101
|
Kitsugi K, Noritake H, Matsumoto M, Hanaoka T, Umemura M, Yamashita M, Takatori S, Ito J, Ohta K, Chida T, Suda T, Kawata K. Simvastatin inhibits hepatic stellate cells activation by regulating the ferroptosis signaling pathway. Biochim Biophys Acta Mol Basis Dis 2023:166750. [PMID: 37268254 DOI: 10.1016/j.bbadis.2023.166750] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND & AIMS Ferroptosis is a form of regulated cell death and its promotion in hepatic stellate cells (HSCs) attenuates liver fibrosis. Statins, which are 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, may induce ferroptosis via the downregulation of glutathione peroxidase 4 (GPX4) by inhibiting the mevalonate pathway. However, little evidence is available regarding the association between statins and ferroptosis. Therefore, we investigated the association between statins and ferroptosis in HSCs. METHODS Two human HSC cell lines, LX-2 and TWNT-1, were treated with simvastatin, an HMG-CoA reductase inhibitor. Mevalonic acid (MVA), farnesyl pyrophosphate (FPP), and geranylgeranyl pyrophosphate (GGPP) were used to determine the involvement of the mevalonate pathway. We performed a detailed analysis of the ferroptosis signaling pathway. We also investigated human liver tissue samples from patients with nonalcoholic steatohepatitis to clarify the effect of statins on GPX4 expression. RESULTS Simvastatin reduced cell mortality and inhibited HSCs activation, accompanied by iron accumulation, oxidative stress, lipid peroxidation, and reduced GPX4 protein expression. These results indicate that simvastatin inhibits HSCs activation by promoting ferroptosis. Furthermore, treatment with MVA, FPP, or GGPP attenuated simvastatin-induced ferroptosis. These results suggest that simvastatin promotes ferroptosis in HSCs by inhibiting the mevalonate pathway. In human liver tissue samples, statins downregulated the expression of GPX4 in HSCs without affecting hepatocytes. CONCLUSIONS Simvastatin inhibits the activation of HSCs by regulating the ferroptosis signaling pathway.
Collapse
Affiliation(s)
- Kensuke Kitsugi
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.
| | - Hidenao Noritake
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Moe Matsumoto
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Tomohiko Hanaoka
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Masahiro Umemura
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Maho Yamashita
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Shingo Takatori
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Jun Ito
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kazuyoshi Ohta
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takeshi Chida
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takafumi Suda
- Division of Respiratory Medicine, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kazuhito Kawata
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
102
|
Xu X, Mao C, Zhang C, Zhang M, Gong J, Wang X. Salvianolic Acid B Inhibits Ferroptosis and Apoptosis during Myocardial Ischemia/Reperfusion Injury via Decreasing the Ubiquitin-Proteasome Degradation of GPX4 and the ROS-JNK/MAPK Pathways. Molecules 2023; 28:molecules28104117. [PMID: 37241859 DOI: 10.3390/molecules28104117] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is related to ferroptosis and apoptosis elicited by reactive oxygen species (ROS). In this research, we investigated the protective effect of salvianolic acid B (SAB) as a natural antioxidant on ferroptosis and apoptosis in the MIRI process, and discussed the protective mechanism inhibiting ubiquitin-proteasome degradation of glutathione peroxidase 4 (GPX4) and the c-Jun N-terminal kinases (JNK) apoptosis signal pathway. We observed that ferroptosis and apoptosis occurred in the MIRI rat model in vivo and the H9c2 cardiomyocyte hypoxia/reoxygenation (H/R) damage model in vitro. SAB can alleviate tissue damage related to ROS, ferroptosis and apoptosis. Ubiquitin-proteasome degradation of GPX4 occurred in H/R models, and SAB reduced the ubiquitin-proteasome degradation of GPX4. SAB downregulates JNK phosphorylation and the expression of BCL2-Associated X (Bax)/B-cell lymphoma-2 (Bcl-2) and Caspase-3 to inhibit apoptosis. The role of GPX4 in the cardioprotection of SAB was further verified by the elimination effect of the GPX4 inhibitor RAS-selective lethal 3 (RSL3). This research shows that SAB may be used as a myocardial protective agent against oxidative stress, ferroptosis and apoptosis, and has potential clinical application prospects.
Collapse
Affiliation(s)
- Xiaojin Xu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Chenhan Mao
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Chengbo Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Meng Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianbin Gong
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xindong Wang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
103
|
Wang X, Zhou Y, Min J, Wang F. Zooming in and out of ferroptosis in human disease. Front Med 2023; 17:173-206. [PMID: 37121959 DOI: 10.1007/s11684-023-0992-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/12/2023] [Indexed: 05/02/2023]
Abstract
Ferroptosis is defined as an iron-dependent regulated form of cell death driven by lipid peroxidation. In the past decade, it has been implicated in the pathogenesis of various diseases that together involve almost every organ of the body, including various cancers, neurodegenerative diseases, cardiovascular diseases, lung diseases, liver diseases, kidney diseases, endocrine metabolic diseases, iron-overload-related diseases, orthopedic diseases and autoimmune diseases. Understanding the underlying molecular mechanisms of ferroptosis and its regulatory pathways could provide additional strategies for the management of these disease conditions. Indeed, there are an expanding number of studies suggesting that ferroptosis serves as a bona-fide target for the prevention and treatment of these diseases in relevant pre-clinical models. In this review, we summarize the progress in the research into ferroptosis and its regulatory mechanisms in human disease, while providing evidence in support of ferroptosis as a target for the treatment of these diseases. We also discuss our perspectives on the future directions in the targeting of ferroptosis in human disease.
Collapse
Affiliation(s)
- Xue Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo, 315000, China
| | - Junxia Min
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Fudi Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
104
|
Guo R, Deng M, Li J, He X, He P, Liu H, Ye Y, Mo Z, He X, Li M, He Q. Depriving Tumor Cells of Ways to Metastasize: Ferroptosis Nanotherapy Blocks Both Hematogenous Metastasis and Lymphatic Metastasis. NANO LETTERS 2023; 23:3401-3411. [PMID: 37036326 DOI: 10.1021/acs.nanolett.3c00365] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Blood and lymph are two main pathways of tumor metastasis; however, hematogenous metastasis and lymphatic metastasis are difficult to inhibit simultaneously. Ferroptosis provides a new breakthrough for metastasis inhibition, but how to effectively trigger ferroptosis in tumor cells remains a major challenge. Metastatic tumor cells are prone to ferroptosis in blood, while they may be protected from ferroptosis in lymph. In this study, a nanoplatform DA/RSL3 was constructed for the intracellular codelivery of the polyunsaturated arachidonic acid (AA) and the GPX4 inhibitor RSL3, which could not only induce ferroptosis but also alleviate ferroptosis resistance. As a result, DA/RSL3 effectively triggered ferroptosis in tumor cells, thereby impairing the ability of tumor cells to metastasize in both blood and lymph. Furthermore, a fucoidan blocking strategy was proposed to maximize the efficacy of DA/RSL3. Fu+DA/RSL3 showed excellent efficacy in 4T1 tumor-bearing mice. This ferroptosis nanotherapy is promising for metastatic cancer treatment.
Collapse
Affiliation(s)
- Rong Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Miao Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xuan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Houqin Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yunxia Ye
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ziyi Mo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xuan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
105
|
Hartmann P, Trufa DI, Hohenberger K, Tausche P, Trump S, Mittler S, Geppert CI, Rieker RJ, Schieweck O, Sirbu H, Hartmann A, Finotto S. Contribution of serum lipids and cholesterol cellular metabolism in lung cancer development and progression. Sci Rep 2023; 13:5662. [PMID: 37024569 PMCID: PMC10079859 DOI: 10.1038/s41598-023-31575-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
Neoplasms of the lungs are the leading cause of cancer incidence and mortality worldwide. Although immunotherapy has increased the overall survival of patients with lung cancer, there is the need to improve this treatment. At this regard, blood lipid levels are thought to be linked to cancer risk and thus a preventive intervention through regulation of the nutrition of patients with lung cancer is gaining much attention. In this study, we therefore asked about the contribution of serum lipids and cholesterol cellular metabolism in lung cancer development and progression. We measured different serum lipids and analyzed cholesterol synthesis enzymes 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) and acetyl-coenzyme A cholesterol acetyltransferase 1 (ACAT1) as well as the cholesterol cellular export protein ATP-binding cassette (ABC) A-1 mRNA by quantitative PCR (qPCR) in the control and tumoral regions of post-surgery lung tissues to analyze the accumulation of cholesterol in cancer cells in a cohort of patients with lung adenocarcinoma (LUAD). We found that triglycerides in serum directly correlated with the body mass index (BMI) in patients with LUAD. By contrast, we found that high-density lipoprotein (HDL) cholesterol inversely correlated with the BMI, C-reactive protein (CRP) and overall survival and total cholesterol inversely correlated with the tumor diameter, serum CRP and overall survival in these LUAD patients. Functionally, the role of cholesterol is indispensable for the growth and development of normal animal cells where it is tightly regulated. Excess of cellular cholesterol regulated by HMGCR is converted to cholesteryl esters by the enzyme ACAT1 and exported extracellularly by the cholesterol transporter ABCA1. Here we found HMGCR and ACAT1 upregulated and ABCA1 downregulated in the lung's tumoral region of our LUAD cohort, indicating cholesterol dysregulated cellular export in lung tumor cells.
Collapse
Affiliation(s)
- Philipp Hartmann
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Denis I Trufa
- Department of Thoracic Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Katja Hohenberger
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Patrick Tausche
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Sonja Trump
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Susanne Mittler
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Ralf J Rieker
- Institute of Pathology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Oliver Schieweck
- Laboratory of Clinic Medicine, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91052, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
- Laboratories of Cellular and Molecular Lung Immunology, Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052, Erlangen, Germany.
| |
Collapse
|
106
|
El-Kenawi A, Berglund A, Estrella V, Zhang Y, Liu M, Putney RM, Yoder SJ, Johnson J, Brown J, Gatenby R. Elevated Methionine Flux Drives Pyroptosis Evasion in Persister Cancer Cells. Cancer Res 2023; 83:720-734. [PMID: 36480167 PMCID: PMC9978888 DOI: 10.1158/0008-5472.can-22-1002] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/29/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
Induction of cell death represents a primary goal of most anticancer treatments. Despite the efficacy of such approaches, a small population of "persisters" develop evasion strategies to therapy-induced cell death. While previous studies have identified mechanisms of resistance to apoptosis, the mechanisms by which persisters dampen other forms of cell death, such as pyroptosis, remain to be elucidated. Pyroptosis is a form of inflammatory cell death that involves formation of membrane pores, ion gradient imbalance, water inflow, and membrane rupture. Herein, we investigate mechanisms by which cancer persisters resist pyroptosis, survive, then proliferate in the presence of tyrosine kinase inhibitors (TKI). Lung, prostate, and esophageal cancer persister cells remaining after treatments exhibited several hallmarks indicative of pyroptosis resistance. The inflammatory attributes of persisters included chronic activation of inflammasome, STING, and type I interferons. Comprehensive metabolomic characterization uncovered that TKI-induced pyroptotic persisters display high methionine consumption and excessive taurine production. Elevated methionine flux or exogenous taurine preserved plasma membrane integrity via osmolyte-mediated effects. Increased dependency on methionine flux decreased the level of one carbon metabolism intermediate S-(5'-adenosyl)-L-homocysteine, a determinant of cell methylation capacity. The consequent increase in methylation potential induced DNA hypermethylation of genes regulating metal ion balance and intrinsic immune response. This enabled thwarting TKI resistance by using the hypomethylating agent decitabine. In summary, the evolution of resistance to pyroptosis can occur via a stepwise process of physical acclimation and epigenetic changes without existing or recurrent mutations. SIGNIFICANCE Methionine enables cancer cells to persist by evading pyroptotic osmotic lysis, which leads to genome-wide hypermethylation that allows persisters to gain proliferative advantages.
Collapse
Affiliation(s)
- Asmaa El-Kenawi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Anders Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Veronica Estrella
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Yonghong Zhang
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Min Liu
- Proteomics and Metabolomics Core Facility, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Ryan M Putney
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Sean J Yoder
- Molecular Genomics Core Facility, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Joseph Johnson
- Analytic Microscopy Core Facility, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Joel Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Robert Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida.,Department of Radiology, H. Lee Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
107
|
Lee J, Shin D, Roh JL. Lipid metabolism alterations and ferroptosis in cancer: Paving the way for solving cancer resistance. Eur J Pharmacol 2023; 941:175497. [PMID: 36621602 DOI: 10.1016/j.ejphar.2023.175497] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Cancer often perturbs lipid metabolism, which leads to the alteration of metabolism intermediates, contributing to their deregulated growth and metastasis. Alteration of lipid metabolism shifting to contain more polyunsaturated fatty acids (PUFAs) in membrane phospholipids (PLs) also leads to cancer therapy resistance. High amounts of PL-PUFAs render cancer cells more vulnerable to lipid peroxidation (LPO), predisposing them towards ferroptosis, a new form of iron-dependent oxidative regulated cell death. The commitment of cancer undergoing ferroptotic cell death depends on the adaptive lipidome remodeling, LPO patterns, and LPO scavenging ability in heterogeneous cancer cells. Ferroptosis is receiving attention in cancer research as treating cancers, altering membrane lipid homeostasis, and refractory from conventional therapies. Therefore, a better understanding of the molecular underpinning of lipid metabolism alterations may provide new opportunities for solving cancer resistance. This review intends to understand altered lipid metabolism in cancers and discuss lipid composition and metabolic processes associated with ferroptosis induction in cancers.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Daiha Shin
- Western Seoul Center, Korea Basic Science Institute, Seoul, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
108
|
Gabanella F, Onori A, Pisani C, Fiore M, Ferraguti G, Colizza A, de Vincentiis M, Ceccanti M, Inghilleri M, Corbi N, Passananti C, Di Certo MG. SMN Deficiency Destabilizes ABCA1 Expression in Human Fibroblasts: Novel Insights in Pathophysiology of Spinal Muscular Atrophy. Int J Mol Sci 2023; 24:ijms24032916. [PMID: 36769246 PMCID: PMC9917534 DOI: 10.3390/ijms24032916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The deficiency of survival motor neuron protein (SMN) causes spinal muscular atrophy (SMA), a rare neuromuscular disease that affects different organs. SMN is a key player in RNA metabolism regulation. An intriguing aspect of SMN function is its relationship with plasma membrane-associated proteins. Here, we provide a first demonstration that SMN affects the ATP-binding cassette transporter A1, (ABCA1), a membrane protein critically involved in cholesterol homeostasis. In human fibroblasts, we showed that SMN associates to ABCA1 mRNA, and impacts its subcellular distribution. Consistent with the central role of ABCA1 in the efflux of free cholesterol from cells, we observed a cholesterol accumulation in SMN-depleted human fibroblasts. These results were also confirmed in SMA type I patient-derived fibroblasts. These findings not only validate the intimate connection between SMN and plasma membrane-associated proteins, but also highlight a contribution of dysregulated cholesterol efflux in SMA pathophysiology.
Collapse
Affiliation(s)
- Francesca Gabanella
- CNR-Institute of Biochemistry and Cell Biology, Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
- Correspondence: (F.G.); (M.G.D.C.)
| | - Annalisa Onori
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Cinzia Pisani
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Marco Fiore
- CNR-Institute of Biochemistry and Cell Biology, Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Colizza
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco de Vincentiis
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Ceccanti
- Center for Rare Neuromuscular Diseases, Department of Human Neuroscience, Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy
| | - Maurizio Inghilleri
- Center for Rare Neuromuscular Diseases, Department of Human Neuroscience, Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy
| | - Nicoletta Corbi
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Claudio Passananti
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Maria Grazia Di Certo
- CNR-Institute of Biochemistry and Cell Biology, Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
- Correspondence: (F.G.); (M.G.D.C.)
| |
Collapse
|
109
|
Abstract
It has been 10 years since the concept of ferroptosis was put forward and research focusing on ferroptosis has been increasing continuously. Ferroptosis is driven by iron-dependent lipid peroxidation, which can be antagonized by glutathione peroxidase 4 (GPX4), ferroptosis inhibitory protein 1 (FSP1), dihydroorotate dehydrogenase (DHODH) and Fas-associated factor 1 (FAF1). Various cellular metabolic events, including lipid metabolism, can modulate ferroptosis sensitivity. It is worth noting that the reprogramming of lipid metabolism in cancer cells can promote the occurrence and development of tumors. The metabolic flexibility of cancer cells opens the possibility for the coordinated targeting of multiple lipid metabolic pathways to trigger cancer cells ferroptosis. In addition, cancer cells must obtain immortality, escape from programmed cell death including ferroptosis, to promote cancer progression, which provides new perspectives for improving cancer therapy. Targeting the vulnerability of ferroptosis has received attention as one of the significant possible strategies to treat cancer given its role in regulating tumor cell survival. We review the impact of iron and lipid metabolism on ferroptosis and the potential role of the crosstalk of lipid metabolism reprogramming and ferroptosis in antitumor immunity and sum up agents targeting lipid metabolism and ferroptosis for cancer therapy.
Collapse
|
110
|
Suresh S, Rabbie R, Garg M, Lumaquin D, Huang TH, Montal E, Ma Y, Cruz NM, Tang X, Nsengimana J, Newton-Bishop J, Hunter MV, Zhu Y, Chen K, de Stanchina E, Adams DJ, White RM. Identifying the Transcriptional Drivers of Metastasis Embedded within Localized Melanoma. Cancer Discov 2023; 13:194-215. [PMID: 36259947 PMCID: PMC9827116 DOI: 10.1158/2159-8290.cd-22-0427] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/25/2022] [Accepted: 10/14/2022] [Indexed: 01/16/2023]
Abstract
In melanoma, predicting which tumors will ultimately metastasize guides treatment decisions. Transcriptional signatures of primary tumors have been utilized to predict metastasis, but which among these are driver or passenger events remains unclear. We used data from the adjuvant AVAST-M trial to identify a predictive gene signature in localized tumors that ultimately metastasized. Using a zebrafish model of primary melanoma, we interrogated the top genes from the AVAST-M signature in vivo. This identified GRAMD1B, a cholesterol transfer protein, as a bona fide metastasis suppressor, with a majority of knockout animals rapidly developing metastasis. Mechanistically, excess free cholesterol or its metabolite 27-hydroxycholesterol promotes invasiveness via activation of an AP-1 program, which is associated with increased metastasis in humans. Our data demonstrate that the transcriptional seeds of metastasis are embedded within localized tumors, suggesting that early targeting of these programs can be used to prevent metastatic relapse. SIGNIFICANCE We analyzed human melanoma transcriptomics data to identify a gene signature predictive of metastasis. To rapidly test clinical signatures, we built a genetic metastasis platform in adult zebrafish and identified GRAMD1B as a suppressor of melanoma metastasis. GRAMD1B-associated cholesterol overload activates an AP-1 program to promote melanoma invasion. This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Shruthy Suresh
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roy Rabbie
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Manik Garg
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Dianne Lumaquin
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York
| | - Ting-Hsiang Huang
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily Montal
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yilun Ma
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York
| | - Nelly M Cruz
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xinran Tang
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
- Biochemistry and Structural Biology, Cellular and Developmental Biology and Molecular Biology Ph.D. Program, Weill Cornell Graduate School of Medical Sciences, New York, New York
| | - Jérémie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Miranda V. Hunter
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuxin Zhu
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kevin Chen
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David J. Adams
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Richard M. White
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
111
|
Yang K, Wang X, Song C, He Z, Wang R, Xu Y, Jiang G, Wan Y, Mei J, Mao W. The role of lipid metabolic reprogramming in tumor microenvironment. Theranostics 2023; 13:1774-1808. [PMID: 37064872 PMCID: PMC10091885 DOI: 10.7150/thno.82920] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/07/2023] [Indexed: 04/18/2023] Open
Abstract
Metabolic reprogramming is one of the most important hallmarks of malignant tumors. Specifically, lipid metabolic reprogramming has marked impacts on cancer progression and therapeutic response by remodeling the tumor microenvironment (TME). In the past few decades, immunotherapy has revolutionized the treatment landscape for advanced cancers. Lipid metabolic reprogramming plays pivotal role in regulating the immune microenvironment and response to cancer immunotherapy. Here, we systematically reviewed the characteristics, mechanism, and role of lipid metabolic reprogramming in tumor and immune cells in the TME, appraised the effects of various cell death modes (specifically ferroptosis) on lipid metabolism, and summarized the antitumor therapies targeting lipid metabolism. Overall, lipid metabolic reprogramming has profound effects on cancer immunotherapy by regulating the immune microenvironment; therefore, targeting lipid metabolic reprogramming may lead to the development of innovative clinical applications including sensitizing immunotherapy.
Collapse
Affiliation(s)
- Kai Yang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaokun Wang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Chenghu Song
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Zhao He
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Ruixin Wang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Yongrui Xu
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Guanyu Jiang
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton 13850, USA
- ✉ Corresponding authors: Wenjun Mao, M.D., Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China. E-mail: . Jie Mei, M.D., Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China. E-mail: . Yuan Wan, Ph.D., The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, No. 65 Murray Hill Rd., Binghamton, 13850, USA. E-mail:
| | - Jie Mei
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing, 210029, China
- ✉ Corresponding authors: Wenjun Mao, M.D., Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China. E-mail: . Jie Mei, M.D., Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China. E-mail: . Yuan Wan, Ph.D., The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, No. 65 Murray Hill Rd., Binghamton, 13850, USA. E-mail:
| | - Wenjun Mao
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
- ✉ Corresponding authors: Wenjun Mao, M.D., Department of Cardiothoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China. E-mail: . Jie Mei, M.D., Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China. E-mail: . Yuan Wan, Ph.D., The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, No. 65 Murray Hill Rd., Binghamton, 13850, USA. E-mail:
| |
Collapse
|
112
|
Lin J, Yang H, Zhang Y, Zou F, He H, Xie W, Zou Z, Liu R, Xu Q, Zhang J, Zhong G, Li Y, Tang Z, Deng Y, Cai S, Wang L, Huang Y, Zhuo Y, Jiang X, Zhong W. Ferrocene-Based Polymeric Nanoparticles Carrying Doxorubicin for Oncotherapeutic Combination of Chemotherapy and Ferroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205024. [PMID: 36398604 DOI: 10.1002/smll.202205024] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/01/2022] [Indexed: 06/16/2023]
Abstract
Mono-chemotherapy has significant side effects and unsatisfactory efficacy, limiting its clinical application. Therefore, a combination of multiple treatments is becoming more common in oncotherapy. Chemotherapy combined with the induction of ferroptosis is a potential new oncotherapy. Furthermore, polymeric nanoparticles (NPs) can improve the antitumor efficacy and decrease the toxicity of drugs. Herein, a polymeric NP, mPEG-b-PPLGFc@Dox, is synthesized to decrease the toxicity of doxorubicin (Dox) and enhance the efficacy of chemotherapy by combining it with the induction of ferroptosis. First, mPEG-b-PPLGFc@Dox is oxidized by endogenous H2 O2 and releases Dox, which leads to an increase of H2 O2 by breaking the redox balance. The Fe(II) group of ferrocene converts H2 O2 into ·OH, inducing subsequent ferroptosis. Furthermore, glutathione peroxidase 4, a biomarker of ferroptosis, is suppressed and the lipid peroxidation level is elevated in cells incubated with mPEG-b-PPLGFc@Dox compared to those treated with Dox alone, indicating ferroptosis induction by mPEG-b-PPLGFc@Dox. In vivo, the antitumor efficacy of mPEG-b-PPLGFc@Dox is higher than that of free Dox. Moreover, the loss of body weight in mice treated mPEG-b-PPLGFc@Dox is lower than in those treated with free Dox, indicating that mPEG-b-PPLGFc@Dox is less toxic than free Dox. In conclusion, mPEG-b-PPLGFc@Dox not only has higher antitumor efficacy but it reduces the damage to normal tissue.
Collapse
Affiliation(s)
- Jundong Lin
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Huikang Yang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Yixun Zhang
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Fen Zou
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Huichan He
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Wenjie Xie
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Zhihao Zou
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Ren Liu
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Qianfeng Xu
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Jie Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518000, China
| | - Guowei Zhong
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Yuejiao Li
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - ZhenFeng Tang
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Yulin Deng
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Shanghua Cai
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510230, China
| | - Linyao Wang
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Yugang Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510180, China
| | - Yangjia Zhuo
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Xinqing Jiang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Weide Zhong
- Department of Urology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| |
Collapse
|
113
|
Zhang C, Xu L, Li X, Chen Y, Shi T, Wang Q. LINC00460 Facilitates Cell Proliferation and Inhibits Ferroptosis in Breast Cancer Through the miR-320a/MAL2 Axis. Technol Cancer Res Treat 2023; 22:15330338231164359. [PMID: 36938678 PMCID: PMC10028644 DOI: 10.1177/15330338231164359] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Background: Emerging evidence suggests that long noncoding RNAs (lncRNAs) play an important role in the progression of multiple human cancers including breast cancer. In this study, we aimed to research novel functions of long intergenic noncoding RNA 460 (LINC00460) on cell proliferation and ferroptosis in breast cancer. Method: UALCAN, TANRIC, and GSE16446 data were used to analyze the expression of LINC00460 in breast cancer tissues. Furthermore, real-time quantitative PCR, western blot, cell proliferation assay, iron assay, and malondialdehyde (MDA) assay were applied to detect the function and mechanism of particular molecules. Results: The LINC00460 expression was significantly increased in breast cancer tissues compared with normal tissues. Importantly, patients with high LINC00460 expression showed a longer overall survival rate. LINC00460 knockdown markedly suppressed the proliferation of breast cancer cells and promoted ferroptosis. Mechanistic analysis revealed that LINC00460 promoted myelin and lymphocyte protein 2 (MAL2) expression by sponging miR-320a. Moreover, both miR-320a knockdown and MAL2 overexpression could reverse the effects of LINC00460 silencing on cell proliferation and ferroptosis. Conclusions: In summary, our results reveal an alternative mechanism by which breast cancer cells can acquire resistance to ferroptosis via the LINC00460/miR-320a/MAL2 axis.
Collapse
Affiliation(s)
- Chuanqiang Zhang
- Department of General Surgery, 627662The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Liang Xu
- Neonatal Department, 604074Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| | - Xiaowei Li
- Department of General Surgery, 627662The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yueqiu Chen
- Institute for Cardiovascular Science, 74566The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of 12582Soochow University, Suzhou, China
| | - Qiang Wang
- Department of General Surgery, 627662The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
114
|
Xia W, Wang H, Zhou X, Wang Y, Xue L, Cao B, Song J. The role of cholesterol metabolism in tumor therapy, from bench to bed. Front Pharmacol 2023; 14:928821. [PMID: 37089950 PMCID: PMC10117684 DOI: 10.3389/fphar.2023.928821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
Cholesterol and its metabolites have important biological functions. Cholesterol is able to maintain the physical properties of cell membrane, play an important role in cellular signaling, and cellular cholesterol levels reflect the dynamic balance between biosynthesis, uptake, efflux and esterification. Cholesterol metabolism participates in bile acid production and steroid hormone biosynthesis. Increasing evidence suggests a strict link between cholesterol homeostasis and tumors. Cholesterol metabolism in tumor cells is reprogrammed to differ significantly from normal cells, and disturbances of cholesterol balance also induce tumorigenesis and progression. Preclinical and clinical studies have shown that controlling cholesterol metabolism suppresses tumor growth, suggesting that targeting cholesterol metabolism may provide new possibilities for tumor therapy. In this review, we summarized the metabolic pathways of cholesterol in normal and tumor cells and reviewed the pre-clinical and clinical progression of novel tumor therapeutic strategy with the drugs targeting different stages of cholesterol metabolism from bench to bedside.
Collapse
Affiliation(s)
- Wenhao Xia
- Cancer Center of Peking University Third Hospital, Beijing, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hao Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Xiaozhu Zhou
- Department of Clinical Pharmacy, School of Pharmacy, Capital Medical University, Beijing, China
| | - Yan Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Third Hospital Institute of Medical Innovation and Research, Beijing, China
| | - Lixiang Xue
- Cancer Center of Peking University Third Hospital, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
- Third Hospital Institute of Medical Innovation and Research, Beijing, China
- *Correspondence: Lixiang Xue, ; Baoshan Cao, ; Jiagui Song,
| | - Baoshan Cao
- Cancer Center of Peking University Third Hospital, Beijing, China
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
- *Correspondence: Lixiang Xue, ; Baoshan Cao, ; Jiagui Song,
| | - Jiagui Song
- Cancer Center of Peking University Third Hospital, Beijing, China
- Third Hospital Institute of Medical Innovation and Research, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University as the Third Responsibility Unit of Song Jiagui, Beijing, China
- *Correspondence: Lixiang Xue, ; Baoshan Cao, ; Jiagui Song,
| |
Collapse
|
115
|
Adaptive activation of EFNB2/EPHB4 axis promotes post-metastatic growth of colorectal cancer liver metastases by LDLR-mediated cholesterol uptake. Oncogene 2023; 42:99-112. [PMID: 36376513 PMCID: PMC9816060 DOI: 10.1038/s41388-022-02519-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/15/2022]
Abstract
The microenvironment of distant organ plays vital roles in regulating tumor metastases. However, little is known about the crosstalk between metastasized tumor cells and target organs. Herein, we found that EFNB2 expression was upregulated in liver metastases (LM) of colorectal cancer (CRC), but not in pulmonary metastases (PM) or primary CRC tumors. EFNB2 played a tumor-promoting role in CRC LM in vitro and in vivo. Through forward signaling, EFNB2-promoted CRC LM by interacting with the EPHB4 receptor. EFNB2/EPHB4 axis enhances LDLR-mediated cholesterol uptake in CRC LM. Subsequently, EFNB2/EPHB4 axis promotes LDLR transcription by regulating STAT3 phosphorylation. Blocking LDLR reversed the role of the EFNB2/EPHB4 axis in promoting CRC LM. Using clinical data, survival analysis revealed that the survival time of patients with CRC LM was decreased in patients with high EFNB2 expression, compared with low EFNB2 expression. Inhibition of the EFNB2/EPHB4 axis markedly prolonged the survival time of BALB/c nude mice with CRC LM with a high cholesterol diet. These findings revealed a key step in the regulation of cholesterol uptake by EFNB2/EPHB4 axis and its tumor-promoting role in CRC LM.
Collapse
|
116
|
Association between diabetes and cancer. Current mechanistic insights into the association and future challenges. Mol Cell Biochem 2022:10.1007/s11010-022-04630-x. [PMID: 36565361 DOI: 10.1007/s11010-022-04630-x] [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: 05/19/2022] [Accepted: 11/30/2022] [Indexed: 12/25/2022]
Abstract
Compelling pieces of epidemiological, clinical, and experimental research have demonstrated that Diabetes mellitus (DM) is a major risk factor associated with increased cancer incidence and mortality in many human neoplasms. In the pathophysiology context of DM, many of the main classical actors are relevant elements that can fuel the different steps of the carcinogenesis process. Hyperglycemia, hyperinsulinemia, metabolic inflammation, and dyslipidemia are among the classic contributors to this association. Furthermore, new emerging actors have received particular attention in the last few years, and compelling data support that the microbiome, the epigenetic changes, the reticulum endoplasmic stress, and the increased glycolytic influx also play important roles in promoting the development of many cancer types. The arsenal of glucose-lowering therapeutic agents used for treating diabetes is wide and diverse, and a growing body of data raised during the last two decades has tried to clarify the contribution of therapeutic agents to this association. However, this research area remains controversial, because some anti-diabetic drugs are now considered as either promotors or protecting elements. In the present review, we intend to highlight the compelling epidemiological shreds of evidence that support this association, as well as the mechanistic contributions of many of these potential pathological mechanisms, some controversial points as well as future challenges.
Collapse
|
117
|
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X, Shi S. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol 2022; 15:174. [PMID: 36482419 PMCID: PMC9733270 DOI: 10.1186/s13045-022-01392-3] [Citation(s) in RCA: 268] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Many types of human cells self-destruct to maintain biological homeostasis and defend the body against pathogenic substances. This process, called regulated cell death (RCD), is important for various biological activities, including the clearance of aberrant cells. Thus, RCD pathways represented by apoptosis have increased in importance as a target for the development of cancer medications in recent years. However, because tumor cells show avoidance to apoptosis, which causes treatment resistance and recurrence, numerous studies have been devoted to alternative cancer cell mortality processes, namely necroptosis, pyroptosis, ferroptosis, and cuproptosis; these RCD modalities have been extensively studied and shown to be crucial to cancer therapy effectiveness. Furthermore, evidence suggests that tumor cells undergoing regulated death may alter the immunogenicity of the tumor microenvironment (TME) to some extent, rendering it more suitable for inhibiting cancer progression and metastasis. In addition, other types of cells and components in the TME undergo the abovementioned forms of death and induce immune attacks on tumor cells, resulting in enhanced antitumor responses. Hence, this review discusses the molecular processes and features of necroptosis, pyroptosis, ferroptosis, and cuproptosis and the effects of these novel RCD modalities on tumor cell proliferation and cancer metastasis. Importantly, it introduces the complex effects of novel forms of tumor cell death on the TME and the regulated death of other cells in the TME that affect tumor biology. It also summarizes the potential agents and nanoparticles that induce or inhibit novel RCD pathways and their therapeutic effects on cancer based on evidence from in vivo and in vitro studies and reports clinical trials in which RCD inducers have been evaluated as treatments for cancer patients. Lastly, we also summarized the impact of modulating the RCD processes on cancer drug resistance and the advantages of adding RCD modulators to cancer treatment over conventional treatments.
Collapse
Affiliation(s)
- Xuhui Tong
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Tang
- grid.452404.30000 0004 1808 0942Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Mingming Xiao
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Xu
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wang
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Zhang
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiang Liu
- grid.452404.30000 0004 1808 0942Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China. .,Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
118
|
Role of Neurite Outgrowth Inhibitor B Receptor in hepatic steatosis. Acta Histochem 2022; 124:151977. [DOI: 10.1016/j.acthis.2022.151977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022]
|
119
|
Yang X, Kawasaki NK, Min J, Matsui T, Wang F. Ferroptosis in heart failure. J Mol Cell Cardiol 2022; 173:141-153. [PMID: 36273661 PMCID: PMC11225968 DOI: 10.1016/j.yjmcc.2022.10.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/18/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
With its complicated pathobiology and pathophysiology, heart failure (HF) remains an increasingly prevalent epidemic that threatens global human health. Ferroptosis is a form of regulated cell death characterized by the iron-dependent lethal accumulation of lipid peroxides in the membrane system and is different from other types of cell death such as apoptosis and necrosis. Mounting evidence supports the claim that ferroptosis is mainly regulated by several biological pathways including iron handling, redox homeostasis, and lipid metabolism. Recently, ferroptosis has been identified to play an important role in HF induced by different stimuli such as myocardial infarction, myocardial ischemia reperfusion, chemotherapy, and others. Thus, it is of great significance to deeply explore the role of ferroptosis in HF, which might be a prerequisite to precise drug targets and novel therapeutic strategies based on ferroptosis-related medicine. Here, we review current knowledge on the link between ferroptosis and HF, followed by critical perspectives on the development and progression of ferroptotic signals and cardiac remodeling in HF.
Collapse
Affiliation(s)
- Xinquan Yang
- The Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Nicholas K Kawasaki
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, USA
| | - Junxia Min
- The Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Takashi Matsui
- Department of Anatomy, Biochemistry & Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoa, Honolulu, HI, USA.
| | - Fudi Wang
- The Fourth Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
120
|
Friedlová N, Zavadil Kokáš F, Hupp TR, Vojtěšek B, Nekulová M. IFITM protein regulation and functions: Far beyond the fight against viruses. Front Immunol 2022; 13:1042368. [PMID: 36466909 PMCID: PMC9716219 DOI: 10.3389/fimmu.2022.1042368] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Interferons (IFNs) are important cytokines that regulate immune responses through the activation of hundreds of genes, including interferon-induced transmembrane proteins (IFITMs). This evolutionarily conserved protein family includes five functionally active homologs in humans. Despite the high sequence homology, IFITMs vary in expression, subcellular localization and function. The initially described adhesive and antiproliferative or pro-oncogenic functions of IFITM proteins were diluted by the discovery of their antiviral properties. The large set of viruses that is inhibited by these proteins is constantly expanding, as are the possible mechanisms of action. In addition to their beneficial antiviral effects, IFITM proteins are often upregulated in a broad spectrum of cancers. IFITM proteins have been linked to most hallmarks of cancer, including tumor cell proliferation, therapeutic resistance, angiogenesis, invasion, and metastasis. Recent studies have described the involvement of IFITM proteins in antitumor immunity. This review summarizes various levels of IFITM protein regulation and the physiological and pathological functions of these proteins, with an emphasis on tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ted R. Hupp
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
121
|
Liu Y, Hu Y, Jiang Y, Bu J, Gu X. Targeting ferroptosis, the achilles' heel of breast cancer: A review. Front Pharmacol 2022; 13:1036140. [PMID: 36467032 PMCID: PMC9709426 DOI: 10.3389/fphar.2022.1036140] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/31/2022] [Indexed: 08/27/2023] Open
Abstract
Ferroptosis is referred as a novel type of cell death discovered in recent years with the feature of the accumulation of iron-dependent lipid reactive oxygen species. Breast cancer is one of the most common malignant cancers in women. There is increasing evidence that ferroptosis can inhibit breast cancer cell growth, improve the sensitivity of chemotherapy and radiotherapy and inhibit distant metastases. Therefore, ferroptosis can be regarded a new target for tumor suppression and may expand the landscape of clinical treatment of breast cancer. This review highlights the ferroptosis mechanism and its potential role in breast cancer treatment to explore new therapeutic strategies of breast cancer.
Collapse
Affiliation(s)
| | | | | | | | - Xi Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
122
|
Research progress on the role of cholesterol in hepatocellular carcinoma. Eur J Pharmacol 2022; 938:175410. [DOI: 10.1016/j.ejphar.2022.175410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
|
123
|
Gong D, Chen M, Wang Y, Shi J, Hou Y. Role of ferroptosis on tumor progression and immunotherapy. Cell Death Dis 2022; 8:427. [PMID: 36289191 PMCID: PMC9605952 DOI: 10.1038/s41420-022-01218-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022]
Abstract
Ferroptosis is triggered by intracellular iron leading to accumulation of lipid peroxidation consequent promotion of cell death. Cancer cell exhibits ability to evade ferroptosis by activation of antioxidant signaling pathways such as SLC7A11/GPX4 axis. In addition to transcriptional regulation on ferroptosis by NRF2, SREBP1, YAP, and p53, ferroptosis is modulated by ubiquitination or autophagic degradation. Moreover, zinc or Ca2+ could modulate ferroptosis by inducing lipid peroxidation and ferroptosis. Induction of ferroptosis enhances immune cell activity such as T cells or macrophages, which is associated with the release of DAMPs (damage-associated molecular patterns) and IFNγ. Therefore, combined immune checkpoint inhibitors with ferroptosis inducers effectively enhance antitumor immunotherapy, whereas induction of ferroptosis could impair T cell activity or survival, suggesting that rational combined therapy for cancer is essential. In this review, we discussed the regulatory role of ferroptosis on tumor progression and immunotherapy.
Collapse
Affiliation(s)
- Deting Gong
- grid.440785.a0000 0001 0743 511XSchool of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013 PR China
| | - Mingjun Chen
- grid.440785.a0000 0001 0743 511XSchool of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013 PR China
| | - Yuhan Wang
- grid.440785.a0000 0001 0743 511XSchool of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013 PR China
| | - Juanjuan Shi
- grid.440785.a0000 0001 0743 511XSchool of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013 PR China
| | - Yongzhong Hou
- grid.440785.a0000 0001 0743 511XSchool of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013 PR China
| |
Collapse
|
124
|
Zhu J, Wang H, Jiang X. mTORC1 beyond anabolic metabolism: Regulation of cell death. J Biophys Biochem Cytol 2022; 221:213609. [PMID: 36282248 PMCID: PMC9606688 DOI: 10.1083/jcb.202208103] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1), a multi-subunit protein kinase complex, interrogates growth factor signaling with cellular nutrient and energy status to control metabolic homeostasis. Activation of mTORC1 promotes biosynthesis of macromolecules, including proteins, lipids, and nucleic acids, and simultaneously suppresses catabolic processes such as lysosomal degradation of self-constituents and extracellular components. Metabolic regulation has emerged as a critical determinant of various cellular death programs, including apoptosis, pyroptosis, and ferroptosis. In this article, we review the expanding knowledge on how mTORC1 coordinates metabolic pathways to impinge on cell death regulation. We focus on the current understanding on how nutrient status and cellular signaling pathways connect mTORC1 activity with ferroptosis, an iron-dependent cell death program that has been implicated in a plethora of human diseases. In-depth understanding of the principles governing the interaction between mTORC1 and cell death pathways can ultimately guide the development of novel therapies for the treatment of relevant pathological conditions.
Collapse
Affiliation(s)
- Jiajun Zhu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,Correspondence to Jiajun Zhu:
| | - Hua Wang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Xuejun Jiang:
| |
Collapse
|
125
|
Sun K, Li C, Liao S, Yao X, Ouyang Y, Liu Y, Wang Z, Li Z, Yao F. Ferritinophagy, a form of autophagic ferroptosis: New insights into cancer treatment. Front Pharmacol 2022; 13:1043344. [PMID: 36339539 PMCID: PMC9635757 DOI: 10.3389/fphar.2022.1043344] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/12/2022] [Indexed: 09/24/2023] Open
Abstract
Ferritinophagy, a form of autophagy, is also an important part of ferroptosis, a type of regulated cell death resulting from abnormal iron metabolism involving the production of reactive oxygen species. As ferroptosis, autophagy and cancer have been revealed, ferritinophagy has attracted increasing attention in cancer development. In this review, we discuss the latest research progress on ferroptosis, autophagy-associated ferroptosis led by ferritinophagy, the regulators of ferritinophagy and promising cancer treatments that target ferritinophagy. Ferritinophagy is at the intersection of ferroptosis and autophagy and plays a significant role in cancer development. The discussed studies provide new insights into the mechanisms of ferritinophagy and promising related treatments for cancer.
Collapse
Affiliation(s)
- Kai Sun
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Chenyuan Li
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Shichong Liao
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinrui Yao
- School of Science, University of Sydney, Sydney, New South Wales, NSW, Australia
| | - Yang Ouyang
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Liu
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong Wang
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiyu Li
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Yao
- Department of Breast and Thyroid Surgery. Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
126
|
Tang Q, Liang B, Zhang L, Li X, Li H, Jing W, Jiang Y, Zhou F, Zhang J, Meng Y, Yang X, Yang H, Huang G, Zhao J. Enhanced CHOLESTEROL biosynthesis promotes breast cancer metastasis via modulating CCDC25 expression and neutrophil extracellular traps formation. Sci Rep 2022; 12:17350. [PMID: 36253427 PMCID: PMC9576744 DOI: 10.1038/s41598-022-22410-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/14/2022] [Indexed: 01/10/2023] Open
Abstract
Neutrophil extracellular traps (NETs) has been demonstrated to regulate the metastasis of breast cancer. In this study, we showed that de novo cholesterol biosynthesis induced by ASPP2 depletion in mouse breast cancer cell 4T1 and human breast cancer cell MDA-MB-231 promoted NETs formation in vitro, as well as in lung metastases in mice intravenously injected with ASPP2-deficient 4T1 cells. Simvastatin and berberine (BBR), cholesterol synthesis inhibitors, efficiently blocked ASPP2-depletion induced NETs formation. Cholesterol biosynthesis greatly enhanced Coiled-coil domain containing protein 25 (CCDC25) expression on cancer cells as well as in lung metastases. CCDC25 expression was co-localized with caveolin-1, a lipid raft molecule, and was damped by inhibitor of lipid rafts formation. Our data suggest that cholesterol biosynthesis promotes CCDC25 expression in a lipid raft-dependent manner. Clinically, the expression of CCDC25 was positively correlated with the expression of 3-hydroxy-3-methylglutaryl-CoAreductase (HMRCG), and citrullinated histone H3 (H3cit), in tissues from breast cancer patients. High expression of CCDC25 and HMGCR was related with worse prognosis in breast cancer patients. In conclusion, our study explores a novel mechanism for de novo cholesterol biosynthesis in the regulation of CCDC25 expression, NETs formation and breast cancer metastasis. Targeting cholesterol biosynthesis may be promising therapeutic strategies to treat breast cancer metastasis.
Collapse
Affiliation(s)
- Qiqi Tang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| | - Beibei Liang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Lisha Zhang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| | - Xuhui Li
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Hengyu Li
- grid.411525.60000 0004 0369 1599Changhai Hospital, Navy Military Medical University, Shanghai, 200438 China
| | - Wei Jing
- grid.411525.60000 0004 0369 1599Changhai Hospital, Navy Military Medical University, Shanghai, 200438 China
| | - Yingjie Jiang
- grid.411525.60000 0004 0369 1599Changhai Hospital, Navy Military Medical University, Shanghai, 200438 China
| | - Felix Zhou
- grid.4991.50000 0004 1936 8948Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - Jian Zhang
- grid.8547.e0000 0001 0125 2443Phase I Clinical Trial Center, Shanghai Cancer Center, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Yanchun Meng
- grid.8547.e0000 0001 0125 2443Phase I Clinical Trial Center, Shanghai Cancer Center, Fudan University, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Xinhua Yang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| | - Hao Yang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Gang Huang
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China
| | - Jian Zhao
- grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Science, 279Th Zhouzhu Road, Shanghai, 201318 China ,grid.507037.60000 0004 1764 1277Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201318 China ,grid.39436.3b0000 0001 2323 5732Shanghai University of Traditional Medicine, Shanghai, 201203 China
| |
Collapse
|
127
|
de Jesus Salazar-Estrada I, Kamath KS, Liu F. Precision Targeting of Endogenous Epidermal Growth Factor Receptor (EGFR) by Structurally Aligned Dual-Modifier Labeling. ACS Pharmacol Transl Sci 2022; 5:859-871. [PMID: 36268127 PMCID: PMC9578136 DOI: 10.1021/acsptsci.2c00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Indexed: 11/28/2022]
Abstract
Covalent modification of endogenous proteins by chemical probes is used for proteome-wide profiling of cellular protein function and drug discovery. However, probe selectivity in the complex cellular environment is a challenge, and new probes with better target selectivity are continuously needed. On the basis of the success of monocovalent activity-based and reactivity-based probes, an approach of structurally aligned dual-modifier labeling (SADL) was investigated here on its potential in improving target precision. Two reactive groups, based on the acrylamide and NHS ester chemistry, were linked with structural alignment to be under the same anilinoquinazoline ligand-directive for targeting the epidermal growth factor receptor (EGFR) protein kinase as the model system for proteome-wide profiling. The SADL approach was compared with its monocovalent precursors in a label-free MaxLFQ workflow using MDA-MB-468 triple negative breast cancer cells. The dual-modifier probe consistently showed labeling of EGFR with improved precision over both monocovalent precursors under various controls. The workflow also labeled endogenous USP34 and PKMYT1 with high selectivity. Precision labeling with two covalent modifiers under a common ligand directive may broaden protein identification opportunities in the native environment to complement genetic and antibody-based approaches for elucidating biological or disease mechanisms, as well as accelerating drug target discovery.
Collapse
Affiliation(s)
| | | | - Fei Liu
- School
of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
128
|
Ferulic Acid Mitigates Growth and Invasion of Esophageal Squamous Cell Carcinoma through Inducing Ferroptotic Cell Death. DISEASE MARKERS 2022; 2022:4607966. [PMID: 36267458 PMCID: PMC9578864 DOI: 10.1155/2022/4607966] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022]
Abstract
Objective Ferroptosis is an iron- and ROS-dependent form of cell death initiated by lipid peroxidation. The rapidly developing study of ferroptosis has facilitated its application in cancer therapeutics. The current study is aimed at investigating the functional property of ferulic acid (FA, a phenolic acid substance) on inducing ferroptosis in antiesophageal squamous cell carcinoma (ESCC). Methods ESCC cells were administrated with gradient doses of FA or with ferroptosis inhibitor deferoxamine. Cellular growth was measured with CCK-8 and colony formation experiments. LDH, caspase-3, MDA, SOD, GSH, and iron were assayed with corresponding kits. Apoptotic level was evaluated through Annexin V-FITC apoptosis staining, with migration and invasion utilizing Transwell assays. Through quantitative RT-PCR, angiogenesis-relevant genes VEGFA and PDGFB were detected. ROS generation was measured via DCFH-DA probe. Immunoblotting was conducted for monitoring ACSL4, SLC7A11, HO-1, and GPX4. Results FA administration observably mitigated cellular viability and colony formation capacity and motivated LDH release, caspase-3 activity, and apoptosis in EC-1 and TE-4 cells. In addition, migration and invasion together with angiogenesis of ESCC cells were restraint by FA. FA exposure led to the increase of MDA content, ROS production, and iron load as well as the reduction of SOD activity and GSH content. Also, FA augmented the activities of ACSL4 and HO-1, with lessening SLC7A11 and GPX4. Nonetheless, deferoxamine restrained the effect of FA on ESCC ferroptosis. Conclusion Altogether, FA may act as a ferroptosis inducer and thus attenuates cell growth and invasion of ESCC, which boosts the clinical application of FA in ESCC therapeutics.
Collapse
|
129
|
Cui Y, Chen Z, Pan B, Chen T, Ding H, Li Q, Wan L, Luo G, Sun L, Ding C, Yang J, Tong X, Zhao J. Neddylation pattern indicates tumor microenvironment characterization and predicts prognosis in lung adenocarcinoma. Front Cell Dev Biol 2022; 10:979262. [PMID: 36176276 PMCID: PMC9513323 DOI: 10.3389/fcell.2022.979262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Lung adenocarcinoma (LUAD) is the most common type of lung cancer with a complex tumor microenvironment. Neddylation, as a type of post-translational modification, plays a vital role in the development of LUAD. To date, no study has explored the potential of neddylation-associated genes for LUAD classification, prognosis prediction, and treatment response evaluation.Methods: Seventy-six neddylation-associated prognostic genes were identified by Univariate Cox analysis. Patients with LUAD were classified into two patterns based on unsupervised consensus clustering analysis. In addition, a 10-gene prognostic signature was constructed using LASSO-Cox and a multivariate stepwise regression approach.Results: Substantial differences were observed between the two patterns of LUAD in terms of prognosis. Compared with neddylation cluster2, neddylation cluster1 exhibited low levels of immune infiltration that promote tumor progression. Additionally, the neddylation-related risk score correlated with clinical parameters and it can be a good predictor of patient outcomes, gene mutation levels, and chemotherapeutic responses.Conclusion: Neddylation patterns can distinguish tumor microenvironment and prognosis in patients with LUAD. Prognostic signatures based on neddylation-associated genes can predict patient outcomes and guide personalized treatment.
Collapse
Affiliation(s)
- Yuan Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bin Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tong Chen
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qifan Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Wan
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, China
| | - Gaomeng Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lang Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Jian Yang, ; Xin Tong, ; Jun Zhao,
| | - Xin Tong
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Jian Yang, ; Xin Tong, ; Jun Zhao,
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Jian Yang, ; Xin Tong, ; Jun Zhao,
| |
Collapse
|
130
|
Yu YQ, Gamez-Belmonte R, Patankar JV, Liebing E, Becker C. The Role of Programmed Necrosis in Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14174295. [PMID: 36077828 PMCID: PMC9455009 DOI: 10.3390/cancers14174295] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Necrosis is a type of cell death characterized by plasma membrane rupture and the induction of inflammation. This review focuses on colorectal cancer and outlines the role of programmed necrosis in tumor development. Potential strategies for anti-tumor treatment via targeting programmed necrosis are also discussed. Abstract For quite a long time, necrosis was considered a chaotic and unorganized form of cell death. However, studies conducted during the past few decades unveiled multiple types of programmed necrosis, such as necroptosis, pyroptosis and ferroptosis. These types of programmed necrosis have been shown to play crucial roles in mediating pathological processes, including tumorigenesis. Almost all key mediators, such as RIPK3 and MLKL in necroptosis, GSDMD and caspase 1/11 in pyroptosis and GPX4 in ferroptosis, are highly expressed in intestinal epithelial cells (IECs). An aberrant increase or decrease in programmed necrosis in IECs has been connected to intestinal disorders. Here, we review the pathways of programmed necrosis and the specific consequences of regulated necrosis in colorectal cancer (CRC) development. Translational aspects of programmed necrosis induction as a novel therapeutic alternative against CRC are also discussed.
Collapse
Affiliation(s)
- Yu-Qiang Yu
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Reyes Gamez-Belmonte
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Jay V. Patankar
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Eva Liebing
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-85-35-886
| |
Collapse
|
131
|
Wang Q, Cao Y, Shen L, Xiao T, Cao R, Wei S, Tang M, Du L, Wu H, Wu B, Yu Y, Wang S, Wen M, OuYang B. Regulation of PD-L1 through direct binding of cholesterol to CRAC motifs. SCIENCE ADVANCES 2022; 8:eabq4722. [PMID: 36026448 PMCID: PMC9417176 DOI: 10.1126/sciadv.abq4722] [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: 04/11/2022] [Accepted: 07/12/2022] [Indexed: 05/24/2023]
Abstract
Cholesterol, an essential molecule for cell structure, function, and viability, plays crucial roles in the development, progression, and survival of cancer cells. Earlier studies have shown that cholesterol-lowering drugs can inhibit the high expression of programmed-death ligand 1 (PD-L1) that contributes to immunoevasion in cancer cells. However, the regulatory mechanism of cell surface PD-L1 abundance by cholesterol is still controversial. Here, using nuclear magnetic resonance and biochemical techniques, we demonstrated that cholesterol can directly bind to the transmembrane domain of PD-L1 through two cholesterol-recognition amino acid consensus (CRAC) motifs, forming a sandwich-like architecture and stabilizing PD-L1 to prevent downstream degradation. Mutations at key binding residues prohibit PD-L1-cholesterol interactions, decreasing the cellular abundance of PD-L1. Our results reveal a unique regulatory mechanism that controls the stability of PD-L1 in cancer cells, providing an alternative method to overcome PD-L1-mediated immunoevasion in cancers.
Collapse
Affiliation(s)
- Qian Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunlei Cao
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijuan Shen
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Taoran Xiao
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruiyu Cao
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shukun Wei
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Tang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingyu Du
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongyi Wu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Wu
- National Facility for Protein Science Shanghai, ZhangJiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yang Yu
- National Facility for Protein Science Shanghai, ZhangJiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Shuqing Wang
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Maorong Wen
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bo OuYang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
132
|
Tao Y, Wei L, You H. Ferroptosis-related gene signature predicts the clinical outcome in pediatric acute myeloid leukemia patients and refines the 2017 ELN classification system. Front Mol Biosci 2022; 9:954524. [PMID: 36032681 PMCID: PMC9403410 DOI: 10.3389/fmolb.2022.954524] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: The prognostic roles of ferroptosis-related mRNAs (FG) and lncRNAs (FL) in pediatric acute myeloid leukemia (P-AML) patients remain unclear. Methods: RNA-seq and clinical data of P-AML patients were downloaded from the TARGET project. Cox and LASSO regression analyses were performed to identify FG, FL, and FGL (combination of FG and FL) prognostic models, and their performances were compared. Tumor microenvironment, functional enrichment, mutation landscape, and anticancer drug sensitivity were analyzed. Results: An FGL model of 22 ferroptosis-related signatures was identified as an independent parameter, and it showed performance better than FG, FL, and four additional public prognostic models. The FGL model divided patients in the discovery cohort (N = 145), validation cohort (N = 111), combination cohort (N = 256), and intermediate-risk group (N = 103) defined by the 2017 European LeukemiaNet (ELN) classification system into two groups with distinct survival. The high-risk group was enriched in apoptosis, hypoxia, TNFA signaling via NFKB, reactive oxygen species pathway, oxidative phosphorylation, and p53 pathway and associated with low immunity, while patients in the low-risk group may benefit from anti-TIM3 antibodies. In addition, patients within the FGL high-risk group might benefit from treatment using SB505124_1194 and JAK_8517_1739. Conclusion: Our established FGL model may refine and provide a reference for clinical prognosis judgment and immunotherapies for P-AML patients.
Collapse
Affiliation(s)
- Yu Tao
- Department of Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Li Wei
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, China
| | - Hua You
- Department of Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Hua You,
| |
Collapse
|
133
|
Gu J, Zhu N, Li HF, Zhao TJ, Zhang CJ, Liao DF, Qin L. Cholesterol homeostasis and cancer: a new perspective on the low-density lipoprotein receptor. Cell Oncol 2022; 45:709-728. [PMID: 35864437 DOI: 10.1007/s13402-022-00694-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Disturbance of cholesterol homeostasis is considered as one of the manifestations of cancer. Cholesterol plays an essential role in the pleiotropic functions of cancer cells, including mediating membrane trafficking, intracellular signal transduction, and production of hormones and steroids. As a single transmembrane receptor, the low-density lipoprotein receptor (LDLR) can participate in intracellular cholesterol uptake and regulate cholesterol homeostasis. It has recently been found that LDLR is aberrantly expressed in a broad range of cancers, including colon cancer, prostate cancer, lung cancer, breast cancer and liver cancer. LDLR has also been found to be involved in various signaling pathways, such as the MAPK, NF-κB and PI3K/Akt signaling pathways, which affect cancer cells and their surrounding microenvironment. Moreover, LDLR may serve as an independent prognostic factor for lung cancer, breast cancer and pancreatic cancer, and is closely related to the survival of cancer patients. However, the role of LDLR in some cancers, such as prostate cancer, remains controversial. This may be due to the lack of normal feedback regulation of LDLR expression in cancer cells and the severe imbalance between LDLR-mediated cholesterol uptake and de novo biosynthesis of cholesterol. CONCLUSIONS The imbalance of cholesterol homeostasis caused by abnormal LDLR expression provides new therapeutic opportunities for cancer. LDLR interferes with the occurrence and development of cancer by modulating cholesterol homeostasis and may become a novel target for the development of anti-cancer drugs. Herein, we systematically review the contribution of LDLR to cancer progression, especially its dysregulation and underlying mechanism in various malignancies. Besides, potential targeting and immunotherapeutic options are proposed.
Collapse
Affiliation(s)
- Jia Gu
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Hong-Fang Li
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Tan-Jun Zhao
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chan-Juan Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Duan-Fang Liao
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Li Qin
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
134
|
Gu J, Zhu N, Li HF, Zhang CJ, Gong YZ, Liao DF, Qin L. Ezetimibe and Cancer: Is There a Connection? Front Pharmacol 2022; 13:831657. [PMID: 35924044 PMCID: PMC9340271 DOI: 10.3389/fphar.2022.831657] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
The high level of serum cholesterol caused by the excessive absorption of cholesterol can lead to hypercholesteremia, thus promoting the occurrence and development of cancer. Ezetimibe is a drug that reduces cholesterol absorption and has been widely used for the treatment of patients with high circulating cholesterol levels for many years. Mechanistically, ezetimibe works by binding to NPC1L1, which is a key mediator of cholesterol absorption. Accumulating data from preclinical models have shown that ezetimibe alone could inhibit the development and progression of cancer through a variety of mechanisms, including anti-angiogenesis, stem cell suppression, anti-inflammation, immune enhancement and anti-proliferation. In the past decade, there has been heated discussion on whether ezetimibe combined with statins will increase the risk of cancer. At present, more and more evidence shows that ezetimibe does not increase the risk of cancers, which supports the role of ezetimibe in anti-cancer. In this review, we discussed the latest progress in the anti-cancer properties of ezetimibe and elucidated its underlying molecular mechanisms. Finally, we highlighted the potential of ezetimibe as a therapeutic agent in future cancer treatment and prevention.
Collapse
Affiliation(s)
- Jia Gu
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Hong-Fang Li
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Chan-Juan Zhang
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Yong-Zhen Gong
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
| | - Li Qin
- Laboratory of Stem Cell Regulation With Chinese Medicine and its Application, HunanUniversity of Chinese Medicine, Changsha, China
- Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Changsha, China
- Hunan Province Engineering Research Center of Bioactive Substance Discovery of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- *Correspondence: Li Qin,
| |
Collapse
|
135
|
Li P, Zhang L, Guo Z, Kang Q, Chen C, Liu X, Ma Q, Zhang J, Hu Y, Wang T. Epimedium koreanum Nakai-Induced Liver Injury-A Mechanistic Study Using Untargeted Metabolomics. Front Pharmacol 2022; 13:934057. [PMID: 35910368 PMCID: PMC9326364 DOI: 10.3389/fphar.2022.934057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Epimedii Folium is widely used worldwide as an herbal supplement, and the risk of its induced liver damage has emerged in recent years. Our preliminary study has found that, among several Epimedii Folium species specified in the Chinese Pharmacopoeia, Epimedium koreanum Nakai has a more severe propensity for hepatotoxicity. However, the mechanism of hepatotoxicity of Epimedium koreanum Nakai is still unclear. In this study, untargeted metabolomics was performed to analyze the serum and liver tissue to explore the mechanism of hepatotoxicity of Epimedium koreanum Nakai. The results of experiments in vivo showed that, after 28 days of exposure to Epimedium koreanum Nakai ethanol extract (EEE), the liver weight, levels of AST, ALP, TBIL, etc. in serum of rats in the EEE group were significantly increased, as well as severe cytoplasmic vacuolation appeared in the liver tissue, which suggested that EEE has significant hepatotoxicity. Subsequently, the results of metabolomics revealed significant changes in the metabolic profile in the liver and serum of rats after EEE exposure, in which metabolites in serum such as flavin mononucleotide, phenylacetylglycine, glutathione, l-tryptophan, and sphingomyelin were able to accurately identify liver injury caused by EEE and could be used as serum markers to reflect EEE-induced liver injury. The KEGG pathway enrichment analysis revealed that EEE caused extensive effects on rats' metabolic pathways. Some of the most affected pathways included glutathione metabolism, glutamate metabolism pathway, primary bile acid biosynthesis pathway, and sphingolipid metabolism pathway, which were all directed to the biological process of ferroptosis. Then, the main markers related to ferroptosis in the liver were examined, and the results demonstrated that the content of malondialdehyde was significantly increased, the activity of superoxide dismutase was significantly reduced, the ferroptosis inhibitory proteins GPX4 and System xc - were significantly downregulated, and the ferroptosis-promoting protein ACSL4 was significantly up-regulated. Judging from these results, we concluded that the mechanism of hepatotoxicity of Epimedium koreanum Nakai was probably related to the induction of ferroptosis in hepatocytes.
Collapse
Affiliation(s)
| | - Lin Zhang
- Beijing Institute of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | | | | | | | | | | | | | | | - Ting Wang
- Beijing Institute of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
136
|
Ma L, Vidana Gamage HE, Tiwari S, Han C, Henn MA, Krawczynska N, Dibaeinia P, Koelwyn GJ, Das Gupta A, Bautista Rivas RO, Wright CL, Xu F, Moore KJ, Sinha S, Nelson ER. The Liver X Receptor Is Selectively Modulated to Differentially Alter Female Mammary Metastasis-associated Myeloid Cells. Endocrinology 2022; 163:bqac072. [PMID: 35569056 PMCID: PMC9188661 DOI: 10.1210/endocr/bqac072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Indexed: 11/19/2022]
Abstract
Dysregulation of cholesterol homeostasis is associated with many diseases such as cardiovascular disease and cancer. Liver X receptors (LXRs) are major upstream regulators of cholesterol homeostasis and are activated by endogenous cholesterol metabolites such as 27-hydroxycholesterol (27HC). LXRs and various LXR ligands such as 27HC have been described to influence several extra-hepatic biological systems. However, disparate reports of LXR function have emerged, especially with respect to immunology and cancer biology. This would suggest that, similar to steroid nuclear receptors, the LXRs can be selectively modulated by different ligands. Here, we use RNA-sequencing of macrophages and single-cell RNA-sequencing of immune cells from metastasis-bearing murine lungs to provide evidence that LXR satisfies the 2 principles of selective nuclear receptor modulation: (1) different LXR ligands result in overlapping but distinct gene expression profiles within the same cell type, and (2) the same LXR ligands differentially regulate gene expression in a highly context-specific manner, depending on the cell or tissue type. The concept that the LXRs can be selectively modulated provides the foundation for developing precision pharmacology LXR ligands that are tailored to promote those activities that are desirable (proimmune), but at the same time minimizing harmful side effects (such as elevated triglyceride levels).
Collapse
Affiliation(s)
- Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Hashni Epa Vidana Gamage
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Srishti Tiwari
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Chaeyeon Han
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Madeline A Henn
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Natalia Krawczynska
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Payam Dibaeinia
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Graeme J Koelwyn
- NYU Cardiovascular Research Center, Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Anasuya Das Gupta
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Rafael Ovidio Bautista Rivas
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Chris L Wright
- Roy J. Carver Biotechnology Center DNA Services, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Fangxiu Xu
- Roy J. Carver Biotechnology Center DNA Services, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Kathryn J Moore
- NYU Cardiovascular Research Center, Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University School of Medicine, New York, NY 10032, USA
| | - Saurabh Sinha
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
137
|
Wu J, Zhang L, Wu S, Liu Z. Ferroptosis: Opportunities and Challenges in Treating Endometrial Cancer. Front Mol Biosci 2022; 9:929832. [PMID: 35847989 PMCID: PMC9284435 DOI: 10.3389/fmolb.2022.929832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022] Open
Abstract
Ferroptosis, a new way of cell death, is involved in many cancers. A growing number of studies have focused on the unique role of ferroptosis on endometrial cancer. In this study, we made a comprehensive review of the relevant articles published to get deep insights in the association of ferroptosis with endometrial cancer and to present a summary of the roles of different ferroptosis-associated genes. Accordingly, we made an evaluation of the relationships between the ferroptosis-associated genes and TNM stage, tumor grade, histological type, primary therapy outcome, invasion and recurrence of tumor, and accessing the different prognosis molecular typing based on ferroptosis-associated genes. In addition, we presented an introduction of the common drugs, which targeted ferroptosis in endometrial cancer. In so doing, we clarified the opportunities and challenges of ferroptosis activator application in treating endometrial cancer, with a view to provide a novel approach to the disease.
Collapse
Affiliation(s)
- Jianfa Wu
- Department of Gynecology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Gynecology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Li Zhang
- Department of Gynecology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Gynecology, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Suqin Wu
- Department of Gynecology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Gynecology, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Suqin Wu, ; Zhou Liu,
| | - Zhou Liu
- Department of Gynecology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Gynecology, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Suqin Wu, ; Zhou Liu,
| |
Collapse
|
138
|
Abstract
Ferroptosis is an iron-dependent form of regulated cell death that is triggered by the toxic build-up of lipid peroxides on cellular membranes. In recent years, ferroptosis has garnered enormous interest in cancer research communities, partly because it is a unique cell death modality that is mechanistically and morphologically different from other forms of cell death, such as apoptosis, and therefore holds great potential for cancer therapy. In this Review, we summarize the current understanding of ferroptosis-inducing and ferroptosis defence mechanisms, dissect the roles and mechanisms of ferroptosis in tumour suppression and tumour immunity, conceptualize the diverse vulnerabilities of cancer cells to ferroptosis, and explore therapeutic strategies for targeting ferroptosis in cancer.
Collapse
Affiliation(s)
- Guang Lei
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
139
|
Kolbrink B, von Samson-Himmelstjerna FA, Messtorff ML, Riebeling T, Nische R, Schmitz J, Bräsen JH, Kunzendorf U, Krautwald S. Vitamin K1 inhibits ferroptosis and counteracts a detrimental effect of phenprocoumon in experimental acute kidney injury. Cell Mol Life Sci 2022; 79:387. [PMID: 35763128 PMCID: PMC9239973 DOI: 10.1007/s00018-022-04416-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
Ferroptosis, a type of iron-dependent programmed cell death distinct from apoptosis, necroptosis, and other types of cell death, is characterized by lipid peroxidation, reactive oxygen species production, and mitochondrial dysfunction. Accumulating evidence has highlighted vital roles for ferroptosis in multiple diseases, including acute kidney injury. Therefore, ferroptosis has become a major focus for translational research. However, despite its involvement in pathological conditions, there are no pharmacologic inhibitors of ferroptosis in clinical use. In the context of drug repurposing, a strategy for identifying new uses for approved drugs outside the original medical application, we discovered that vitamin K1 is an efficient inhibitor of ferroptosis. Our findings are strengthened by the fact that the vitamin K antagonist phenprocoumon significantly exacerbated ferroptotic cell death in vitro and also massively worsened the course of acute kidney injury in vivo, which is of utmost clinical importance. We therefore assign vitamin K1 a novel role in preventing ferroptotic cell death in acute tubular necrosis during acute kidney injury. Since the safety, tolerability, pharmacokinetics, and pharmacodynamics of vitamin K1 formulations are well documented, this drug is primed for clinical application, and provides a new strategy for pharmacological control of ferroptosis and diseases associated with this mode of cell death.
Collapse
Affiliation(s)
- Benedikt Kolbrink
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Campus Kiel, Fleckenstr. 4, 24105, Kiel, Germany
| | | | - Maja Lucia Messtorff
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Campus Kiel, Fleckenstr. 4, 24105, Kiel, Germany
| | - Theresa Riebeling
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Campus Kiel, Fleckenstr. 4, 24105, Kiel, Germany
| | - Raphael Nische
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Campus Kiel, Fleckenstr. 4, 24105, Kiel, Germany
| | - Jessica Schmitz
- Nephropathology Unit, Institute of Pathology, University of Hannover, 30625, Hannover, Germany
| | - Jan Hinrich Bräsen
- Nephropathology Unit, Institute of Pathology, University of Hannover, 30625, Hannover, Germany
| | - Ulrich Kunzendorf
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Campus Kiel, Fleckenstr. 4, 24105, Kiel, Germany
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Campus Kiel, Fleckenstr. 4, 24105, Kiel, Germany.
| |
Collapse
|
140
|
Li Y, Yang Y, Yang Y. Multifaceted Roles of Ferroptosis in Lung Diseases. Front Mol Biosci 2022; 9:919187. [PMID: 35813823 PMCID: PMC9263225 DOI: 10.3389/fmolb.2022.919187] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/20/2022] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a distinct type of programmed cell death (PCD) that depends on iron and is characterized by the accumulation of intracellular iron, exhaustion of glutathione, deactivation of glutathione peroxidase, and promotion of lipid peroxidation. Recently, accumulated investigations have demonstrated that ferroptosis is strongly correlated with the initiation and development of many lung diseases. In this review, we summarized the contribution of ferroptosis to the pathologic process of lung diseases, namely, obstructive lung diseases (chronic obstructive pulmonary disease, asthma, and cystic fibrosis), interstitial lung diseases (pulmonary fibrosis of different causes), pulmonary diseases of vascular origin (ischemia-reperfusion injury and pulmonary hypertension), pulmonary infections (bacteria, viruses, and fungi), acute lung injury, acute respiratory distress syndrome, obstructive sleep apnea, pulmonary alveolar proteinosis, and lung cancer. We also discussed the therapeutic potential of targeting ferroptosis for these lung diseases.
Collapse
Affiliation(s)
- Yi Li
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Yang
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Yongfeng Yang
- Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, China
- Precision Medicine Key Laboratory, West China Hospital, Sichuan University, Chengdu, China
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yongfeng Yang,
| |
Collapse
|
141
|
Liang D, Minikes AM, Jiang X. Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell 2022; 82:2215-2227. [PMID: 35390277 DOI: 10.1016/j.molcel.2022.03.022] [Citation(s) in RCA: 392] [Impact Index Per Article: 196.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
Ferroptosis, a newly emerged form of regulated necrotic cell death, has been demonstrated to play an important role in multiple diseases including cancer, neurodegeneration, and ischemic organ injury. Mounting evidence also suggests its potential physiological function in tumor suppression and immunity. The execution of ferroptosis is driven by iron-dependent phospholipid peroxidation. As such, the metabolism of biological lipids regulates ferroptosis via controlling phospholipid peroxidation, as well as various other cellular processes relevant to phospholipid peroxidation. In this review, we provide a comprehensive analysis by focusing on how lipid metabolism impacts the initiation, propagation, and termination of phospholipid peroxidation; how multiple signal transduction pathways communicate with ferroptosis via modulating lipid metabolism; and how such intimate cross talk of ferroptosis with lipid metabolism and related signaling pathways can be exploited for the development of rational therapeutic strategies.
Collapse
Affiliation(s)
- Deguang Liang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY 10065, USA
| | - Alexander M Minikes
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Ave., New York, NY 10065, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY 10065, USA.
| |
Collapse
|
142
|
Xu L, Huang X, Lou Y, Xie W, Zhao H. Regulation of apoptosis, autophagy and ferroptosis by non‑coding RNAs in metastatic non‑small cell lung cancer (Review). Exp Ther Med 2022; 23:352. [PMID: 35493430 PMCID: PMC9019694 DOI: 10.3892/etm.2022.11279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/10/2022] [Indexed: 11/06/2022] Open
Abstract
Non-small cell lung cancer (NSCLC), a common type of cancer worldwide, is normally associated with a poor prognosis. It is difficult to treat successfully as it often metastasizes into brain or bone. Methods to facilitate the induction of effective programmed cell death (PCD) in NSCLC cells to reverse drug resistance, or to inhibit the invasion and migration of NSCLC cells, are currently under investigation. The present study summarized the regulatory functions of PCD, including apoptosis, autophagy and ferroptosis, in the context of NSCLC metastasis. It further summarized how regulatory agents, including long non-coding RNAs, circular RNAs and microRNAs, regulate PCD during the metastasis of NSCLC and characterized new potential diagnostic biomarkers of NSCLC metastasis.
Collapse
Affiliation(s)
- Lei Xu
- Department of Orthopedics, Chengdu Seventh People's Hospital, Chengdu, Sichuan 610213, P.R. China
| | - Xin Huang
- Department of Orthopedics, Chengdu Seventh People's Hospital, Chengdu, Sichuan 610213, P.R. China
| | - Yan Lou
- Department of Orthopedic Oncology, Spine Tumor Center, Changzheng Hospital, Naval Military Medical University, Shanghai 200003, P.R. China
| | - Wei Xie
- Department of Orthopedics, Chengdu Seventh People's Hospital, Chengdu, Sichuan 610213, P.R. China
| | - Hangyu Zhao
- Department of Orthopedics, Chengdu Seventh People's Hospital, Chengdu, Sichuan 610213, P.R. China
| |
Collapse
|
143
|
Anisman H, Kusnecov AW. Dietary components associated with being overweight, having obesity, and cancer. Cancer 2022. [DOI: 10.1016/b978-0-323-91904-3.00018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
144
|
Xu T, Peng B, Liu M, Liu Q, Yang J, Qu M, Liu N, Lin L, Wu J. Favorable Genotypes of Type III Interferon Confer Risk of Dyslipidemia in the Population With Obesity. Front Endocrinol (Lausanne) 2022; 13:871352. [PMID: 35784542 PMCID: PMC9243353 DOI: 10.3389/fendo.2022.871352] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Studies have indicated that the chronic state of inflammation caused by obesity leads to dyslipidemia. However, how the polymorphisms involved in these inflammatory pathways affect the lipid metabolism in people with obesity is poorly understood. We investigated the associations of inflammation-related gene polymorphisms with dyslipidemia in individuals with obesity living in China. METHODS This case-control study in a population with obesity involved 194 individuals with dyslipidemia and 103 individuals without dyslipidemia. Anthropometric indices of obesity, fasting plasma glucose, blood pressure, blood lipids, and C-reactive protein were evaluated. The genes we tested were IL6 (interleukin 6), IL6R (interleukin 6 receptor), FOXP3 (forkhead box P3), TLR2 (toll-like receptor 2), TLR4 (toll-like receptor 4), IFNL3 (interferon lambda 3, formerly known as IL28B), and IFNL4 (interferon lambda 4, formerly known as IL29). Polymorphisms were genotyped using matrix-assisted laser desorption/ionization-time of flight mass spectrometry. RESULTS There were significant differences in the allelic and genotype frequencies of IFNL3 (IL28B) rs12971396, rs8099917, rs11882871, rs12979860, rs4803217 between non-dyslipidemia and dyslipidemia groups in people with obesity. These single nucleotide polymorphisms (SNPs) of IFNL3 were highly linked (D' and r > 0.90), so the result of one SNP could represent the result of other SNPs. For IFNL3 rs12971396, people with the homozygous genotype (the major group) carried a higher risk of dyslipidemia than people with the heterozygous genotype (P < 0.001, OR = 4.46, 95%CI, 1.95-10.22). CONCLUSIONS The favorable genotypes of type III interferon, which have a beneficial role in anti-virus function, were associated with dyslipidemia in a Chinese population with obesity. Type III interferon could have a pathologic role and confer risk of dyslipidemia in people with obesity and chronic inflammation.
Collapse
Affiliation(s)
- Tiantian Xu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Bo Peng
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Mengmeng Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qingjing Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Junya Yang
- School of Health and Related Research, University of Sheffield, Sheffield, United Kingdom
| | - Minli Qu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lizhen Lin
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jing Wu,
| |
Collapse
|
145
|
Guo Q, Li L, Hou S, Yuan Z, Li C, Zhang W, Zheng L, Li X. The Role of Iron in Cancer Progression. Front Oncol 2021; 11:778492. [PMID: 34858857 PMCID: PMC8631356 DOI: 10.3389/fonc.2021.778492] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/15/2021] [Indexed: 01/19/2023] Open
Abstract
Iron is an essential trace element for the human body, and its deficiency or excess can induce a variety of biological processes. Plenty of evidences have shown that iron metabolism is closely related to the occurrence and development of tumors. In addition, iron plays an important role in cell death, which is very important for the development of potential strategies for tumor treatment. Here, we reviewed the latest research about iron metabolism disorders in various types of tumors, the functions and properties of iron in ferroptosis and ferritinophagy, and new opportunities for iron-based on treatment methods for tumors, providing more information regarding the prevention and treatment of tumors.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Liwen Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shanshan Hou
- Department of Pharmacy, Zhejiang Pharmaceutical College, Ningbo, China
| | - Ziqiao Yuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chenhui Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenzhou Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|