Batel A, Polović M, Glumac M, Šuman O, Jadrijević S, Lozić B, Petrović M, Samardžija B, Bradshaw NJ, Skube K, Palada V, Acman M, Marinović Terzić I. SPRTN is involved in hepatocellular carcinoma development through the ER stress response. Cancer Gene Ther 2024; 31:376-386. [PMID: 38086993 DOI: 10.1038/s41417-023-00708-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 03/16/2024]
Abstract
Endoplasmic reticulum (ER) stress, prompted by the accumulation of misfolded or unfolded proteins, triggers the activation of the unfolded protein response (UPR) pathway to restore ER homeostasis. This stress response is implicated in the development of hepatocellular carcinoma (HCC). A biallelic mutation in SPRTN is currently the only known single-gene mutation implicated in the early onset of HCC. However, the exact mechanism linking SPRTN mutations to HCC remains unclear. In our study, we analyzed SPRTN and UPR in 21 human HCC tissue samples using RT-qPCR, immunoblot, and immunohistochemistry. We found alterations in the expression levels of SPRTN and UPR-related genes and proteins in HCC samples. The impact of SPRTN on the ER stress response was assessed in SPRTN-depleted HepG2 cells through RNA sequencing, pull-down assay, comet assay, and mitotic index calculation. We demonstrated that SPRTN interacts with the UPR sensor GRP78. Furthermore, we observed a decrease in SPRTN levels during ER stress, and increased sensitivity to ER stress in SPRTN-depleted cells. These findings suggest an essential role for SPRTN in the ER stress response and provide new insights into HCC pathogenesis. This newly discovered function of SPRTN could significantly enhance our understanding and treatment of HCC.
Collapse
Affiliation(s)
- Anja Batel
- Laboratory for Cancer Research, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Mirjana Polović
- Laboratory for Cancer Research, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Mateo Glumac
- Laboratory for Cancer Research, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Oliver Šuman
- Department of Abdominal Surgery, Merkur Clinical Hospital, Zajčeva 19, 10000, Zagreb, Croatia
| | - Stipislav Jadrijević
- Department of Abdominal Surgery, Merkur Clinical Hospital, Zajčeva 19, 10000, Zagreb, Croatia
| | - Bernarda Lozić
- Laboratory for Cancer Research, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
- Laboratory for Human Genetics, University Hospital Split, Spinčićeva 1, 21000, Split, Croatia
| | - Marija Petrović
- Laboratory for Human Genetics, University Hospital Split, Spinčićeva 1, 21000, Split, Croatia
| | - Bobana Samardžija
- Faculty of Biotechnology & Drug Development, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia
| | - Nicholas J Bradshaw
- Faculty of Biotechnology & Drug Development, University of Rijeka, Radmile Matejčić 2, 51000, Rijeka, Croatia
| | - Karlo Skube
- Selvita, Prilaz baruna Filipovića 29, 10000, Zagreb, Croatia
| | - Vinko Palada
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Mislav Acman
- Omics solutions, trg 101. Brigade HV 1, 10000, Zagreb, Croatia
| | - Ivana Marinović Terzić
- Laboratory for Cancer Research, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia.
| |
Collapse
|
12
|
Venkatesan N, Doskey LC, Malhi H. The Role of Endoplasmic Reticulum in Lipotoxicity during Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1887-1899. [PMID: 37689385 PMCID: PMC10699131 DOI: 10.1016/j.ajpath.2023.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
Perturbations in lipid and protein homeostasis induce endoplasmic reticulum (ER) stress in metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease. Lipotoxic and proteotoxic stress can activate the unfolded protein response (UPR) transducers: inositol requiring enzyme1α, PKR-like ER kinase, and activating transcription factor 6α. Collectively, these pathways induce expression of genes that encode functions to resolve the protein folding defect and ER stress by increasing the protein folding capacity of the ER and degradation of misfolded proteins. The ER is also intimately connected with lipid metabolism, including de novo ceramide synthesis, phospholipid and cholesterol synthesis, and lipid droplet formation. Following their activation, the UPR transducers also regulate lipogenic pathways in the liver. With persistent ER stress, cellular adaptation fails, resulting in hepatocyte apoptosis, a pathological marker of liver disease. In addition to the ER-nucleus signaling activated by the UPR, the ER can interact with other organelles via membrane contact sites. Modulating intracellular communication between ER and endosomes, lipid droplets, and mitochondria to restore ER homeostasis could have therapeutic efficacy in ameliorating liver disease. Recent studies have also demonstrated that cells can convey ER stress by the release of extracellular vesicles. This review discusses lipotoxic ER stress and the central role of the ER in communicating ER stress to other intracellular organelles in MASLD pathogenesis.
Collapse
Affiliation(s)
- Nanditha Venkatesan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Luke C Doskey
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
13
|
Luna-Marco C, Ubink A, Kopsida M, Heindryckx F. Endoplasmic Reticulum Stress and Metabolism in Hepatocellular Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1377-1388. [PMID: 36309104 DOI: 10.1016/j.ajpath.2022.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/23/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, accounting for 85% to 90% of all liver cancer cases. It is a hepatocyte-derived primary tumor, causing 550,000 deaths per year, ranking it as one of the most common cancers worldwide. The liver is a highly metabolic organ with multiple functions, including digestion, detoxification, breakdown of fats, and production of bile and cholesterol, in addition to storage of vitamins, glycogen, and minerals, and synthesizing plasma proteins and clotting factors. Due to these fundamental and diverse functions, the malignant transformation of hepatic cells can have a severe impact on the liver's metabolism. Furthermore, tumorigenesis is often accompanied by activation of the endoplasmic reticulum (ER) stress pathways, which are known to be highly intertwined with several metabolic pathways. Because HCC is characterized by changes in the metabolome and by an aberrant activation of the ER stress pathways, the aim of this review was to summarize the current knowledge that links ER stress and metabolism in HCC, thereby focusing on potential therapeutic targets.
Collapse
Affiliation(s)
- Clara Luna-Marco
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Anna Ubink
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Maria Kopsida
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Femke Heindryckx
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Ishteyaque S, Yadav KS, Verma S, Washimkar KR, Mugale MN. CYP2E1 triggered GRP78/ATF6/CHOP signaling axis inhibit apoptosis and promotes progression of hepatocellular carcinoma. Arch Biochem Biophys 2023; 745:109701. [PMID: 37499993 DOI: 10.1016/j.abb.2023.109701] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death worldwide. Cytochrome P450 2E1 (CYP2E1) is an enzyme, primarily involved in the metabolism of xenobiotics and procarcinogens. The present study was designed to investigate the potential role of CYP2E1 triggered endoplasmic reticulum stress in the progression of HCC through inhibition of apoptosis. In vitro CYP2E1 promotes HepG2 cell migration, reduced chromatin condensation, enhanced intracellular ROS accumulation and induce cell cycle progression. Conversely this effect was averted by CYP2E1 siRNA, selective inhibitor Diallyl sulphide (DAS) and antioxidants (vitamin C and E). In vivo Diethylnitrosamine (DEN) induced HCC rats showed decreased body weight and increased relative liver weight. Moreover, macro trabecular-massive HCC (MTM-HCC) histological subtyping showed pathological features like well-differentiated tumors, micro-trabecular and pseudo glandular patterns, megakaryocytes and cholestasis. Masson's trichrome staining revealed an intensive accumulation of collagen fibers in the extracellular matrix (ECM). Increased CYP2E1, VEGF and PCNA enhance the carcinogenicity as revealed in immunohistochemistry results. Immunoblot analysis showed reduced expression of copper-zinc superoxide dismutase (CuZnSOD) and manganese superoxide dismutase (MnSOD) in cytosolic as well as mitochondrial fraction of rat liver tissue respectively. Also, increased level of CYP2E1 stimulated the upregulation of unfolded proteins response (UPR) and ER stress-related proteins such as Glucose regulatory protein 78 (GRP78), activating transcription factor 6 (ATF6) and CCAAT enhancer-binding protein (C/EBP) homologous protein (CHOP). Meanwhile, CYP2E1 stimulated ER-stress reduces BCL2 and downregulates the cleaved caspase 3 thus suppresses apoptosis. in. Furthermore, immunofluorescence revealed increased expression level of α-SMA in the HCC rat liver tissue. The level of CYP2E1 mRNA was significantly increased. Altogether, these findings indicate that CYP2E1 has a dynamic role in the pathogenesis of HCC and might be a budding agent in liver carcinogenesis therapy.
Collapse
Affiliation(s)
- Sharmeen Ishteyaque
- Division of Cancer Biology CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Karan Singh Yadav
- Division of Cancer Biology CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Smriti Verma
- Division of Cancer Biology CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kaveri R Washimkar
- Division of Cancer Biology CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Madhav Nilakanth Mugale
- Division of Cancer Biology CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
15
|
He F, Zhang P, Liu J, Wang R, Kaufman RJ, Yaden BC, Karin M. ATF4 suppresses hepatocarcinogenesis by inducing SLC7A11 (xCT) to block stress-related ferroptosis. J Hepatol 2023; 79:362-377. [PMID: 36996941 PMCID: PMC11332364 DOI: 10.1016/j.jhep.2023.03.016] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC), a leading cause of cancer-related death, is associated with viral hepatitis, non-alcoholic steatohepatitis (NASH), and alcohol-related steatohepatitis, all of which trigger endoplasmic reticulum (ER) stress, hepatocyte death, inflammation, and compensatory proliferation. Using ER stress-prone MUP-uPA mice, we established that ER stress and hypernutrition cooperate to cause NASH and HCC, but the contribution of individual stress effectors, such as activating transcription factor 4 (ATF4), to HCC and their underlying mechanisms of action remained unknown. METHODS Hepatocyte-specific ATF4-deficient MUP-uPA mice (MUP-uPA/Atf4Δhep) and control MUP-uPA/Atf4F/F mice were fed a high-fat diet to induce NASH-related HCC, and Atf4F/F and Atf4Δhep mice were injected with diethylnitrosamine to model carcinogen-induced HCC. Histological, biochemical, and RNA-sequencing analyses were performed to identify and define the role of ATF4-induced solute carrier family 7a member 11 (SLC7A11) expression in hepatocarcinogenesis. Reconstitution of SLC7A11 in ATF4-deficient primary hepatocytes and mouse livers was used to study its effects on ferroptosis and HCC development. RESULTS Hepatocyte ATF4 ablation inhibited hepatic steatosis, but increased susceptibility to ferroptosis, resulting in accelerated HCC development. Although ATF4 activates numerous genes, ferroptosis susceptibility and hepatocarcinogenesis were reversed by ectopic expression of a single ATF4 target, Slc7a11, coding for a subunit of the cystine/glutamate antiporter xCT, which is needed for glutathione synthesis. A ferroptosis inhibitor also reduced liver damage and inflammation. ATF4 and SLC7A11 amounts were positively correlated in human HCC and livers of patients with NASH. CONCLUSIONS Despite ATF4 being upregulated in established HCC, it serves an important protective function in normal hepatocytes. By maintaining glutathione production, ATF4 inhibits ferroptosis-dependent inflammatory cell death, which is known to promote compensatory proliferation and hepatocarcinogenesis. Ferroptosis inhibitors or ATF4 activators may also blunt HCC onset. IMPACT AND IMPLICATIONS Liver cancer or hepatocellular carcinoma (HCC) is associated with multiple aetiologies. Most HCC aetiologies cause hepatocyte stress and death, as well as subsequent inflammation, and compensatory proliferation, thereby accelerating HCCdevelopment. The contribution of individual stress effectors to HCC and their underlying mechanisms of action were heretofore unknown. This study shows that the stress-responsive transcription factor ATF4 blunts liver damage and cancer development by suppressing iron-dependent cell death (ferroptosis). Although ATF4 ablation prevents hepatic steatosis, it also increases susceptibility to ferroptosis, due to decreased expression of the cystine/glutamate antiporter SLC7A11, whose expression in human HCC and NASH correlates with ATF4. These findings reinforce the notion that benign steatosis may be protective and does not increase cancer risk unless accompanied by stress-induced liver damage. These results have important implications for prevention of liver damage and cancer.
Collapse
Affiliation(s)
- Feng He
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, San Diego, CA, USA.
| | - Peng Zhang
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Junlai Liu
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Ruolei Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, SBP Medical Discovery Institute, La Jolla, CA, USA
| | - Benjamin C Yaden
- Diabetes Novel Therapies and External Innovation, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, San Diego, CA, USA; Department of Pathology, School of Medicine, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
16
|
Ajoolabady A, Kaplowitz N, Lebeaupin C, Kroemer G, Kaufman RJ, Malhi H, Ren J. Endoplasmic reticulum stress in liver diseases. Hepatology 2023; 77:619-639. [PMID: 35524448 PMCID: PMC9637239 DOI: 10.1002/hep.32562] [Citation(s) in RCA: 154] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 02/02/2023]
Abstract
The endoplasmic reticulum (ER) is an intracellular organelle that fosters the correct folding of linear polypeptides and proteins, a process tightly governed by the ER-resident enzymes and chaperones. Failure to shape the proper 3-dimensional architecture of proteins culminates in the accumulation of misfolded or unfolded proteins within the ER, disturbs ER homeostasis, and leads to canonically defined ER stress. Recent studies have elucidated that cellular perturbations, such as lipotoxicity, can also lead to ER stress. In response to ER stress, the unfolded protein response (UPR) is activated to reestablish ER homeostasis ("adaptive UPR"), or, conversely, to provoke cell death when ER stress is overwhelmed and sustained ("maladaptive UPR"). It is well documented that ER stress contributes to the onset and progression of multiple hepatic pathologies including NAFLD, alcohol-associated liver disease, viral hepatitis, liver ischemia, drug toxicity, and liver cancers. Here, we review key studies dealing with the emerging role of ER stress and the UPR in the pathophysiology of liver diseases from cellular, murine, and human models. Specifically, we will summarize current available knowledge on pharmacological and non-pharmacological interventions that may be used to target maladaptive UPR for the treatment of nonmalignant liver diseases.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Neil Kaplowitz
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Research Center for Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cynthia Lebeaupin
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Randal J. Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jun Ren
- Department of Cardiology, Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
17
|
Song Y, Yin D, Zhang Z, Chi L. Research progress of treatment of functional dyspepsia with traditional Chinese medicine compound based on cell signal pathway. Front Pharmacol 2023; 13:1089231. [PMID: 36699059 PMCID: PMC9868459 DOI: 10.3389/fphar.2022.1089231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Functional dyspepsia (FD) is the most common clinical gastrointestinal disease, with complex and prolonged clinical symptoms. The prevalence of FD is increasing year by year, seriously affecting the quality of life of patients. The main causes of FD are related to abnormal gastrointestinal dynamics, increased visceral sensitivity, Helicobacter pylori (HP) infection, intestinal flora disturbance and psychological factors. A review of the relevant literature reveals that the mechanisms of traditional Chinese medicine (TCM) in the treatment of FD mainly involve the following pathways:5-HT signal pathway, AMPK signal pathway,C-kit signal pathway, CRF signal pathway, PERK signal pathway,NF-κB signal pathway. Based on a holistic concept, TCM promotes gastrointestinal motility, regulates visceral sensitivity and alleviates gastrointestinal inflammation through multiple signal pathways, reflecting the advantages of multi-level, multi-pathway and multi-targeted treatment of FD.
Collapse
Affiliation(s)
- Yujiao Song
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| | - Defei Yin
- Digestive System Department II, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| | - Zhenyi Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| | - Lili Chi
- Digestive System Department II, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| |
Collapse
|
18
|
Pavlović N, Heindryckx F. Targeting ER stress in the hepatic tumor microenvironment. FEBS J 2022; 289:7163-7176. [PMID: 34331743 DOI: 10.1111/febs.16145] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/30/2021] [Indexed: 01/13/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. It currently ranks as one of the most aggressive and deadly cancers worldwide, with an increasing mortality rate and limited treatment options. An important hallmark of liver pathologies, such as liver fibrosis and HCC, is the accumulation of misfolded and unfolded proteins in the lumen of the endoplasmic reticulum (ER), which induces ER stress and leads to the activation of the unfolded protein response (UPR). Upon accumulation of misfolded proteins, ER stress is sensed through three transmembrane proteins, IRE1α, PERK, and ATF6, which trigger the UPR to either alleviate ER stress or induce apoptosis. Increased expression of ER stress markers has been widely shown to correlate with fibrosis, inflammation, drug resistance, and overall HCC aggressiveness, as well as poor patient prognosis. While preclinical in vivo cancer models and in vitro approaches have shown promising results by pharmacologically targeting ER stress mediators, the major challenge of this therapeutic strategy lies in specifically and effectively targeting ER stress in HCC. Furthermore, both ER stress inducers and inhibitors have been shown to ameliorate HCC progression, adding to the complexity of targeting ER stress players as an anticancer strategy. More studies are needed to better understand the dual role and molecular background of ER stress in HCC, as well as its therapeutic potential for patients with liver cancer.
Collapse
Affiliation(s)
- Nataša Pavlović
- Department of Medical Cell Biology, Uppsala University, Sweden
| | | |
Collapse
|
19
|
Nguyen LC, Renner DM, Silva D, Yang D, Parenti NA, Medina KM, Nicolaescu V, Gula H, Drayman N, Valdespino A, Mohamed A, Dann C, Wannemo K, Robinson-Mailman L, Gonzalez A, Stock L, Cao M, Qiao Z, Moellering RE, Tay S, Randall G, Beers MF, Rosner MR, Oakes SA, Weiss SR. SARS-CoV-2 Diverges from Other Betacoronaviruses in Only Partially Activating the IRE1α/XBP1 Endoplasmic Reticulum Stress Pathway in Human Lung-Derived Cells. mBio 2022; 13:e0241522. [PMID: 36125275 PMCID: PMC9600248 DOI: 10.1128/mbio.02415-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has killed over 6 million individuals worldwide and continues to spread in countries where vaccines are not yet widely available or its citizens are hesitant to become vaccinated. Therefore, it is critical to unravel the molecular mechanisms that allow SARS-CoV-2 and other coronaviruses to infect and overtake the host machinery of human cells. Coronavirus replication triggers endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR), a key host cell pathway widely believed to be essential for viral replication. We examined the master UPR sensor IRE1α kinase/RNase and its downstream transcription factor effector XBP1s, which is processed through an IRE1α-mediated mRNA splicing event, in human lung-derived cells infected with betacoronaviruses. We found that human respiratory coronavirus OC43 (HCoV-OC43), Middle East respiratory syndrome coronavirus (MERS-CoV), and murine coronavirus (MHV) all induce ER stress and strongly trigger the kinase and RNase activities of IRE1α as well as XBP1 splicing. In contrast, SARS-CoV-2 only partially activates IRE1α through autophosphorylation, but its RNase activity fails to splice XBP1. Moreover, while IRE1α was dispensable for replication in human cells for all coronaviruses tested, it was required for maximal expression of genes associated with several key cellular functions, including the interferon signaling pathway, during SARS-CoV-2 infection. Our data suggest that SARS-CoV-2 actively inhibits the RNase of autophosphorylated IRE1α, perhaps as a strategy to eliminate detection by the host immune system. IMPORTANCE SARS-CoV-2 is the third lethal respiratory coronavirus, after MERS-CoV and SARS-CoV, to emerge this century, causing millions of deaths worldwide. Other common coronaviruses such as HCoV-OC43 cause less severe respiratory disease. Thus, it is imperative to understand the similarities and differences among these viruses in how each interacts with host cells. We focused here on the inositol-requiring enzyme 1α (IRE1α) pathway, part of the host unfolded protein response to virus-induced stress. We found that while MERS-CoV and HCoV-OC43 fully activate the IRE1α kinase and RNase activities, SARS-CoV-2 only partially activates IRE1α, promoting its kinase activity but not RNase activity. Based on IRE1α-dependent gene expression changes during infection, we propose that SARS-CoV-2 prevents IRE1α RNase activation as a strategy to limit detection by the host immune system.
Collapse
Affiliation(s)
- Long C. Nguyen
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - David M. Renner
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Dongbo Yang
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Nicholas A. Parenti
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kaeri M. Medina
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vlad Nicolaescu
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
| | - Haley Gula
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
| | - Nir Drayman
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Andrea Valdespino
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Adil Mohamed
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Christopher Dann
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Kristin Wannemo
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | | | - Alan Gonzalez
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Letícia Stock
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Mengrui Cao
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Zeyu Qiao
- Department of Chemistry, University of Chicago, Chicago, Illinois, USA
| | | | - Savas Tay
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Glenn Randall
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
| | - Michael F. Beers
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Scott A. Oakes
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Susan R. Weiss
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Endoplasmic Reticulum Stress-Related Signature for Predicting Prognosis and Immune Features in Hepatocellular Carcinoma. J Immunol Res 2022; 2022:1366508. [PMID: 36003068 PMCID: PMC9393196 DOI: 10.1155/2022/1366508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/29/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) with cancer cells under endoplasmic reticulum (ER) stress has a poor prognosis. This study is aimed at discovering credible biomarkers for predicting the prognosis of HCC based on ER stress-related genes (ERSRGs). We constructed a novel four-ERSRG prognostic risk model, including PON1, AGR2, SSR2, and TMCC1, through a series of bioinformatic approaches, which can accurately predict survival outcomes in HCC patients. Higher risk scores were linked to later grade, recurrence, advanced TNM stage, later T stage, and HBV infection. In addition, 20 fresh frozen tumors and normal tissues from HCC patients were collected and used to validate the genes expressed in the signature by qRT-PCR and immunohistochemical (IHC) assays. Moreover, we found the ER stress-related signature could reflect the infiltration levels of different immune cells in the tumor microenvironment (TME) and forecast the efficacy of immune checkpoint inhibitor (ICI) treatment. Finally, we created a nomogram incorporating this ER stress-related signature. In conclusion, our constructed four-gene risk model associated with ER stress can accurately predict survival outcomes in HCC patients, and the model's risk score is associated with the poor clinical classification.
Collapse
|
21
|
Gentile D, Esposito M, Grumati P. Metabolic adaption of cancer cells toward autophagy: Is there a role for ER-phagy? Front Mol Biosci 2022; 9:930223. [PMID: 35992272 PMCID: PMC9382244 DOI: 10.3389/fmolb.2022.930223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is an evolutionary conserved catabolic pathway that uses a unique double-membrane vesicle, called autophagosome, to sequester cytosolic components, deliver them to lysosomes and recycle amino-acids. Essentially, autophagy acts as a cellular cleaning system that maintains metabolic balance under basal conditions and helps to ensure nutrient viability under stress conditions. It is also an important quality control mechanism that removes misfolded or aggregated proteins and mediates the turnover of damaged and obsolete organelles. In this regard, the idea that autophagy is a non-selective bulk process is outdated. It is now widely accepted that forms of selective autophagy are responsible for metabolic rewiring in response to cellular demand. Given its importance, autophagy plays an essential role during tumorigenesis as it sustains malignant cellular growth by acting as a coping-mechanisms for intracellular and environmental stress that occurs during malignant transformation. Cancer development is accompanied by the formation of a peculiar tumor microenvironment that is mainly characterized by hypoxia (oxygen < 2%) and low nutrient availability. Such conditions challenge cancer cells that must adapt their metabolism to survive. Here we review the regulation of autophagy and selective autophagy by hypoxia and the crosstalk with other stress response mechanisms, such as UPR. Finally, we discuss the emerging role of ER-phagy in sustaining cellular remodeling and quality control during stress conditions that drive tumorigenesis.
Collapse
Affiliation(s)
- Debora Gentile
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Marianna Esposito
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Scuola Superiore Meridionale, Naples, Italy
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
- *Correspondence: Paolo Grumati,
| |
Collapse
|
22
|
Nguyen LC, Renner DM, Silva D, Yang D, Parenti N, Medina KM, Nicolaescu V, Gula H, Drayman N, Valdespino A, Mohamed A, Dann C, Wannemo K, Robinson-Mailman L, Gonzalez A, Stock L, Cao M, Qiao Z, Moellering RE, Tay S, Randall G, Beers MF, Rosner MR, Oakes SA, Weiss SR. SARS-CoV-2 diverges from other betacoronaviruses in only partially activating the IRE1α/XBP1 ER stress pathway in human lung-derived cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.12.30.474519. [PMID: 35821981 PMCID: PMC9275661 DOI: 10.1101/2021.12.30.474519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has killed over 6 million individuals worldwide and continues to spread in countries where vaccines are not yet widely available, or its citizens are hesitant to become vaccinated. Therefore, it is critical to unravel the molecular mechanisms that allow SARS-CoV-2 and other coronaviruses to infect and overtake the host machinery of human cells. Coronavirus replication triggers endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR), a key host cell pathway widely believed essential for viral replication. We examined the master UPR sensor IRE1α kinase/RNase and its downstream transcription factor effector XBP1s, which is processed through an IRE1α-mediated mRNA splicing event, in human lung-derived cells infected with betacoronaviruses. We found human respiratory coronavirus OC43 (HCoV-OC43), Middle East respiratory syndrome coronavirus (MERS-CoV), and murine coronavirus (MHV) all induce ER stress and strongly trigger the kinase and RNase activities of IRE1α as well as XBP1 splicing. In contrast, SARS-CoV-2 only partially activates IRE1α through autophosphorylation, but its RNase activity fails to splice XBP1. Moreover, while IRE1α was dispensable for replication in human cells for all coronaviruses tested, it was required for maximal expression of genes associated with several key cellular functions, including the interferon signaling pathway, during SARS-CoV-2 infection. Our data suggest that SARS-CoV-2 actively inhibits the RNase of autophosphorylated IRE1α, perhaps as a strategy to eliminate detection by the host immune system. IMPORTANCE SARS-CoV-2 is the third lethal respiratory coronavirus after MERS-CoV and SARS-CoV to emerge this century, causing millions of deaths world-wide. Other common coronaviruses such as HCoV-OC43 cause less severe respiratory disease. Thus, it is imperative to understand the similarities and differences among these viruses in how each interacts with host cells. We focused here on the inositol-requiring enzyme 1α (IRE1α) pathway, part of the host unfolded protein response to virus-induced stress. We found that while MERS-CoV and HCoV-OC43 fully activate the IRE1α kinase and RNase activities, SARS-CoV-2 only partially activates IRE1α, promoting its kinase activity but not RNase activity. Based on IRE1α-dependent gene expression changes during infection, we propose that SARS-CoV-2 prevents IRE1α RNase activation as a strategy to limit detection by the host immune system.
Collapse
Affiliation(s)
- Long C. Nguyen
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, U.S.A
| | - David M. Renner
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, IL 60637, U.S.A
| | - Dongbo Yang
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, U.S.A
| | - Nicholas Parenti
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kaeri M. Medina
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vlad Nicolaescu
- Department of Microbiology, University of Chicago, Chicago, IL 60637, U.S.A
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Haley Gula
- Department of Microbiology, University of Chicago, Chicago, IL 60637, U.S.A
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Nir Drayman
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, U.S.A
| | - Andrea Valdespino
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, U.S.A
| | - Adil Mohamed
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, U.S.A
| | - Christopher Dann
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, U.S.A
| | - Kristin Wannemo
- Department of Pathology, University of Chicago, Chicago, IL 60637, U.S.A
| | | | - Alan Gonzalez
- Department of Pathology, University of Chicago, Chicago, IL 60637, U.S.A
| | - Letícia Stock
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, U.S.A
| | - Mengrui Cao
- Department of Pathology, University of Chicago, Chicago, IL 60637, U.S.A
| | - Zeyu Qiao
- Department of Chemistry, University of Chicago, Chicago, IL 60637, U.S.A
| | | | - Savas Tay
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, U.S.A
| | - Glenn Randall
- Department of Microbiology, University of Chicago, Chicago, IL 60637, U.S.A
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Michael F. Beers
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marsha Rich Rosner
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, U.S.A
| | - Scott A. Oakes
- Department of Pathology, University of Chicago, Chicago, IL 60637, U.S.A
| | - Susan R. Weiss
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
23
|
Flessa C, Kyrou I, Nasiri‐Ansari N, Kaltsas G, Kassi E, Randeva HS. Endoplasmic reticulum stress in nonalcoholic (metabolic associated) fatty liver disease (NAFLD/MAFLD). J Cell Biochem 2022; 123:1585-1606. [PMID: 35490371 DOI: 10.1002/jcb.30247] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023]
Affiliation(s)
- Christina‐Maria Flessa
- Department of Biological Chemistry, Medical School National and Kapodistrian University of Athens Athens Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) University Hospitals Coventry and Warwickshire NHS Trust Coventry UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) University Hospitals Coventry and Warwickshire NHS Trust Coventry UK
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School University of Warwick Coventry UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing Coventry University Coventry UK
- Aston Medical School, College of Health and Life Sciences Aston University Birmingham UK
- Department of Food Science & Human Nutrition Agricultural University of Athens Athens Greece
| | - Narjes Nasiri‐Ansari
- Department of Biological Chemistry, Medical School National and Kapodistrian University of Athens Athens Greece
| | - Gregory Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital National and Kapodistrian University of Athens Athens Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School National and Kapodistrian University of Athens Athens Greece
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital National and Kapodistrian University of Athens Athens Greece
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) University Hospitals Coventry and Warwickshire NHS Trust Coventry UK
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School University of Warwick Coventry UK
| |
Collapse
|
24
|
Ward CP, Peng L, Yuen S, Chang M, Karapetyan R, Nyangau E, Mohammed H, Palacios H, Ziari N, Joe LK, Frakes AE, Dandan M, Dillin A, Hellerstein MK. ER Unfolded Protein Response in Liver In Vivo Is Characterized by Reduced, Not Increased, De Novo Lipogenesis and Cholesterol Synthesis Rates with Uptake of Fatty Acids from Adipose Tissue: Integrated Gene Expression, Translation Rates and Metabolic Fluxes. Int J Mol Sci 2022; 23:ijms23031073. [PMID: 35162995 PMCID: PMC8835023 DOI: 10.3390/ijms23031073] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
The unfolded protein response in the endoplasmic reticulum (UPRER) is involved in a number of metabolic diseases. Here, we characterize UPRER-induced metabolic changes in mouse livers in vivo through metabolic labeling and mass spectrometric analysis of lipid and proteome-wide fluxes. We induced UPRER by tunicamycin administration and measured synthesis rates of proteins, fatty acids and cholesterol, as well as RNA-seq. Contrary to reports in isolated cells, hepatic de novo lipogenesis and cholesterogenesis were markedly reduced, as were mRNA levels and synthesis rates of lipogenic proteins. H&E staining showed enrichment with lipid droplets while electron microscopy revealed ER morphological changes. Interestingly, the pre-labeling of adipose tissue prior to UPRER induction resulted in the redistribution of labeled fatty acids from adipose tissue to the liver, with replacement by unlabeled glycerol in the liver acylglycerides, indicating that the liver uptake was of free fatty acids, not whole glycerolipids. The redistribution of adipose fatty acids to the liver was not explicable by altered plasma insulin, increased fatty acid levels (lipolysis) or by reduced food intake. Synthesis of most liver proteins was suppressed under UPRER conditions, with the exception of BiP, other chaperones, protein disulfide isomerases, and proteins of ribosomal biogenesis. Protein synthesis rates generally, but not always, paralleled changes in mRNA. In summary, this combined approach, linking static changes with fluxes, revealed an integrated reduction of lipid and cholesterol synthesis pathways, from gene expression to translation and metabolic flux rates, under UPRER conditions. The reduced lipogenesis does not parallel human fatty liver disease. This approach provides powerful tools to characterize metabolic processes underlying hepatic UPRER in vivo.
Collapse
Affiliation(s)
- Catherine P. Ward
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Lucy Peng
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Samuel Yuen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Michael Chang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Rozalina Karapetyan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Edna Nyangau
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Hussein Mohammed
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Hector Palacios
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Naveed Ziari
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Larry K. Joe
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 92093, USA; (L.K.J.); (A.E.F.); (A.D.)
| | - Ashley E. Frakes
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 92093, USA; (L.K.J.); (A.E.F.); (A.D.)
| | - Mohamad Dandan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Andrew Dillin
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 92093, USA; (L.K.J.); (A.E.F.); (A.D.)
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
- Correspondence:
| |
Collapse
|
25
|
Cai C, Yang L, Zhou K. 8DEstablishment and validation of a hypoxia-related signature predicting prognosis in hepatocellular carcinoma. BMC Gastroenterol 2021; 21:463. [PMID: 34895169 PMCID: PMC8667367 DOI: 10.1186/s12876-021-02057-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Background Hypoxia plays a crucial role in immunotherapy of hepatocellular carcinoma (HCC) by changing the tumor microenvironment. Until now the association between hypoxia genes and prognosis of HCC remains obscure. We attempt to construct a hypoxia model to predict the prognosis in HCC.
Results We screened out 3 hypoxia genes (ENO1, UGP2, TPI1) to make the model, which can predict prognosis in HCC. And this model emerges as an independent prognostic factor for HCC. A Nomogram was drawn to evaluate the overall survival in a more accurate way. Furthermore, immune infiltration state and immunosuppressive microenvironment of the tumor were detected in high-risk patients. Conclusion We establish and validate a risk prognostic model developed by 3 hypoxia genes, which could effectively evaluate the prognosis of HCC patients. This prognostic model can be used as a guidance for hypoxia modification in HCC patients undergoing immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-021-02057-0.
Collapse
Affiliation(s)
- Congbo Cai
- Emergency Department of Yinzhou No.2 Hospital, Ningbo, 315000, Zhejiang, China
| | - Lei Yang
- Emergency Department of Yinzhou No.2 Hospital, Ningbo, 315000, Zhejiang, China
| | - Kena Zhou
- Gastroenterology Department of Ningbo No. 9 Hospital, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
26
|
Yu M, Lun J, Zhang H, Wang L, Zhang G, Zhang H, Fang J. Targeting UPR branches, a potential strategy for enhancing efficacy of cancer chemotherapy. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1417-1427. [PMID: 34664059 DOI: 10.1093/abbs/gmab131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer cells are often exposed to cell intrinsic stresses and environmental perturbations that may lead to accumulation of unfolded and/or misfolded proteins in the lumen of endoplasmic reticulum (ER), a cellular condition known as ER stress. In response to ER stress, the cells elicit an adaptive process called unfolded protein response (UPR) to cope with the stress, supporting cellular homeostasis and survival. The ER stress sensors inositol requiring protein 1α (IRE1α), eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3, also called PERK), and activating transcription factor 6 (ATF6) constitute the three branches of UPR to resolve ER stress. IRE1α, PERK, and ATF6 play an important role in tumor cell growth and survival. They are also involved in chemotherapy resistance of cancers. These have generated widespread interest in targeting these UPR branches for cancer treatment. In this review, we provide an overview of the role of IRE1α, PERK, and ATF6 in cancer progression and drug resistance and we summarize the research advances in targeting these UPR branches to enhance the efficacy of chemotherapy of cancers.
Collapse
Affiliation(s)
- Mengchao Yu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute of Qingdao University, Qingdao 266061, China
| | - Jie Lun
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute of Qingdao University, Qingdao 266061, China
| | - Hongwei Zhang
- Oncology Department, Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| | - Lei Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute of Qingdao University, Qingdao 266061, China
| | - Gang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute of Qingdao University, Qingdao 266061, China
| | - Haisheng Zhang
- Center for Cancer Targeted Therapies, Signet Therapeutics Inc., Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Jing Fang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Cancer Institute of Qingdao University, Qingdao 266061, China
| |
Collapse
|
27
|
Karan D. CCL23 in Balancing the Act of Endoplasmic Reticulum Stress and Antitumor Immunity in Hepatocellular Carcinoma. Front Oncol 2021; 11:727583. [PMID: 34671553 PMCID: PMC8522494 DOI: 10.3389/fonc.2021.727583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/17/2021] [Indexed: 11/15/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is a cellular process in response to stress stimuli in protecting functional activities. However, sustained hyperactive ER stress influences tumor growth and development. Hepatocytes are enriched with ER and highly susceptible to ER perturbations and stress, which contribute to immunosuppression and the development of aggressive and drug-resistant hepatocellular carcinoma (HCC). ER stress-induced inflammation and tumor-derived chemokines influence the immune cell composition at the tumor site. Consequently, a decrease in the CCL23 chemokine in hepatic tumors is associated with poor survival of HCC patients and could be a mechanism hepatic tumor cells use to evade the immune system. This article describes the prospective role of CCL23 in alleviating ER stress and its impact on the HCC tumor microenvironment in promoting antitumor immunity. Moreover, approaches to reactivate CCL23 combined with immune checkpoint blockade or chemotherapy drugs may provide novel opportunities to target hepatocellular carcinoma.
Collapse
Affiliation(s)
- Dev Karan
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
28
|
Ko P, Choi JH, Song S, Keum S, Jeong J, Hwang YE, Kim JW, Rhee S. Microtubule Acetylation Controls MDA-MB-231 Breast Cancer Cell Invasion through the Modulation of Endoplasmic Reticulum Stress. Int J Mol Sci 2021; 22:ijms22116018. [PMID: 34199510 PMCID: PMC8199658 DOI: 10.3390/ijms22116018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 12/27/2022] Open
Abstract
During aggressive cancer progression, cancer cells adapt to unique microenvironments by withstanding various cellular stresses, including endoplasmic reticulum (ER) stress. However, the mechanism whereby cancer cells overcome the ER stress to survive remains to be elucidated. Herein, we demonstrated that microtubule acetylation in cancer cells grown on a stiff matrix promotes cancer progression by preventing excessive ER stress. Downregulation of microtubule acetylation using shRNA or CRSIPR/Cas9 techniques targeting ATAT1, which encodes α-tubulin N-acetyltransferase (αTAT1), resulted in the upregulation of ER stress markers, changes in ER morphology, and enhanced tunicamycin-induced UPR signaling in cancer cells. A set of genes involved in cancer progression, especially focal adhesion genes, were downregulated in both ATAT1-knockout and tunicamycin-treated cells, whereas ATAT1 overexpression restored the gene expression inhibited by tunicamycin. Finally, the expression of ATAT1 and ER stress marker genes were negatively correlated in various breast cancer types. Taken together, our results suggest that disruption of microtubule acetylation is a potent therapeutic tool for preventing breast cancer progression through the upregulation of ER stress. Moreover, ATAT1 and ER stress marker genes may be useful diagnostic markers in various breast cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sangmyung Rhee
- Correspondence: ; Tel.: +82-2-820-5818; Fax: +82-2-825-5206
| |
Collapse
|
29
|
Flessa CM, Kyrou I, Nasiri-Ansari N, Kaltsas G, Papavassiliou AG, Kassi E, Randeva HS. Endoplasmic Reticulum Stress and Autophagy in the Pathogenesis of Non-alcoholic Fatty Liver Disease (NAFLD): Current Evidence and Perspectives. Curr Obes Rep 2021; 10:134-161. [PMID: 33751456 DOI: 10.1007/s13679-021-00431-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Non-alcoholic fatty liver disease (NAFLD) is one of the most common causes of chronic liver disease with rising prevalence worldwide. Herein, we provide a comprehensive overview of the current knowledge supporting the role of ER stress and autophagy processes in NAFLD pathogenesis and progression. We also highlight the interrelation between these two pathways and the impact of ER stress and autophagy modulators on NAFLD treatment. RECENT FINDINGS The pathophysiological mechanisms involved in NAFLD progression are currently under investigation. The endoplasmic reticulum (ER) stress and the concomitant unfolded protein response (UPR) seem to contribute to its pathogenesis mainly due to high ER content in the liver which exerts significant metabolic functions and can be dysregulated. Furthermore, disruption of autophagy processes has also been identified in NAFLD. The crucial role of these two pathways in NAFLD is underlined by the fact that they have recently emerged as promising targets of therapeutic interventions. There is a greater need for finding the natural/chemical compounds and drugs which can modulate the ER stress pathway and autophagy for the treatment of NAFLD. Clarifying the inter-relation between these two pathways and their interaction with inflammatory and apoptotic mechanisms will allow the development of additional therapeutic options which can better target and reprogram the underlying pathophysiological pathways, aiming to attenuate NAFLD progression.
Collapse
Affiliation(s)
- Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, B4 7ET, Birmingham, UK
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Gregory Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece.
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece.
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK.
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
30
|
Maintenance of Endoplasmic Reticulum Protein Homeostasis in Cancer: Friend or Foe. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021. [PMID: 34050868 DOI: 10.1007/978-3-030-67696-4_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
The endoplasmic reticulum, as the site of synthesis for proteins in the secretory pathway has evolved select machineries to ensure the correct folding and modification of proteins. However, sometimes these quality control mechanisms fail and proteins are misfolded. Other factors, such as nutrient deprivation, hypoxia or an increased demand on protein synthesis can also cause the accumulation of unfolded or misfolded proteins in the endoplasmic reticulum. There are mechanisms that recognise and deal with this accumulation of protein through degradation and/or export. Many diseases are associated with aberrant quality control mechanisms, and among these, cancer has emerged as a group of diseases that rely on endoplasmic reticulum homeostasis to sustain development and growth. The knowledge of how protein quality control operates in cancer has identified opportunities for these pathways to be pharmacologically targeted, which could lead to newer or more effective treatments in the future.
Collapse
|
31
|
Yukimoto A, Watanabe T, Sunago K, Nakamura Y, Tanaka T, Koizumi Y, Yoshida O, Tokumoto Y, Hirooka M, Abe M, Hiasa Y. The long noncoding RNA of RMRP is downregulated by PERK, which induces apoptosis in hepatocellular carcinoma cells. Sci Rep 2021; 11:7926. [PMID: 33846370 PMCID: PMC8041825 DOI: 10.1038/s41598-021-86592-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress plays an important role in hepatocyte degeneration, especially in patients with chronic liver injury. Protein kinase R-like endoplasmic reticulum kinase (PERK) is a key molecule in ER stress. PERK may contribute to apoptotic cell death in HCC, however the details of the mechanism are not clear. In this study, we identified PERK-associated molecules using transcriptome analysis. We modulated PERK expression using a plasmid, tunicamycin and siRNA against PERK, and then confirmed the target gene expression with real-time PCR and Northern blotting. We further analyzed the apoptotic function. Transcriptome analysis revealed that expression of the RNA component of mitochondrial RNA processing endoribonuclease (RMRP), which is a long noncoding RNA, was strongly correlated with the function of PERK. The expression of RMRP was correlated with the expression of PERK in experiments with the siRNA and PERK plasmid in both HCC cell lines and human HCC tissue. Furthermore, RMRP downregulation induced apoptotic cell death. RMRP is downregulated by PERK, which induces apoptosis in HCC. RMRP could be a new therapeutic target to regulate HCC in patients with chronic liver diseases associated with ER stress.
Collapse
Affiliation(s)
- Atsushi Yukimoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Takao Watanabe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Kotaro Sunago
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Yoshiko Nakamura
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Takaaki Tanaka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Yohei Koizumi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Yoshio Tokumoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Masanori Abe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, 791-0295, Japan.
| |
Collapse
|
32
|
Wei J, Fang D. Endoplasmic Reticulum Stress Signaling and the Pathogenesis of Hepatocarcinoma. Int J Mol Sci 2021; 22:ijms22041799. [PMID: 33670323 PMCID: PMC7918477 DOI: 10.3390/ijms22041799] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC), also known as hepatoma, is a primary malignancy of the liver and the third leading cause of cancer mortality globally. Although much attention has focused on HCC, its pathogenesis remains largely obscure. The endoplasmic reticulum (ER) is a cellular organelle important for regulating protein synthesis, folding, modification and trafficking, and lipid metabolism. ER stress occurs when ER homeostasis is disturbed by numerous environmental, physiological, and pathological challenges. In response to ER stress due to misfolded/unfolded protein accumulation, unfolded protein response (UPR) is activated to maintain ER function for cell survival or, in cases of excessively severe ER stress, initiation of apoptosis. The liver is especially susceptible to ER stress given its protein synthesis and detoxification functions. Experimental data suggest that ER stress and unfolded protein response are involved in HCC development, aggressiveness and response to treatment. Herein, we highlight recent findings and provide an overview of the evidence linking ER stress to the pathogenesis of HCC.
Collapse
|
33
|
Xia SW, Wang ZM, Sun SM, Su Y, Li ZH, Shao JJ, Tan SZ, Chen AP, Wang SJ, Zhang ZL, Zhang F, Zheng SZ. Endoplasmic reticulum stress and protein degradation in chronic liver disease. Pharmacol Res 2020; 161:105218. [PMID: 33007418 DOI: 10.1016/j.phrs.2020.105218] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Endoplasmic reticulum (ER) stress is easily observed in chronic liver disease, which often causes accumulation of unfolded or misfolded proteins in the ER, leading to unfolded protein response (UPR). Regulating protein degradation is an integral part of UPR to relieve ER stress. The major protein degradation system includes the ubiquitin-proteasome system (UPS) and autophagy. All three arms of UPR triggered in response to ER stress can regulate UPS and autophagy. Accumulated misfolded proteins could activate these arms, and then generate various transcription factors to regulate the expression of UPS-related and autophagy-related genes. The protein degradation process regulated by UPR has great significance in many chronic liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, liver fibrosis, and hepatocellular carcinoma(HCC). In most instances, the degradation of excessive proteins protects cells with ER stress survival from apoptosis. According to the specific functions of protein degradation in chronic liver disease, choosing to promote or inhibit this process is promising as a potential method for treating chronic liver disease.
Collapse
Affiliation(s)
- Si-Wei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhi-Min Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Su-Min Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Su
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhang-Hao Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiang-Juan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shan-Zhong Tan
- Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - An-Ping Chen
- Department of Pathology, School of Medicine, Saint Louis University, MO 63104, USA
| | - Shi-Jun Wang
- Shandong University of Traditional Chinese Medicine, Jinan 250035, China
| | - Zi-Li Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shi-Zhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
34
|
Ridlo MR, Kim GA, Taweechaipaisankul A, Kim EH, Lee BC. Zinc supplementation alleviates endoplasmic reticulum stress during porcine oocyte in vitro maturation by upregulating zinc transporters. J Cell Physiol 2020; 236:2869-2880. [PMID: 32944961 DOI: 10.1002/jcp.30052] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/29/2020] [Accepted: 09/02/2020] [Indexed: 01/16/2023]
Abstract
Endoplasmic reticulum (ER) stress is a major contributor to embryonic development failure. Mammalian oocytes have a high risk of exposure to cellular stress during in vitro embryo production. We investigated the effects of zinc supplementation during in vitro maturation under ER stress. We evaluated cumulus expansion, embryonic development derived by parthenogenetic activation, reactive oxygen species, protein expression of X-box binding protein 1 (XBP1), and expression of genes related to ER stress. Supplementation with 1 μg/ml zinc significantly increased the nuclear maturation of oocytes, cleavage and blastocyst formation rates, and total blastocyst cell number (p < .05). Under ER stress, zinc significantly reduced protein expression of XBP1, and increased cleavage and blastocyst rates (p < .05). Concomitantly, zinc supplementation upregulated the expression of zinc transporters (SLC39A14 and SLC39A10), PTGS2, and downregulated ER stress-related genes (sXBP1, uXBP1, ATF4, and PTPN1/PTP1B), and caspase 3. These results suggest that zinc supplementation alleviates ER stress by providing essential metal-ion transporters for oocyte maturation and subsequent embryonic development.
Collapse
Affiliation(s)
- Muhammad Rosyid Ridlo
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Department of Bioresources Technology and Veterinary, Vocational College, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Geon A Kim
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejon, Republic of Korea
| | - Anukul Taweechaipaisankul
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Eui Hyun Kim
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Endoplasmic Reticulum Stress Regulates Cardiomyocyte Apoptosis in Myocardial Fibrosis Development via PERK-Mediated Autophagy. Cardiovasc Toxicol 2020; 20:618-626. [PMID: 32632848 DOI: 10.1007/s12012-020-09586-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endoplasmic reticulum stress (ERS) is involved in a variety of diseases. Recently, it was found that ERS induces not only apoptosis but also autophagy. Previous studies showed that inhibition of autophagy alleviates cell injury. The purpose of our study was to investigate the involvement of the R-like ER kinase (PERK) in ERS-induced autophagy in H9c2 cardiomyoblasts. To address this aim, therefore, H9c2 cells were treated with PERK agonist and inhibitor after establishment of rapamycin-induced ERS models in H9c2 cardiomyoblasts. Transmission electron microscopy and immunofluorescence staining were used to detect degrees of ERS-induced autophagy, apoptosis and myocardial fibrosis. Western blotting was employed to detect the levels of total and phosphorylated PERK, light chain 3 (LC3), P62, Caspase3, Bcl2 and Bax. Immunofluorescence staining was used to assess α-SMA density. TGF-β induced H9c2 cardiomyoblasts time-dependently upregulated col I, col III, FN, and LC3 expressions, PERK phosphorylation and α-SMA density, and downregulated P62 level compared with control cells. Treatment with PERK agonist and inhibitor respectively increased and decreased LC3 expression, conversely in P62 level, which is consistent with effect of ERS agonists and inhibitors. And a PERK inhibitor upregulated the expressions of Caspase3 and Bax, and downregulated Bcl2 level, which developed H9c2 cardiomyoblasts. Moreover, siRNA-mediated knockdown of PERK reduced ERS mediated autophagy activity and increased cells apoptosis. On the other hand, elevated autophagy activity could downregulated PERK level. Our finding showed that PERK activity mediates upregulation of ERS-induced autophagy and regulation of cardiomyocyte apoptosis in H9c2 cardiomyoblasts.
Collapse
|
36
|
Song J, Ding W, Liu B, Liu D, Xia Z, Zhang L, Cui L, Luo Y, Jia X, Feng L. Anticancer effect of caudatin in diethylnitrosamine‑induced hepatocarcinogenesis in rats. Mol Med Rep 2020; 22:697-706. [PMID: 32626931 PMCID: PMC7339819 DOI: 10.3892/mmr.2020.11135] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
An overwhelming endoplasmic reticulum stress (ERS) and the following unfolded protein response (UPR) can induce hepatic inflammation, fibrosis and hepatocellular carcinoma (HCC). Caudatin, one of the species of C-21 steroidal glycosides mainly isolated from the roots of Cynanchum bungei Decne, exhibits potent anticancer activities in vivo. However, the effect of caudatin on HCC remains unclear. In the present study, a diethylnitrosamine (DEN)-induced HCC model was established. Nodules and tumors in rat livers were monitored by T2-/T1-weighted-magnetic resonance imaging (MRI) using a 1.5 T scanner. Caudatin reduced the number and size of nodules and alleviated the inflammatory foci in the liver. In addition, the hepatic pro-inflammatory levels of interleukin (IL) 6, monocyte chemoattractant protein 1 and IL-1β were decreased in caudatin-treated rats. The DEN-induced surge in malondialdehyde, aspartate aminotransferase, alanine transaminase and TBIL were alleviated following caudatin treatment. The expression of ERS chaperones glucose-regulated protein, 94 kDa, glucose-regulated protein, 78 kDa and protein disulfide-isomerase A4 and the proliferation marker Ki-67 in liver nodules were all downregulated by caudatin as demonstrated by immunohistochemistry, reverse transcription-quantitative PCR and western blot analysis. Caudatin reduced the cytoprotective ERS sensor activating transcription factor 6-mediated signal transduction and inhibited the PKR-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 pathway. However, the effect of caudatin on inositol requiring enzyme 1 signaling was negligible. In conclusion, restoration of the dysregulated UPR program was involved in the antitumor efficacy of caudatin without inducing cumulative hepatotoxicity.
Collapse
Affiliation(s)
- Jie Song
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Wenbo Ding
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Bojia Liu
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Dan Liu
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Zhi Xia
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Li Zhang
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Li Cui
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Yi Luo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| |
Collapse
|
37
|
Lequoy M, Gigante E, Couty JP, Desbois-Mouthon C. Hepatocellular carcinoma in the context of non-alcoholic steatohepatitis (NASH): recent advances in the pathogenic mechanisms. Horm Mol Biol Clin Investig 2020; 41:/j/hmbci.ahead-of-print/hmbci-2019-0044/hmbci-2019-0044.xml. [PMID: 32112699 DOI: 10.1515/hmbci-2019-0044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer. HCC is particularly aggressive and is one of the leading causes of cancer mortality. In recent decades, the epidemiological landscape of HCC has undergone significant changes. While chronic viral hepatitis and excessive alcohol consumption have long been identified as the main risk factors for HCC, non-alcoholic steatohepatitis (NASH), paralleling the worldwide epidemic of obesity and type 2 diabetes, has become a growing cause of HCC in the US and Europe. Here, we review the recent advances in epidemiological, genetic, epigenetic and pathogenic mechanisms as well as experimental mouse models that have improved the understanding of NASH progression toward HCC. We also discuss the clinical management of patients with NASH-related HCC and possible therapeutic approaches.
Collapse
Affiliation(s)
- Marie Lequoy
- Service d'Hépato-Gastro-Entérologie, AP-HP, F-75012 Paris, France
- Centre de Recherche Saint-Antoine, INSERM, Sorbonne Université, F-75012 Paris, France
| | - Elia Gigante
- Service d'Hépato-Gastro-Entérologie, AP-HP, F-75012 Paris, France
| | - Jean-Pierre Couty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006 Paris, France
| | - Christèle Desbois-Mouthon
- Centre de Recherche des Cordeliers, INSERM UMR_S1138, 15 rue de l'école de médecine, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006 Paris, France
| |
Collapse
|
38
|
Chen Y, Zhang F, Wang D, Li L, Si H, Wang C, Liu J, Chen Y, Cheng J, Lu Y. Mesenchymal Stem Cells Attenuate Diabetic Lung Fibrosis via Adjusting Sirt3-Mediated Stress Responses in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8076105. [PMID: 32089781 PMCID: PMC7024095 DOI: 10.1155/2020/8076105] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/25/2019] [Accepted: 01/23/2020] [Indexed: 02/05/2023]
Abstract
Diabetes affects a variety of organs such as the kidneys, eyes, and liver, and there is increasing evidence that the lung is also one of the target organs of diabetes and imbalance of Sirt3-mediated stress responses such as inflammation, oxidative stress, apoptosis, autophagy, and ER stress may contribute to diabetic lung fibrosis. Although previous studies have reported that mesenchymal stem cells (MSCs) have beneficial effects on various diabetic complications, the effect and mechanisms of MSCs on diabetes-induced lung injury are not clear. In this study, the STZ-induced diabetes model was constructed in rats, and the effect and potential mechanisms of bone marrow MSCs on diabetic lung fibrosis were investigated. The results revealed that fibrotic changes in the lung were successfully induced in the diabetic rats, while MSCs significantly inhibited or even reversed the changes. Specifically, MSCs upregulated the expression levels of Sirt3 and SOD2 and then activated the Nrf2/ARE signaling pathway, thereby controlling MDA, GSH content, and iNOS and NADPH oxidase subunit p22phox expression levels in the lung tissue. Meanwhile, high levels of Sirt3 and SOD2 induced by MSCs reduced the expression levels of IL-1β, TNF-α, ICAM-1, and MMP9 by suppressing the NF-κB/HMGB1/NLRP3/caspase-1 signaling pathway, as well as regulating the expression levels of cleaved caspasese-3, Bax, and Bcl2 by upregulating the expression level of P-Akt, thereby inhibiting the apoptosis of the lung tissue. In addition, MSCs also regulated the expression levels of LC3, P62, BiP, Chop, and PERK, thereby enhancing autophagy and attenuating endoplasmic reticulum stress. Taken together, our results suggest that MSCs effectively attenuate diabetic lung fibrosis via adjusting Sirt3-mediated responses, including inflammation, oxidative stress, apoptosis, autophagy, and endoplasmic reticulum stress, providing a theoretical foundation for further exploration of MSC-based diabetic therapeutics.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Fuping Zhang
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Di Wang
- Research Core Facility, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Haibo Si
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengshi Wang
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Regenerative Medicine Research Center, Sichuan University, Chengdu 610041, China
| |
Collapse
|
39
|
Lebeaupin C, Yong J, Kaufman RJ. The Impact of the ER Unfolded Protein Response on Cancer Initiation and Progression: Therapeutic Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1243:113-131. [PMID: 32297215 PMCID: PMC7243802 DOI: 10.1007/978-3-030-40204-4_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cellular stress induced by the accumulation of misfolded proteins in the endoplasmic reticulum (ER) activates an elaborate signalling network termed the unfolded protein response (UPR). This adaptive response is mediated by the transmembrane signal transducers IRE1, PERK, and ATF6 to decide cell fate of recovery or death. In malignant cells, UPR signalling may be required to maintain ER homeostasis and survival in the tumor microenvironment characterized by oxidative stress, hypoxia, lactic acidosis and compromised protein folding. Here we provide an overview of the ER response to cellular stress and how the sustained activation of this network enables malignant cells to develop tumorigenic, metastatic and drug-resistant capacities to thrive under adverse conditions. Understanding the complexity of ER stress responses and how to target the UPR in disease will have significant potential for novel future therapeutics.
Collapse
Affiliation(s)
- Cynthia Lebeaupin
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Yong
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
40
|
Abstract
Endoplasmic reticulum (ER) stress occurs when ER homeostasis is perturbed with accumulation of unfolded/misfolded protein or calcium depletion. The unfolded protein response (UPR), comprising of inositol-requiring enzyme 1α (IRE1α), PKR-like ER kinase (PERK) and activating transcription factor 6 (ATF6) signaling pathways, is a protective cellular response activated by ER stress. However, UPR activation can also induce cell death upon persistent ER stress. The liver is susceptible to ER stress given its synthetic and other biological functions. Numerous studies from human liver samples and animal disease models have indicated a crucial role of ER stress and UPR signaling pathways in the pathogenesis of liver diseases, including non-alcoholic fatty liver disease, alcoholic liver disease, alpha-1 antitrypsin deficiency, cholestatic liver disease, drug-induced liver injury, ischemia/reperfusion injury, viral hepatitis and hepatocellular carcinoma. Extensive investigations have demonstrated the potential underlying mechanisms of the induction of ER stress and the contribution of UPR pathways during the development of the diseases. Moreover ER stress and the UPR proteins and genes have become emerging therapeutic targets to treat liver diseases.
Collapse
Affiliation(s)
- Xiaoying Liu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tarry Building 15-709, 303 East Superior Street, Chicago, IL 60611, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Corresponding author: Xiaoying-liu@northwestern
| | - Richard M. Green
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tarry Building 15-709, 303 East Superior Street, Chicago, IL 60611, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
41
|
Hypoxia Induced ER Stress Response as an Adaptive Mechanism in Cancer. Int J Mol Sci 2019; 20:ijms20030749. [PMID: 30754624 PMCID: PMC6387291 DOI: 10.3390/ijms20030749] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/23/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
It is evident that regions within tumors are deprived of oxygen, which makes the microenvironment hypoxic. Cancer cells experiencing hypoxia undergo metabolic alterations and cytoprotective adaptive mechanisms to survive such stringent conditions. While such mechanisms provide potential therapeutic targets, the mechanisms by which hypoxia regulates adaptive responses-such as ER stress response, unfolded protein response (UPR), anti-oxidative responses, and autophagy-remain elusive. In this review, we summarize the complex interplay between hypoxia and the ER stress signaling pathways that are activated in the hypoxic microenvironment of the tumors.
Collapse
|
42
|
Lebeaupin C, Vallée D, Hazari Y, Hetz C, Chevet E, Bailly-Maitre B. Endoplasmic reticulum stress signalling and the pathogenesis of non-alcoholic fatty liver disease. J Hepatol 2018; 69:927-947. [PMID: 29940269 DOI: 10.1016/j.jhep.2018.06.008] [Citation(s) in RCA: 631] [Impact Index Per Article: 90.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/22/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
Abstract
The global epidemic of obesity has been accompanied by a rising burden of non-alcoholic fatty liver disease (NAFLD), with manifestations ranging from simple steatosis to non-alcoholic steatohepatitis, potentially developing into hepatocellular carcinoma. Although much attention has focused on NAFLD, its pathogenesis remains largely obscure. The hallmark of NAFLD is the hepatic accumulation of lipids, which subsequently leads to cellular stress and hepatic injury, eventually resulting in chronic liver disease. Abnormal lipid accumulation often coincides with insulin resistance in steatotic livers and is associated with perturbed endoplasmic reticulum (ER) proteostasis in hepatocytes. In response to chronic ER stress, an adaptive signalling pathway known as the unfolded protein response is triggered to restore ER proteostasis. However, the unfolded protein response can cause inflammation, inflammasome activation and, in the case of non-resolvable ER stress, the death of hepatocytes. Experimental data suggest that the unfolded protein response influences hepatic tumour development, aggressiveness and response to treatment, offering novel therapeutic avenues. Herein, we provide an overview of the evidence linking ER stress to NAFLD and discuss possible points of intervention.
Collapse
Affiliation(s)
| | - Deborah Vallée
- Université Côte d'Azur, INSERM, U1065, C3M, 06200 Nice, France
| | - Younis Hazari
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, 02115 Boston, MA, USA
| | - Eric Chevet
- "Chemistry, Oncogenesis, Stress, Signaling", Inserm U1242, Université de Rennes, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | | |
Collapse
|
43
|
Asha K, Sharma-Walia N. Virus and tumor microenvironment induced ER stress and unfolded protein response: from complexity to therapeutics. Oncotarget 2018; 9:31920-31936. [PMID: 30159133 PMCID: PMC6112759 DOI: 10.18632/oncotarget.25886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/21/2018] [Indexed: 12/28/2022] Open
Abstract
Endoplasmic reticulum (ER) stress can be activated by various pathological and physiological conditions including the unfolded protein response (UPR) to restore homeostasis. The UPR signaling pathways initiated by double-stranded RNA-activated protein kinase (PKR) like ER kinase (PERK), inositol requiring enzyme 1 α (IRE1α), and activating transcription factor 6 (ATF6) are vital for tumor growth, aggressiveness, microenvironment remodeling, and resistance to cancer therapeutics. This review focuses on the role of ER stress and activity of UPR signaling pathways involved in tumor formation and uncontrolled cell proliferation during various cancers and viral malignancies.
Collapse
Affiliation(s)
- Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, U.S.A
| | - Neelam Sharma-Walia
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, U.S.A
| |
Collapse
|
44
|
Ogawa H, Kaira K, Takahashi K, Shimizu A, Altan B, Yoshinari D, Asao T, Oyama T. Prognostic role of BiP/GRP78 expression as ER stress in patients with gastric adenocarcinoma. Cancer Biomark 2018; 20:273-281. [PMID: 28854502 DOI: 10.3233/cbm-170062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE The glucose-regulated protein 78 (GRP78), also referred to as immunoglobulin heavy chain binding protein (BiP) (BiP/GRP78), is a major molecular chaperone in the endoplasmic reticulum (ER) and is extensively expressed in human neoplasms. Although the enhanced expression of BiP/GRP78 has been described to be associated with poor prognosis in gastric cancer (GC), details regarding its prognostic significance remain unclear. The aim of this study was to elucidate the prognostic role of BiP/GRP78 in patients with GC. METHODS Study subjects included 328 patients who underwent surgical resection. Tumor specimens of primary tumors underwent immunohistochemical staining for BiP/GRP78. RESULTS BiP/GRP78 was highly expressed in 57% (188/328) of patients. High expression of BiP/GRP78 was significantly associated with older age, male, disease staging, T factor, lymph node metastases, differentiation, lymphatic permeation, and vascular invasion. According to univariate analysis, age, disease staging, T factor, N factor, lymphatic permeation, vascular invasion, and BiP/GRP78 expression were significant prognostic factors for OS. In particular, high BiP/GRP78 expression was proven to be a significant predictor of prognosis in patients with older age, female sex, early disease stage, T1-2 factor, well or moderately differentiated tumors, and negative vascular invasion. CONCLUSION BiP/GRP78 is significantly associated with tumor aggressiveness and progression. The increased expression of BiP/GRP78 was identified as an independent factor for predicting poor OS in patients with early stage of disease, especially T1-2 factor.
Collapse
Affiliation(s)
- Hiroomi Ogawa
- Department of Thoracic and Visceral Organ Surgery, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Kyoichi Kaira
- Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Kengo Takahashi
- Department of Thoracic and Visceral Organ Surgery, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Akira Shimizu
- Department of Dermatology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Bolag Altan
- Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Daisuke Yoshinari
- Department of Thoracic and Visceral Organ Surgery, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Takayuki Asao
- Department of Thoracic and Visceral Organ Surgery, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| |
Collapse
|
45
|
Vandewynckel YP, Coucke C, Laukens D, Devisscher L, Paridaens A, Bogaerts E, Vandierendonck A, Raevens S, Verhelst X, Van Steenkiste C, Libbrecht L, Geerts A, Van Vlierberghe H. Next-generation proteasome inhibitor oprozomib synergizes with modulators of the unfolded protein response to suppress hepatocellular carcinoma. Oncotarget 2018; 7:34988-5000. [PMID: 27167000 PMCID: PMC5085204 DOI: 10.18632/oncotarget.9222] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/31/2016] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) responds poorly to conventional systemic therapies. The first-in-class proteasome inhibitor bortezomib has been approved in clinical use for hematologic malignancies and has shown modest activity in solid tumors, including HCC. However, a considerable proportion of patients fail to respond and experience important adverse events. Recently, the next-generation orally bioavailable irreversible proteasome inhibitor oprozomib was developed. Here, we assessed the efficacy of oprozomib and its effects on the unfolded protein response (UPR), a signaling cascade activated through the ATF6, PERK and IRE1 pathways by accumulation of unfolded proteins in the endoplasmic reticulum, in HCC. The effects of oprozomib and the role of the UPR were evaluated in HCC cell lines and in diethylnitrosamine-induced and xenograft mouse models for HCC. Oprozomib dose-dependently reduced the viability and proliferation of human HCC cells. Unexpectedly, oprozomib-treated cells displayed diminished cytoprotective ATF6-mediated signal transduction as well as unaltered PERK and IRE1 signaling. However, oprozomib increased pro-apoptotic UPR-mediated protein levels by prolonging their half-life, implying that the proteasome acts as a negative UPR regulator. Supplementary boosting of UPR activity synergistically improved the sensitivity to oprozomib via the PERK pathway. Oral oprozomib displayed significant antitumor effects in the orthotopic and xenograft models for HCC, and importantly, combining oprozomib with different UPR activators enhanced the antitumor efficacy by stimulating UPR-induced apoptosis without cumulative toxicity. In conclusion, next-generation proteasome inhibition by oprozomib results in dysregulated UPR activation in HCC. This finding can be exploited to enhance the antitumor efficacy by combining oprozomib with clinically applicable UPR activators.
Collapse
Affiliation(s)
| | - Céline Coucke
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | - Debby Laukens
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | - Lindsey Devisscher
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | - Annelies Paridaens
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | - Eliene Bogaerts
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | | | - Sarah Raevens
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | - Xavier Verhelst
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | | | - Louis Libbrecht
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Anja Geerts
- Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium
| | | |
Collapse
|
46
|
Protein biosynthesis, a target of sorafenib, interferes with the unfolded protein response (UPR) and ferroptosis in hepatocellular carcinoma cells. Oncotarget 2018; 9:8400-8414. [PMID: 29492203 PMCID: PMC5823558 DOI: 10.18632/oncotarget.23843] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/16/2017] [Indexed: 01/19/2023] Open
Abstract
Sorafenib is the first line treatment for advanced hepatocellular carcinoma (HCC). We explored its impact on the proteostasis of cancer cells, i.e. the processes that regulate the synthesis, maturation and turn-over of cellular proteins. We observed that sorafenib inhibits the production of the tumour marker alpha-foetoprotein (AFP) in two different HCC cell lines, an effect that correlated with a radical inhibition of protein biosynthesis. This effect was observed at clinically relevant concentrations of sorafenib and was not related to the effect of sorafenib on the transport of amino acids across the plasma membrane or the induction of the unfolded protein response (UPR). Instead, we observed that sorafenib inhibits translation initiation and the mechanistic target of rapamycin (mTOR) signaling cascade, as shown by the analysis of phosphorylation levels of the protein 4EBP1 (eukaryotic translation initiation factor 4E binding protein 1). We explored the consequences of this inhibition in HCC cells. We observed that overall sorafenib is a weak inducer of the UPR that can paradoxically prevent the UPR induced by tunicamycin. We also found no direct synergistic anticancer effect between sorafenib and various strategies that inhibit the UPR. In agreement with the possibility that translation inhibition might be an adaptive stress response in HCC cells, we noted that it protects cancer cell from ferroptosis, a form of oxidative necrosis. Our findings point to the modulation of protein biosynthesis and mTOR signaling as being important, yet complex determinants of the response of HCC cells to sorafenib.
Collapse
|
47
|
Corazzari M, Gagliardi M, Fimia GM, Piacentini M. Endoplasmic Reticulum Stress, Unfolded Protein Response, and Cancer Cell Fate. Front Oncol 2017; 7:78. [PMID: 28491820 PMCID: PMC5405076 DOI: 10.3389/fonc.2017.00078] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/10/2017] [Indexed: 12/24/2022] Open
Abstract
Perturbation of endoplasmic reticulum (ER) homeostasis results in a stress condition termed "ER stress" determining the activation of a finely regulated program defined as unfolded protein response (UPR) and whose primary aim is to restore this organelle's physiological activity. Several physiological and pathological stimuli deregulate normal ER activity causing UPR activation, such as hypoxia, glucose shortage, genome instability, and cytotoxic compounds administration. Some of these stimuli are frequently observed during uncontrolled proliferation of transformed cells, resulting in tumor core formation and stage progression. Therefore, it is not surprising that ER stress is usually induced during solid tumor development and stage progression, becoming an hallmark of such malignancies. Several UPR components are in fact deregulated in different tumor types, and accumulating data indicate their active involvement in tumor development/progression. However, although the UPR program is primarily a pro-survival process, sustained and/or prolonged stress may result in cell death induction. Therefore, understanding the mechanism(s) regulating the cell survival/death decision under ER stress condition may be crucial in order to specifically target tumor cells and possibly circumvent or overcome tumor resistance to therapies. In this review, we discuss the role played by the UPR program in tumor initiation, progression and resistance to therapy, highlighting the recent advances that have improved our understanding of the molecular mechanisms that regulate the survival/death switch.
Collapse
Affiliation(s)
- Marco Corazzari
- Department of Health Sciences, University of Piemonte Orientale "A. Avogadro", Novara, Italy.,Department Clinical Epidemiology and Translational Research, INMI-IRCCS "L. Spallanzani", Rome, Italy
| | - Mara Gagliardi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Gian Maria Fimia
- Department Clinical Epidemiology and Translational Research, INMI-IRCCS "L. Spallanzani", Rome, Italy.,Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Mauro Piacentini
- Department Clinical Epidemiology and Translational Research, INMI-IRCCS "L. Spallanzani", Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
48
|
A Systems Biological View of Life-and-Death Decision with Respect to Endoplasmic Reticulum Stress-The Role of PERK Pathway. Int J Mol Sci 2017; 18:ijms18010058. [PMID: 28067773 PMCID: PMC5297693 DOI: 10.3390/ijms18010058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/06/2016] [Accepted: 12/19/2016] [Indexed: 12/16/2022] Open
Abstract
Accumulation of misfolded/unfolded proteins in the endoplasmic reticulum (ER) leads to the activation of three branches (Protein kinase (RNA)-like endoplasmic reticulum kinase [PERK], Inositol requiring protein 1 [IRE-1] and Activating trascription factor 6 [ATF6], respectively) of unfolded protein response (UPR). The primary role of UPR is to try to drive back the system to the former or a new homeostatic state by self-eating dependent autophagy, while excessive level of ER stress results in apoptotic cell death. Our study focuses on the role of PERK- and IRE-1-induced arms of UPR in life-or-death decision. Here we confirm that silencing of PERK extends autophagy-dependent survival, whereas the IRE-1-controlled apoptosis inducer is downregulated during ER stress. We also claim that the proper order of surviving and self-killing mechanisms is controlled by a positive feedback loop between PERK and IRE-1 branches. This regulatory network makes possible a smooth, continuous activation of autophagy with respect to ER stress, while the induction of apoptosis is irreversible and switch-like. Using our knowledge of molecular biological techniques and systems biological tools we give a qualitative description about the dynamical behavior of PERK- and IRE-1-controlled life-or-death decision. Our model claims that the two arms of UPR accomplish an altered upregulation of autophagy and apoptosis inducers during ER stress. Since ER stress is tightly connected to aging and age-related degenerative disorders, studying the signaling pathways of UPR and their role in maintaining ER proteostasis have medical importance.
Collapse
|
49
|
Targeting the angio-proteostasis network: Combining the forces against cancer. Pharmacol Ther 2016; 167:1-12. [DOI: 10.1016/j.pharmthera.2016.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/14/2016] [Indexed: 01/24/2023]
|
50
|
Axten JM. Protein kinase R(PKR)–like endoplasmic reticulum kinase (PERK) inhibitors: a patent review (2010-2015). Expert Opin Ther Pat 2016; 27:37-48. [DOI: 10.1080/13543776.2017.1238072] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|